WO2020117701A1 - Methods of determining ph and calcium or chloride concentration in samples - Google Patents

Methods of determining ph and calcium or chloride concentration in samples Download PDF

Info

Publication number
WO2020117701A1
WO2020117701A1 PCT/US2019/064053 US2019064053W WO2020117701A1 WO 2020117701 A1 WO2020117701 A1 WO 2020117701A1 US 2019064053 W US2019064053 W US 2019064053W WO 2020117701 A1 WO2020117701 A1 WO 2020117701A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
fluorophore
label
stranded nucleic
cells
Prior art date
Application number
PCT/US2019/064053
Other languages
French (fr)
Inventor
Yamuna Krishnan
Kasturi CHAKRABORTY
Ka Ho LEUNG
Anand SAMINATHAN
Original Assignee
The University Of Chicago
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The University Of Chicago filed Critical The University Of Chicago
Priority to JP2021531481A priority Critical patent/JP7307797B2/en
Priority to US17/295,704 priority patent/US20220011325A1/en
Priority to CN201980090973.XA priority patent/CN113383075A/en
Priority to EP19893431.7A priority patent/EP3887519A4/en
Publication of WO2020117701A1 publication Critical patent/WO2020117701A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/84Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving inorganic compounds or pH
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • C12Q1/6825Nucleic acid detection involving sensors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/582Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with fluorescent label
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y306/00Hydrolases acting on acid anhydrides (3.6)
    • C12Y306/03Hydrolases acting on acid anhydrides (3.6) acting on acid anhydrides; catalysing transmembrane movement of substances (3.6.3)
    • C12Y306/03014H+-transporting two-sector ATPase (3.6.3.14), i.e. F1 ATPase

Definitions

  • This disclosure relates to methods for determining pH and also calcium (Ca 2+ ) concentration or chloride (Cl-) concentration in biological samples. More particularly, this disclosure relates to methods capable of simultaneously determining pH and Ca 2+ concentration, or pH and Cl- concentration using nucleic acid complexes.
  • Lysosomes are, by and large, regarded as a single population while assaying for a specific lysosomal function.
  • recent promising studies have considered that sub populations of lysosomes might perform sub-sets of tasks.
  • many cell types have evolved specialized lysosomes that perform distinct functions. For instance in addition to lysosomes, skin cells have melanosomes, neutrophils have azurophil granules, cytotoxic T- cells have secretory lysosomes while every cell has autolysosomes.
  • Functional imaging based on physical parameters such as lysosome movement, morphology or spatial position within cells have revealed sub-populations that exhibit different behaviors and functions.
  • autolysosomes and lysosomes adopt tubulovesicular and vesicular morphologies respectively.
  • Lysosomes have also been sorted into two populations based on how actively they move within the cell. Spatial positioning of lysosomes is emerging as a correlate of lysosome function. Nevertheless, the capacity to chemically discriminate between lysosome populations in live cells would significantly aid the understanding of lysosome biology by providing the ability to quantitatively correlate chemotypes with function. For example, in 1960s electron microscopy and bright field imaging could only distinguish up to three stages in melanosome maturation based on morphology and melanin content respectively.
  • H + and Cl- are two highly abundant ions in the lysosome that are critical to its function. In fact, no other organelle has a greater concentration of either ion. Lysosomal pH is critical to lysosome maturation, cargo degradation and recycling of degraded material.
  • High lumenal CF in the lysosome is required for the activity of certain lysosome-resident hydrolases. Unlike other organelles, however, lumenal Cl- level in the lysosome is independent of lumenal pH.
  • Ca 2+ regulates diverse cellular functions upon its controlled release from different intracellular stores that initiates signaling cascades. Lysosomes have recently been recognized as“acidic Ca 2+ stores”, and lumenal Ca 2+ is central to its diverse functions. For example, risk genes for Parkinson's disease such as LRRK2, ATP6AP2, ATP13A2, and genetic risk associated GBA1 gene, are predicted to act in lysosomal pathways.
  • Lysosomal Ca 2+ release channels are amenable to investigation because Ca 2+ release can be tracked using cytosolic Ca 2+ dyes or genetically encoded Ca 2+ indicators anchored to the cytoplasmic face of the lysosome. Upon Ca 2+ release, these probes indicate cytosolic Ca 2+ in the area surrounding lysosomes. In contrast, lumenal Ca 2+ cannot be quantitated, impeding the study of lysosomal Ca 2+ importers. Consequently, lysosomal Ca 2+ importers have not yet been identified in animals, with the closest evidence being that the Xenopus CAX gene localizes in lysosomes upon overexpression.
  • novel nucleic acid complexes of the disclosure can efficiently and accurately determine pH in addition to Ca 2+ concentration or CF concentration in samples.
  • novel nucleic acid complexes of the disclosure simultaneously determine pH and Ca 2+ concentration, or pH and CF concentration in samples.
  • nucleic acid complexes including:
  • nucleic acid complex further comprises a first label conjugated to the first single-stranded nucleic acid molecule or the second single-stranded nucleic acid molecule and the first label is capable of producing a signal.
  • nucleic acid complex comprising
  • a first single-stranded nucleic acid molecule comprising a Ca 2+ fluorophore or a Cl- fluorophore crosslinked to the first strand;
  • Another aspect of the disclosure relates to a methods for screening a candidate drug in a model cell or organism, the method including delivering the nucleic acid complex of the disclosure to the cell or organism; contacting the cell or organism with the candidate drug, measuring the intensity of the signal; and determining the pH, and Ca 2+ or Cl- from the measured signal.
  • the model cell or organism is a model for a lysosomal storage disease.
  • the disease is a lysosomal storage disease.
  • Another aspect of the disclosure relates to a method for detecting the severity of a disease, the progression of the disease, or the presence of a disease, the method including delivering the nucleic acid complex of the disclosure to a sample; measuring the intensity of the signal; and determining the pH, and Ca 2+ or Cl- from the measured signal.
  • Figures 1A-1D illustrate design and characterization of a nucleic acid complex according to one embodiment of the disclosure, CalipHluor Ly .
  • Figure 1 A provides a working principle of CalipHluor Ly . pH-induced FRET between Alexa488 (donor, D, sphere) and Alexa647 (acceptor, A, star) reports on pH ratiometrically.
  • Alexa488 donor, D, sphere
  • Alexa647 acceptor, A, star
  • Figures 2A-2J illustrate in vivo sensing characteristics of a nucleic acid complex according to one embodiment of the disclosure, CalipHluor Ly .
  • Figure 2A illustrates representative CalipHluor Ly labelled coelomocytes imaged in the donor (D,i), acceptor (A,ii), Rhod-5F (O, iii) and Alexa647 (R, iv) channels.
  • D/A (v) and O/R (vi) are the corresponding pixel-wise pseudocolor images.
  • Figures 3A-3M illustrate the pH and [Ca 2+ ] maps accompanying endosomal maturation of a nucleic acid complex according to one embodiment of the disclosure, CalipHluor.
  • Figure 3A illustrates CalipHluor marks the indicated organelles in coelomocytes time dependently, by scavenger receptor mediated endocytosis.
  • Figure 3B illustrates colocalization of CalipHluor and GFP-tagged markers of endocytic organelles at indicated time points post-injection in nematodes.
  • Figure 3L illustrates mean endosomal pH of EE, LE and Ly.
  • Figure 3M illustrates mean endosomal [Ca 2+ ] in EE, LE and Ly. Data represented as the mean ⁇ S.E.M. Experiments were repeated thrice independently with similar results.
  • Figure 5C illustrates pseudocolor pH and Ca 2+ maps of lysosomes in normal and L6025 fibroblasts.
  • Figure 5D illustrates mean lysosomal pH
  • Data represent mean ⁇ S.E.M. Experiments were repeated thrice independently with similar results.
  • Figure 6A illustrates fluorescence emission spectra of Rhod-5F (left) and Alexa 647 (right) with increasing [Ca2 + ] upon exciting Rhod-5F and Alexa647 at 560 nm and 650 nm respectively.
  • Figure 6B illustrates normalized O/R ratio of Rhod-5F/Alexa 647 with increasing [Ca2 + ] at pH 7.2 and 5.5. Error bar represents ⁇ S.E.M of three independent experiments.
  • Figures 7A-7G illustrate characterization of nucleic acid complexes according to certain embodiments of the disclosure, CalipHluorCy and CalipHluor.
  • Figure 7A illustrates gel showing the conjugation of Rhod-5F to D2-DBCO strand. Gels were visualized in EtBr and TMR channels.
  • Figure 7B illustrates native PAGE showing formation of CalipHluor Ly . Gels were visualized in Alexa 488, TMR and Alexa 647 channels.
  • Figure 7C is a schematic of working principle of CalipHluor. A pH-induced FRET changes between Alexa488 (donor, sphere) and Alexa647 (acceptor, star) is used to report pH ratiometrically.
  • FIG. 7D illustrates gel showing the conjugation of Rhod-5F to 03-DBCO strand. Gels were visualized in EtBr and TMR channels.
  • Figure 7E illustrates native PAGE showing formation of CalipHluor. Gels were visualized in Alexa 488, TMR and Alexa 647 channels.
  • Figure 7F shows emission spectra of CalipHluor at pH values ranging from 7.5 to 5.0 upon excitation at 488 nm.
  • Figure 7G shows normalized ratio of fluorescence intensity of donor to that of acceptor (D/A) of CalipHluor as a function of pH.
  • Error bar represents ⁇ S.E.M. of three independent experiments.
  • Figures 8A-8B illustrate in vivo performance of a nucleic acid complex according to one embodiments of the disclosure, CalipHluor Ly .
  • Figure 8A provides representative pseudo color images of coelomocytes labeled with CalipHluor Ly and clamped at the indicated pH.
  • Figure 8B provides representative pseudo color images of coelomocytes labeled with CalipHluor Ly and clamped at different free [Ca 2+ ] at pH 5.5. Scale bar 5 pm. Experiments were repeated thrice independently with similar results.
  • Figures 9A-9H illustrate comparison of in vitro and in vivo pH and Ca 2+ calibration profile of a nucleic acid complex according to one embodiments of the disclosure
  • Figures 10A-10C illustrate endocytic trafficking of a nucleic acid complex according to one embodiments of the disclosure, CaiipHiuor ⁇ 7 in coelomocytes. Representative confocal images taken 5 min, 17 min, and 60 min following injection of CaiipHiuor ⁇ 7 in worms expressing GFP::RAB-5, GFP::RAB-7 and LMP-1 ::GFP. Scale bar 5 pm.
  • FIG 11C shows representative images of worms expressing LMP-1 ::GFP (the left panels) in the background of various indicated RNAi, which were injected with CalipHluor LyA647 (the middle panels) and imaged 60 mins post-injection. Scale bar: 5 pm.
  • Figures 12A-12H illustrate design and characterization of a nucleic acid complex according to one embodiment of the disclosure, CalipHluor mLy .
  • Figure 12A is a schematic of working principle of CalipHluor wLy .
  • Figures 12B and 12C provide calibration curves comparing in vitro (solid gray) and on beads (light gray) calibration at pH 4.6 and pH 5.1 , respectively.
  • Figures 12D and 12E provide comparison of K d and fold change (FC), respectively, in O/R of CalipHluor Ly (solid) and CalipHluor wLy (hatched).
  • Figure 12F provides representative images.
  • Figures 14A-14E illustrate design and characterization of ChloropHore.
  • Figure 14A is a schematic of the working principle: A pH-induced change in FRET between Alexa546 (donor, sphere) and Alexa647 (acceptor, star) reports pH ratiometrically. A Cl- sensitive fluorophore (BAC, triangle) and Alexa647 report Cl- ratiometrically.
  • Figure 14B shows a pH and Cl- response profiles of ChloropHore : Normalized fluorescence intensity ratio (D/A) of donor (D) and acceptor (A) upon donor excitation in vitro as a function of pH and 50 mM Cl- (bottom, light gray).
  • Figures 15D and 15G show ChloropHore labels lysosomes in HDF cells.
  • Figure 15D are representative images of co localization between lysosomes of HDF cells labeled with FITC Dextran (middle panel) and LAMP-1 RFP (left panel) with the corresponding Pearson’s correlation coefficient (Figure 15E).
  • Figure 15F are representative images of lysosomes of HDF cells labelled with TMR dextran (TMR; middle panel) and ChloropHore (Alexa647, left panel).
  • Figures 16A-160 illustrate intracellular calibration of ChloropHore and
  • FIGS. 16A are fluorescence images of primary HDF cells labeled with ChloropHore , clamped at the indicated pH and [Cl-], imaged in the donor (D), acceptor (A), reference (R), BAC (G) channel and the corresponding pseudocolour D/A (pH) and R/G (Cl-) maps.
  • Figures 16B and 16C show in cell calibration surface corresponding to the pH and Cl- response profiles of the sensing modules in ChloropHore at various [Cl-] and pH values respectively.
  • Figure 16D is a representative scatter plot of D/A versus the R/G values of endosomes in primary HDF cells from a normal individual clamped at the indicated pH and [Cl-].
  • Figure 16E is the scatter plot in Figure 16D, represented as a density plot in pseudocolour where red and blue correspond to populations with higher and lower frequencies of occurrence, i.e., a 2-IM profile.
  • Figures 16F and 16H show 2-IM profiles of HDF cells clamped in varying pH and fixed [Cl-].
  • Figures 161 and 16K show 2-IM profiles of HDF cells at fixed pH and increasing [Cl-].
  • Figure 16L is a single endosome clamping in HDF cells. ChloropHore labeled cells clamped at indicated pH (i) and Cl- (ii) were clamped to a different indicated pH and same Cl- (iv)
  • Figure 16M shows 2D-scatter plots and their projected histograms on a single axis of each clamping step of the same endosomes are shown. Gray arrow represents the direction of change in D/A (pH) values for each endosome.
  • Figure 16N is Chlorophore LY labeled HDF clamped at indicated pH (i) and Cl- (ii) were clamped to the same pH (iii) but different [Cl-] (iv)
  • Figures 17A-17G illustrate that 2-IM chemically resolves lysosome populations.
  • Figure 17A is a respective pseudocolour D/A and R/G map of HDF cells derived from normal individuals 1 , NP-A patient 1 and NP-C patient 1 labeled with Chlorophore LY .
  • 2-IM profiles and the histograms of D/A, R/G ratios of ( Figures 17B, 171) normal individual (N.l.) and in presence of bafilomycin A1 ( Figure 17C) or NPPB ( Figure 17D).
  • FIG 19 illustrates characterization of ChloropHore by CD spectroscopy
  • Thermal denaturation was carried out on ChloropHore at (b) pH 4.0 as well as at (c) pH 7.5 to demonstrate i-motif formation. Experiments were performed in triplicate.
  • Figure 20 illustrates response of ChloropHore (200 nM) given by the fold change in R/G from ⁇ 0 mM to 100 mM of all indicated ions and 30% glycerol. *2 mM. Error bars indicate the mean ⁇ s.e.m. of three independent measurements.
  • Figures 21A-21E illustrates design and characterization of ChloropHore Ly .
  • Figure 21 A is a schematic of the working principle of ChloropHore Ly in the“open state” (low FRET) at high pH and in the“closed” state (high FRET) at low pH.
  • Cl sensitive fluorophore BAC displays high fluorescence in the absence of Cl ion whereas the fluorescence has been quenched by increasing concentration of Cl ions. Both pH and Cl sensing modules are independent of each other.
  • Performance of the pH and Cl sensing modules are in light gray and dark gray, respectively. Plot of fold change of pH sensing modules in indicated Cl concentration (light gray hatched bars, on the right). Fold change (dark gray hatched bars, in the middle) and K sv (solid gray) for Cl sensing module versus pH. Fluorescence intensity ratio ( Figure 21 D) D/A and ( Figure 21 E) R/G of ChloropHore Ly as a function of Cl and pH. Error bars indicate the mean ⁇ s.e.m. of three independent measurements.
  • Figure 22 illustrates stability of ChloropHore in HDF cells
  • Figure 24 illustrates intracellular calibration profile of ChloropHore.
  • (a) D/A and (b) R/G ratio of ChloropHore as a function of pH and Cl- in endosomes of human dermal fibroblasts each showing the intracellular calibration profile in the specified pH/CI- clamped condition (color dots). Error bars indicate the mean of three independent experiments ⁇ s.e.m. (n 15 cells, 150 endosomes).
  • Figure 25 illustrates performance of / mLY in fibroblast
  • a Schematic of the working principle of l mLY at low and high pH. pH indicator - Oregon green (OG) was conjugated to DNA and hybridize with its Alexa 647 labeled complementary strand
  • b In vitro (dark gray) and in cellulo (light gray) calibration curve of l mLY . Histograms showing spread of G/R ratios of lysosomes of cells from a normal individual (c) without or (d) 500 nM bafilomycin A1.
  • Figure 26 illustrates quantitative performance of l mLY within single lysosome.
  • (b) G/R change of lysosomes of cells that clamped with pH 7.5 and pH 4.5. Experiments were performed in triplicate (n 15 lysosomes).
  • Figure 27 illustrates labeling lysosomes of BHK-21 cells
  • a-b ChloropHore internalization by BHK-21 cells is competed out by excess maleylated BSA (mBSA, 10 pM), revealing uptake is by scavenger receptors. Cells are imaged in Alexa647 channel (c)
  • Figure 28 illustrates labeling lysosome of T47D cells (a-b) ChloropHore
  • T47D cells internalization by T47D cells is competed out by excess maleylated BSA (mBSA, 10 mM), revealing uptake is by scavenger receptors.
  • mBSA maleylated BSA
  • Cells are imaged in Alexa647 channel (c)
  • FIG 29 illustrates 2-IM profiles of lysosomes in various cell types.
  • a combination reporter for pH and Ca 2+ can map both ions in parallel in the same endosome with single endosome addressability, achieving highly accurate measures of lumenal Ca 2+ .
  • the pH reporter module of the combination reporter the pH is determined in individual endosomes.
  • the affinity of the Ca 2+ sensitive module for example, dissociation constant (K d ) and changes with pH, a K d correction factor suited to the lumenal pH of each endosome may be applied to compute the true value of lumenal Ca 2+ with single-endosome resolution.
  • K d was computed at every pixel in the pH map to generate a K d map. From the K d map and the O/R map, the true Ca 2+ map of the acidic organelle can be constructed.
  • DNA nanodevices are versatile chemical reporters that can quantitatively map second messengers in real time, in living systems.
  • the modularity of DNA allows integration of distinct functions in precise stoichiometries into a single assembly. These include, for example, (i) a module to fluorescently sense a given ion (ii) a normalizing module for ratiometric quantitation, and/or (iii) a targeting module to localize the reporter in a specific organelle.
  • a nucleic acid complex according to one embodiment of the disclosure, CalipHluor is used in the methods of the disclosure to map organellar pH and Ca 2+ simultaneously and with single organelle addressability.
  • ATP13A2 (a risk gene for Parkinson’s disease) transports divalent ions such as Mg 2+ , Mn 2+ , Cd 2+ , Zn 2+ yet, has not been tested for its ability to transport Ca 2+ .
  • the human homolog, ATP13A2 was also shown herein to facilitate lysosomal Ca 2+ entry by measuring lysosomal Ca 2+ in fibroblasts derived from patients with Kufor Rakeb Syndrome. This constitutes the first example of a lysosomal Ca 2+ importer in the animal kingdom.
  • the ability to map pH and Ca 2+ or with single organelle addressability is important to discriminate between lysosomal hypo-acidification and Ca 2+ dysregulation.
  • the nucleic acid complexes of the disclosure in certain embodiments, can be used to map lumenal Ca 2+ changes in diverse organelles. As a result, the nucleic acid complexes of the disclosure, for example, can provide new insights into organellar Ca 2+ regulation.
  • the inventors have also shown that by measuring the pH and Cl- - simultaneously in the same lysosome (referred herein as“2-IM”) and retaining this information with single lysosome addressability, one can resolve lysosomal sub-populations quantitatively in live cells.
  • Methods such as 2-IM have proved elusive to realize thus far for several reasons.
  • CP- sensitive small molecule probes offer the necessary chemical selectivity, molar brightness and long wavelength excitation, but not the required spatial addressability or organelle targetability.
  • Genetically encoded CP sensors offer stable spatial localization, but the response of these reporters to CP is pH sensitive. This complicates analysis of most organelles as lumenal CP entry is coupled to their acidification.
  • DNA nanodevices comprise a range of biologically interfacable, quantitative imaging probes that unite the photophysical advantages of small molecules, the stable localization provided by proteins along with the precision of organelle targeting that is accessible to endocytic tracers.
  • nucleic acid complexes of the disclosure that can ratiometrically image pH and (CP) simultaneously with single lysosome addressability, lysosome chemotypes on a two-dimensional map that correlates lumenal pH with lumenal (CP) can be discriminated.
  • nucleic acid complexes of the disclosure as describe herein comprises the 1 :1 stoichiometry of DNA hybridization to integrate four functions with stoichiometric precision onto a single probe: (i) a pH sensing function (ii) a CP sensing function (iii) an internal standard for simultaneous ratiometric quantitation of both CP & pH and (iv) a lysosome targeting function for addressability.
  • 2-IM is a highly sensitive method that chemically resolved a high-chloride, high acidity lysosome population in human fibroblasts isolated from skin biopsies of normal, healthy humans.
  • nucleic acid complexes include a Ca 2+ fluorophore or a Cl- fluorophore crosslinked to the first single-stranded nucleic acid molecule.
  • Ca 2+ fluorophore known in the art may be used in the complexes and methods of the disclosure. Some examples include, but are not limited to, Rhod-5F, XRhod-5F, Rhod- FF, XRhod-FF, Oregon Green 488 BAPTA-6F, Fluo 5F, Fluo 4FF, Oregon Green BAPTA- 5N, Fluo-5 N, and Mag-Fluo-4 indicator. Additional Ca 2+ fluorophores may be selected from the labels disclosed herein.
  • the Ca 2+ fluorophore includes Rhod-5F, XRhod-5F, or Rhod-FF indicator.
  • the Ca 2+ fluorophore Rhod-5F indicator has the following formula:
  • any available position may be functionalized as to allow for crosslinking to the first strand.
  • a hydrogen atom on the Rhod-5F molecule provided above may be replaced by a functional group that is configured to crosslink to the first strand.
  • the Ca 2+ fluorophore as described herein can be crosslinked to the first single- stranded nucleic acid molecule using linkers and methods known in the art.
  • the Ca 2+ fluorophore can be crosslinked using peptide chemistry, click chemistry, by forming ester, ether, thioether, disulfide, amine reactive N-Hydroxysuccinimidyl (NHS) esters, isocyanates, and isothiocyanates bonds, etc.
  • the Ca 2+ fluorophore is crosslinked to the first strand through a linker moiety stable under physiological conditions.
  • the Ca 2+ fluorophore of the disclosure as described herein includes the following formula:
  • the nucleic acid molecules may be labeled with a“fluorescent dye” or a“fluorophore.”
  • Dyes that may be used in the disclosed methods include, but are not limited to, the following dyes sold under the following trade names: 1 ,5 IAEDANS; 1 ,8-ANS; 4-methylumbelliferone; 5-carboxy-2,7-dichlorofluorescein; 5-carboxyfluorescein (5-FAM); 5- carboxytetramethylrhodamine (5-TAMRA); 5-hydroxy tryptamine (HAT); 5-ROX (carboxy-X- rhodamine); 6-carboxyrhodamine 6G; 6-JOE; 7-amino-4-methylcoumarin; 7- aminoactinomycin D (7-AAD); 7-hydroxy-4-methylcoumarin; 9-amino-6-chloro-2- methoxyacridine; ABQ; Acid Fuchsin; ACMA (9-amino-6-chloro-2- meth
  • LysoSensor Blue LysoSensor Green; LysoSensor Yellow/Blue; Mag Green; Magdala Red (Phloxin B); Mag-Fura Red; Mag-Fura-2; Mag-Fura-5; Mag-lndo-1 ; Magnesium Green;
  • PYMPO Propidium lodid
  • Pyrene Pyronine
  • Pyronine B Pyrozal Brilliant Flavin 7GF
  • QSY 7 Quinacrine Mustard; Red 613 [PE-TexasRed]; Resorufin; RH 414; Rhod-2;
  • Rhodamine Rhodamine
  • Rhodamine 110 Rhodamine 123; Rhodamine 5 GLD; Rhodamine 6G;
  • Rhodamine B Rhodamine B 200; Rhodamine B extra; Rhodamine BB; Rhodamine BG; Rhodamine Green; Rhodamine Phallicidine; Rhodamine Phalloidine; Rhodamine Red;
  • Rhodamine WT Rose Bengal; R-phycocyanine; R-phycoerythrin (PE); RsGFP; S65A; S65C; S65L; S65T; Sapphire GFP; SBFI; Serotonin; Sevron Brilliant Red 2B; Sevron Brilliant Red 4G; Sevron Brilliant Red B; Sevron Orange; Sevron Yellow L; sgBFPTM; sgBFPTM (super glow BFP); sgGFPTM; sgGFPTM (super glow GFP); SITS; SITS (Primuline); SITS (Stilbene Isothiosulphonic Acid); SNAFL calcein; SNAFL-1 ; SNAFL-2; SNARF calcein; SNARF1 ; Sodium Green; Spectru Aqua; SpectrumGreen; Spectru Orange; Spectrum Red; SPQ (6- methoxy-N-(3-sulfopropyl)quinolinium); Stilbene; Sulphorhodamine B can C;
  • Sulphorhodamine G Extra SYTO 11 ; SYTO 12; SYTO 13; SYTO 14; SYTO 15; SYTO 16; SYTO 17; SYTO 18; SYTO 20; SYTO 21 ; SYTO 22; SYTO 23; SYTO 24; SYTO 25; SYTO 40; SYTO 41 ; SYTO 42; SYTO 43; SYTO 44; SYTO 45; SYTO 59; SYTO 60; SYTO 61 ; SYTO 62; SYTO 63; SYTO 64; SYTO 80; SYTO 81 ; SYTO 82; SYTO 83; SYTO 84; SYTO 85; SYTOX Blue; SYTOX Green; SYTOX Orange; TETTM; Tetracycline;
  • Thiadicarbocyanine (DiSC3); Thiazine Red R; Thiazole Orange; Thioflavin 5; Thioflavin S; Thioflavin TON; Thiolyte; Thiozole Orange; Tinopol CBS (Calcofluor White); TMR; TO-PRO- 1 ; TO-PRO-3; TO-PRO-5; TOTO-1 ; TOTO-3; Tricolor (PE-Cy5); TRITC
  • TetramethyIRodaminelsoThioCyanate True Blue; TruRed; Ultralite; Uranine B; Uvitex SFC; VIC®; wt GFP; WW 781 ; X-Rhodamine; XRITC; Xylene Orange; Y66F; Y66H; Y66W; Yellow GFP; YFP; YO-PRO-1 ; YO-PRO-3; YOYO-1 ; YOYO-3; pHrodoTM (available from Thermo Fischer Scientific, Inc. Waltham, MA), and salts thereof.
  • Fluorescent dyes or fluorophores may include derivatives that have been modified to facilitate conjugation to another reactive molecule.
  • fluorescent dyes or fluorophores may include amine-reactive derivatives such as isothiocyanate derivatives and/or succinimidyl ester derivatives of the fluorophore.
  • the nucleic acid molecules of the disclosed compositions and methods may be labeled with a quencher.
  • Quenching may include dynamic quenching (e.g., by FRET), static quenching, or both.
  • Illustrative quenchers may include Dabcyl.
  • Illustrative quenchers may also include dark quenchers, which may include black hole quenchers sold under the tradename“BHQ” (e.g., BHQ-0, BHQ-1 , BHQ-2, and BHQ-3, Biosearch Technologies, Novato, Calif.). Dark quenchers also may include quenchers sold under the tradename “QXLTM” (Anaspec, San Jose, Calif.). Dark quenchers also may include DNP-type non- fluorophores that include a 2,4-dinitrophenyl group.
  • the proximity of the two labels can be detected using fluorescence resonance energy transfer (FRET) or fluorescence polarization.
  • FRET is a distance-dependent interaction between the electronic excited states of two dye molecules in which excitation is transferred from a donor molecule to an acceptor molecule without emission of a photon.
  • donor/acceptor dye pairs for FRET are known in the art and may include fluorophores and quenchers described herein such as
  • the labels can be conjugated to the nucleic acid molecules directly or indirectly by a variety of techniques. Depending upon the precise type of label used, the label can be located at the 5' or 3' end of the oligonucleotide, located internally in the oligonucleotide's nucleotide sequence, or attached to spacer arms extending from the oligonucleotide and having various sizes and compositions to facilitate signal interactions.
  • nucleic acid molecules containing functional groups e.g., thiols or primary amines
  • a phosphoramidite dye to the 5' hydroxyl of the 5' base by the formation of a phosphate bond, or internally, via an appropriately protected phosphoramidite.
  • Nucleic acid molecules may also incorporate functionalizing reagents having one or more sulfhydryl, amino or hydroxyl moieties into the nucleic acid sequence.
  • a 5' phosphate group can be incorporated as a radioisotope by using polynucleotide kinase and [y32P] ATP to provide a reporter group.
  • Biotin can be added to the 5' end by reacting an aminothymidine residue, introduced during synthesis, with an N-hydroxysuccinimide ester of biotin.
  • Labels at the 3' terminus for example, can employ polynucleotide terminal transferase to add the desired moiety, such as for example, cordycepin, 35S-dATP, and biotinylated dUTP.
  • Oligonucleotide derivatives are also available as labels.
  • etheno-dA and etheno-A are known fluorescent adenine nucleotides which can be incorporated into a reporter.
  • etheno-dC is another analog that can be used in reporter synthesis.
  • the reporters containing such nucleotide derivatives can be hydrolyzed to release much more strongly fluorescent mononucleotides by the polymerase's 5' to 3' nuclease activity as nucleic acid polymerase extends a primer during PCR.
  • a first label is conjugated to the second single-stranded nucleic acid molecule.
  • a second label is conjugated to the first single-stranded nucleic acid molecule or the second single-stranded nucleic acid molecule.
  • a first label is conjugated to the second single-stranded nucleic acid molecule and the second label is conjugated to the first single-stranded nucleic acid molecule, wherein the first label is capable of producing a signal, and wherein the intensity of the signal varies as a function of the conformation of the nucleic acid complex.
  • a second label is conjugated to the third single-stranded nucleic acid.
  • the first label is conjugated to the second single-stranded nucleic acid molecule and the second label is conjugated to the third single-stranded nucleic acid, wherein the first label is capable of producing a signal, and wherein the intensity of the signal varies as a function of the conformation of the nucleic acid complex.
  • the intensity of the signal is irrelevant of the distance between the first and second labels and/or the relative orientation of the first and second labels.
  • the intensity of the signal varies as a function of at least one of the distance between the first and second labels and the relative orientation of the first and second labels.
  • the first and second labels comprise a donor and acceptor pair.
  • the signal is measured using a FRET technique.
  • the signal can be measured at 2 different wavelengths.
  • the signal can be measured at 4 different wavelengths.
  • at least one label is selected from the group consisting of an Atto dye, an Alexa Flour® dye, a Cy® dye, and a BODIPY dye.
  • the donor and acceptor pair are FITC and TRITC, Cy3 and Cy5, or Alexa-488 and Alexa-647.
  • the donor and acceptor pair are labels described herein.
  • the first and second label comprise a donor fluorophore and an acceptor quencher.
  • the signal and label is directionally dependent (anisotropy).
  • Non-limiting examples of such labels include Atto dyes, BODipy dyes, Alexa dyes,
  • the nucleic acid complexes of the disclosure include a first single- stranded molecule and a second single-stranded nucleic acid molecule that is partially or fully complementary to the first single-stranded molecule.
  • the nucleic acid complexes of the disclosure include a first single-stranded molecule, a second single-stranded nucleic acid molecule that is partially complementary to the first single- stranded molecule, and a third single-stranded nucleic acid molecule that is partially complementary to the first single-stranded molecule.
  • the second nucleic acid strand and/or the third nucleic acid strand is one that is only partially complementary to the first nucleic acid.
  • a nucleic acid strand is fully complementary when all bases are capable of forming conventional Watson-Crick base pairing (e.g. G-C and A-T base pairing).
  • a nucleic acid strand is partially complementary when at least one of the base pairs is not complementary to the opposing strand.
  • the second single nucleic acid strand comprises at least 4 non
  • the second single nucleic acid strand comprises at least, at most, or exactly 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 (or any derivable range therein) non-complementary nucleic acid bases.
  • each of the first single-stranded nucleic acid molecule the second single-stranded nucleic acid molecule, and/or the third single-stranded nucleic acid molecule is independently less than 200 nucleotides.
  • the nucleic acid complexes described herein are capable of determining the Ca 2+ concentration in a range of 10 nM to 1 mM, or 10 nM to 100 nM, or 10 nM to 500 nM, or 100 nM to 500 nM, or 100 nM to 1 pM, or 500 nM to 1 pM, or 500 nM to 50 pM, or 1 pM to 1 mM, or 1 pM to 10 mM, or 10 pM to 10 mM, or 100 pM to 1 mM, or 100 pM to 10 mM.
  • the recited Ca 2+ concentration is excluded.
  • nucleic acid complexes described herein are useful as CF concentration sensors, and have high sensitivity without a substantial change in cooperativity.
  • the nucleic acid complexes described herein are capable of determining the Cl- concentration in a range of 1 mM to 100 mM, the range is inclusive of the recited CF concentration.
  • the nucleic acid complexes described herein are capable of determining the CF concentration in a range of 1 mM to 50 mM, 1 mM to 25 mM, or 1 mM to 10 mM, or 5 mM to 100 mM, or 5 mM to 50 mM, 5 mM to 25 mM, or 5 mM to 10 mM, or 10 mM to 25 mM, or 10 mM to 50 mM, or 10 mM to 75 mM, or 20 mM to 80 mM, or 30 mM to 70 mM.
  • the recited CFconcentration is excluded.
  • nucleic acid complexes described herein are also useful as pH sensors, and have high sensitivity without a substantial change in cooperativity.
  • the nucleic acid complexes described herein are capable of determining the pH of is less than 5.5 or more than 7.0.
  • the nucleic acid complexes described herein are capable of determining the pH of less than or exactly pH 5.8, 5.7, 5.6, 5.5, 5.4,
  • the nucleic acid complexes described herein are capable of determining the pH of more than or exactly pH 6.8, 6.9, 7.0, 7.1 , 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 8.0, 8.2, 8.4, 8.6, 8.8 or 9.0 (or any derivable range therein).
  • the nucleic acid complexes described herein are capable of determining the pH in the range of 5.5 to 7.
  • the nucleic acid complexes described herein are capable of determining the pH in the range of 5.8 and 6.8, or 5.8 to 7, or 5.6 to 6.8, or 5.4 to 6.8.
  • Cytosine rich DNA sequences are found in human genomes such as in telomeres and in promoters of several oncogenes, e.g., c-myc.
  • the nucleic acid complexes of the disclosure can form a special tetraplex structure under slightly acidic condition where two parallel duplexes paired through OCH+ pairs intercalated with each other in head to tail orientation called the i-motif.
  • The“i-motif” is a nucleic acid (DNA and/or RNA) containing complex characterized by the presence of cytosine-rich stretches or stretches rich in cytosine derivatives, including two parallel-stranded duplexes in which the cytosines or derivatives thereof form base pairs, and the two duplexes are associated anti parallel to one another.
  • the pairs of cytosine or derivatives thereof of one duplex are intercalated with those of the other duplex.
  • the structure of an i-motif differs from that of the usual DNA duplex because the base pairing scheme involves hemiprotonated cytosines which result in the formation of OC+ base pairs. Specifically, one of the cytosines contained in each pair is protonated.
  • the i-motif may also exist as a tetramer formed by the association of two duplexes as described above.
  • the nucleic acid complexes of the disclosure may be synthesized from oligonucleotide sequences including a stretch of at least two, at least three, or at least four consecutive cytosines.
  • oligonucleotide sequences including a stretch of at least two, at least three, or at least four consecutive cytosines.
  • a cytosine-rich stretch may contain one or two non-cytosine base(s) in between the cytosines. However, this may reduce the stability of the i-motif.
  • the cytosine stretches which comprise the i-motif may belong to different strands of nucleic acids;
  • any two of them may also be linked together covalently or non-covalently. Also, any two of them may be part of a single nucleic acid strand wherein they are separated by a stretch of specified bases.
  • the second single-stranded nucleic acid molecule of the nucleic acid complexes of the disclosure comprises the sequence C n XC n YC n ZC n (SEQ ID NO. 13), wherein C is cytosine; X, Y and Z are each one or more adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues of the nucleic acid molecule are modified. In some embodiments, at least or exactly 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, or 18 (or any derivable range therein) cytosine residues of the second single-stranded nucleic acid molecule are modified.
  • the number of cytosine residues of the second single-stranded nucleic acid molecule that are modified is in the range of 2 to 18, or 4 to 18, or 10 to 18, or 14 to 18, or 2 to 14, or 4 to 14, or 10 to 14, or 2 to 10, or 4 to 10, or 8 to 16.
  • each of X, Y, and Z is independently AA or TAA. In certain embodiments, each of X, Y, and Z is independently TAA. In some embodiments, n is 3, 4, or 7. In further embodiments, n is at least, at most, or exactly 3, 4, 5, 6, 7, 8, or 9 (or any derivable range therein). In some embodiments, n is 4. In some embodiments, the modification is a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group. In some embodiments, the cytosine is modified with a methyl or bromo group. In some
  • the modification is at the 5’ position of the cytosine. In some embodiments, all the cytosines in the second nucleic acid molecule are modified with the same
  • all the cytosines in the second nucleic acid molecule are modified with a negatively charged modification. In some embodiments, all the cytosines in the second nucleic acid molecule are modified with a positively charged modification.
  • the second single-stranded molecule comprises the sequence ((C a ) n X(C ) n Y(C c ) n Z(C d ) n (SEQ ID NO. 14) wherein C a , C b , C c , and C d are equal to n number of consecutive cytosine residues; X, Y, and Z are one or more adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 3.
  • each of C a , C , C c , and C d comprise at least one modified cytosine.
  • each of C a , C b , C c , and C d comprise at least, at most, or exactly 1 , 2, 3, 4, or 5 modified cytosines (or any derivable range therein).
  • the modified cytosine is the first or last consecutive cytosine in each of C a , C , C c , and C d .
  • n 3 and the modified cytosine is the second consecutive cytosine in each of C a , C , C c , and C d .
  • n 4 and the modified cytosine is the second or third consecutive cytosine in each of C a , C , C c , and C d .
  • each of C and C c comprise at least two modified cytosines and each of C a and C d consist of unmodified cytosine.
  • C a or C d consists of modified cytosine residues.
  • C a , C , C c , and/or C d consist of or comprise of at least, at most, or exactly 1 ,
  • the second single-stranded nucleic acid molecule and/or the third single-stranded nucleic acid molecule of the nucleic acid complexes of the disclosure comprises the sequence C n XC n , wherein C is cytosine; X and Y are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified. In some embodiments, at least or exactly 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, or 18 (or any derivable range therein) cytosine residues of the second single-stranded nucleic acid molecule are modified.
  • the number of cytosine residues of the second single-stranded nucleic acid molecule that are modified is in the range of 2 to 18, or 4 to 18, or 10 to 18, or 14 to 18, or 2 to 14, or 4 to 14, or 10 to 14, or 2 to 10, or 4 to 10, or 8 to 16.
  • X and Y are independently AA or TAA. In certain embodiments, X and Y are independently AA or TAA. In certain
  • X and Y are independently AA.
  • n is 3, 4, or 7.
  • n is at least, at most, or exactly 3, 4, 5, 6, 7, 8, or 9 (or any derivable range therein).
  • n is 4.
  • the modification is a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group.
  • the cytosine is modified with a methyl or bromo group.
  • the modification is at the 5’ position of the cytosine.
  • all the cytosines in the second nucleic acid molecule are modified with the same modification.
  • all the cytosines in the second nucleic acid molecule are modified with a negatively charged modification. In some embodiments, all the cytosines in the second nucleic acid molecule are modified with a positively charged modification.
  • the nucleic acid complexes described herein are useful as pH sensors, and have high sensitivity (as evidenced by fold change of D/A ratio) without a substantial change in cooperativity.
  • the method further comprises calculating a D/A ratio from the signal intensity values.
  • the D/A ratio is a normalized value.
  • the fold change of the D/A ratio is at least 4.1 , 5, 6, 7, or 7.5.
  • the fold change of the D/A ratio is at least or exactly 4.5, 5, 6, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11 , 11.5, 12, 12.5, 13, or 14 (or any derivable range therein) and the cooperativity, compared to the unmodified nucleic acid complexes, is changed by less than 1.75, 1.5, 1.25. 1 , 0.75. 0.5, 0.25, 0.2, or 0.1 fold or any derivable range therein.
  • f is altered without substantially increasing the cooperativity.
  • f is at least, at most, or exactly 5.0, 5.2, 5.4, 5.6, 5.8, 6.0, 6.2, 6.4, 6.6, 6.8, 7.0, 7.2, 7.4, 7.6,
  • the pH haif compared to the un-modified nucleic acid complexes, is at least, at most, or exactly 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1 , 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 2.9, or 3.0 pH units different (or any range derivable therein).
  • the pH haif compared to the un-modified nucleic acid complexes, is at least, at most, or exactly 3, 5, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 24,
  • the measured value described herein i.e. , signal intensity, pH ha
  • the first and second single-stranded nucleic acid molecules are capable of forming an i-motif under acidic conditions.
  • the first nucleic acid strand is capable of forming an intramolecular complex comprising two parallel- stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
  • the nucleic acid molecules and complexes of the disclosure in some embodiments, comprise a targeting moiety, such as a nucleic acid, small molecule, or polypeptide that has an affinity for a certain target or, by virtue of its chemical makeup, is targeted to a particular location in the cell.
  • the targeting moiety can act as a handle to target the nucleic acid complexes of the disclosure to different subcellular locations.
  • the targeting moiety may be a nucleic acid that binds to a receptor protein, and the receptor protein may be one that is intracellularly targeted or conjugated to a protein that is intracellularly targeted.
  • the targeting moiety is a nucleic acid sequence.
  • the targeting moiety has a cognate artificial protein receptor.
  • the artificial receptor may be, for example, a single chain variable fragment (scFv), transcription factor, Zn-fingered protein, leucine zipper, or DNA binding immunoglobulin,
  • the targeting moiety is encoded on the same nucleic acid strand as the first and/or second single-stranded nucleic acid molecule.
  • the targeting moiety is selected from an aptamer, a duplex domain targeted to an artificial protein receptor, a nucleic acid sequence that binds an anionic-ligand binding receptor, and an endocytic ligand.
  • the sub-cellular localization sequence targets the nucleic acid complex to a region of the cell selected from the group consisting of: the cytosol, the endoplasmic reticulum, the mitochondrial matrix, the chloroplast lumen, the medial trans-Golgi cistemae, the lumen of lysosome, the lumen of an endosome, the peroxisome, the nucleus, and a specific spatial location on the plasma membrane.
  • the sub-cellular organelle is one that exchanges membrane directly or indirectly with the plasma membrane.
  • nucleic acids may be used in combination with additional nucleic acid based sensors, such as those described in
  • PNA or PNA strand or PNA sequence is used
  • DNA or DNA strand or DNA sequence is used interchangeably and has the same scope or meaning.
  • RNA or RNA strand or RNA sequence is used interchangeably and has the same scope or meaning.
  • the nucleic acid complex of the present disclosure self assembles two or all three strands through Watson-Crick base pairing, which is stable under physiological conditions.
  • A) DNA only and B) a combination of DNA and RNA are used: A) DNA only and B) a combination of DNA and RNA.
  • the targeting moiety comprising only DNA hybridizes to normalizing module to form the dsDNA domain required for intracellular targeting via an anionic ligand binding receptor (ALBR).
  • ABR anionic ligand binding receptor
  • the RNA sequence used in combination with DNA in the targeting moiety is used to achieve targeting to Transferrin pathway.
  • a DNA strand is used as first strand and/or the second strand.
  • the nucleic acid complex has a dsDNA part (minimum 15 bp sequence) resulting from the hybridization of the first strand and the second strand, or the first strand and the third strand, or the first strand with the second strand and the third strand.
  • the nucleic acid complex comprises d(AT)4 sequence and hence is targeted to any given compartment in any cell that expresses scFv tagged protein of choice.
  • the nucleic acid complex as described herein comprises the first strain has the sequence 5’-the Ca 2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and the second strain has the sequence 5’-the first label-CCC CTA ACC CCT AAC CCC TAA CCC CAT ATA TAT CCT AGA ACG ACA GAC AAA CAG TGA GTC-3’(SEQ ID NO:08).
  • the nucleic acid complex as described herein comprises the first strain has the sequence 5’-TTA TAG GAT CCT GCG GTC GG/the Ca 2+ fluorophore/ GGC ACC AGG CGT AAA ATG TA-3’(SEQ ID NO:09); the second strain has the sequence: 5’-the first label-CCC CAA CCC CAA TAC ATT TTA CGC CTG GTG CC- 3’ (SEQ ID NO:10); and the third strain has the sequence: 5’-CCG ACC GCA GGA TCC TAT AAA ACC CCA ACC CC-the second label-3 (SEQ ID NO: 11).
  • the nucleic acid complex as described herein comprises the first strain has the sequence 5’-the Ca 2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and the second strain has the sequence 5'- the first label - AT AAC ACA TAA CAC ATAACAAAA TAT ATA TCC TAG AAC GAC AGA CAA ACA GTG AGT
  • the nucleic acid complex as described herein comprises the first strain has the sequence 5’-the Cl- fluorophore-ATC AAC ACT GCA-Lys-COOH (SEQ ID NO:22); the second strain has the sequence 5'-TAT TGT GTA TTG TGT ATT GTT TTA TAT AT /the first label/ A TAG GAT CTT GCT GTC TGG TGT GCA GTG TTG AT-3'(SEQ ID NO:23); and the third strain has the sequence: 5’-CAC CAG ACA GCA AGA TCC TAT ATA TAT ACC CCA ATC CCC AAT CCC CAA TCC CC-the second label-3' (SEQ ID NO:24).
  • the methods described herein may be used to monitor the pH changes and/or Ca 2+ or Cl- concentration in real-time during cellular processes.
  • the methods are for monitoring endocytosis. While not wishing to be limited by theory, acidification plays a major role in facilitating cargo dissociation from receptors or in mediating cellular entry of toxins and viruses during endocytosis.
  • the nucleic acid complex exhibits a pH response inside cells illustrated by the capture of spatiotemporal pH changes associated with endocytosis in living cells.
  • Fluorescence in the sample can be measured in a variety of ways, such as using a fluorometer or fluorescence microscopy.
  • excitation radiation from an excitation source having a first wavelength, passes through excitation optics.
  • the excitation optics cause the excitation radiation to excite the sample.
  • labels in the sample emit radiation which has a wavelength that is different from the excitation wavelength.
  • the device can include a temperature controller to maintain the sample at a specific temperature while it is being scanned.
  • a multi-axis translation stage can be used to move a microtiter plate holding a plurality of samples in order to position different wells to be exposed.
  • the multi-axis translation stage, temperature controller, auto-focusing feature, and electronics associated with imaging and data collection can be managed by an appropriately programmed digital computer.
  • the computer also can transform the data collected during the assay into another format for presentation.
  • an“increase” (or“decrease”) in a signal from the nucleic acid complex refers to the change in a signal in the sample compared to a reference sample.
  • the reference sample may be a control sample (e.g., an untreated population of cells where the effects of a drug or agent are being examined), or it may be the same sample at a different period of time, for instance, where the intracellular pH and/or Ca 2+ or CF concentration is being monitored to follow one or more cellular processes.
  • the term“detectable” refers to a property of the nucleic acid complex that allows one to determine the pH and/or Ca 2+ or CF concentration of a biological sample by detecting activity, e.g., fluorescence activity, possessed by the nucleic acid complex under certain conditions.
  • the signal from the nucleic acid complex is normalized by plotting the donor/acceptor (D/A) signal ratio as a function of pH in a standard reference sample. pH variation on a doubly-labeled nucleic acid complex changes the ratio between its closed and open states thereby resulting in different ratios of the donor and acceptor intensities (D/A) because of FRET in the closed state due to i-motif formation.
  • a pH calibration curve and/or Ca 2+ or CF concentration calibration curve may be generated to which test samples may be compared and normalized.
  • An intracellular calibration curve may be generated according to methods described in U.S. Patent Application Publication No.: 2010/0304370, which is herein incorporated by reference. Briefly, for the pH calibration curve, cells are pulsed, washed, incubated with an ionophore in buffers at a given pH and then mildly fixed. Donor and acceptor FRET images are acquired from which D/A ratios are obtained. The mean D/A of individual cells or regions thereof at each pH are plotted as a function of pH for the intracellular pH calibration curve. The D/A ratio of the test sample can be compared to the calibration curve. Similar approach may be used to generate Ca 2+ concentration calibration curve and CF concentration calibration curve. Related methods are also described in the Examples of the application.
  • intracellular pH and/or Ca 2+ or CF concentration may be monitored for the purposes of examining cellular phenomena and/or screening the effects of various compounds, wherein the level of the signal from a nucleic acid complex (e.g., increased or decreased signal) in a test sample at a first time point is determined and compared with the level found in a test sample obtained at a later time point.
  • the change in signal may reflect a relative change in pH and/or Ca 2+ or CF concentration between the two samples. For example, where a FRET pair is used as a label, an increase in signal from one time point to another may indicate an increase in pH between the two time points.
  • a decrease in signal from one point to another may indicate a decrease in pH or decrease in Ca 2+ or CF concentration.
  • the absolute level of signal may be compared to a reference sample of known standards or reference samples in order to determine the precise pH and/or Ca 2+ or CF concentration of the sample.
  • the sample can be classified or assigned to a particular pH value based on how similar the measured levels were compared to the control levels for a given group.
  • the standard deviations of the control group levels can be used to make a probabilistic determination and the method of this disclosure are applicable over a wide range of probability-based determinations.
  • the measured level of signal falls within 2.5 standard deviations of the mean of any of the control groups, then that sample may be assigned to that group.
  • the measured level of signal falls within 2.0 standard deviations of the mean of any of the control groups then that sample may be assigned to that group.
  • the measured level of signal falls within 1.5 standard deviations of the mean of any of the control groups then that sample may be assigned to that group.
  • the measured level of signal is 1.0 or less standard deviations of the mean of any of the control groups levels then that sample may be assigned to that group.
  • this process allows determination, with various degrees of probability, in which group a specific sample should be placed.
  • Statistical methods can also be used to set thresholds for determining when the signal intensity in a test sample can be considered to be different than or similar to the reference level.
  • statistics can be used to determine the validity of the difference or similarity observed between a test sample's signal intensity and the reference level.
  • concentration sensors may vary in their respective pKa, and the differences in pKa can be used to select the most suitable nucleic acid complex for a particular application.
  • a sensor should be used whose pKa is close to the pH of the sample to be measured.
  • the pKa may be within 1.5 pH unit, within 1.0 pH unit, or within 0.5 pH units of the sample.
  • the nucleic acid complexes described herein are useful as pH and Cl- concentration sensors.
  • the signal of the Cl- fluorophore is linearly dependent on CF concentration with constant Stern-Volmer quenching constant (K sv ).
  • the signal of the Cl- fluorophore is insensitive to physiological change in pH and to cations, non-halide anions (nitrate, phosphate, bicarbonate, sulfate), and albumin.
  • the fluorescence of the nucleic acid complex can be compared to the fluorescence of a second sensor, e.g., a second nucleic acid complex that is also present in the measured sample.
  • the second nucleic acid complex should have an emission spectra distinct from the first nucleic acid complex so that the emission spectra of the two sensors can be distinguished. Because experimental conditions such as focusing and cell movement will affect fluorescence of the second sensor as well as the first sensor, comparing the relative fluorescence of the two sensors may allow for the normalization of fluorescence.
  • FLIM is used to measure the conformational change upon i- motif formation.
  • anisotropy imaging is used to measure the conformational change.
  • Fluorescence-lifetime imaging microscopy FLIM is an imaging technique for producing an image based on the differences in the exponential decay rate of the fluorescence from a fluorescent sample. It can be used as an imaging technique in confocal microscopy, two-photon excitation microscopy, and multiphoton tomography. The lifetime of the fluorophore signal, rather than its intensity, is used to create the image in FLIM.
  • FLIM is used to gain pH information, as one of the photo physical properties of the dyes that would change when the nucleic acid complex changes conformation due to a change in the pH.
  • the dye is an Atto dye, BODIPY dye, Alexa dye, TMR/TAMRA dye, or Cy dye.
  • the signal intensity changes by at least twenty percent as the Ca 2+ concentration is raised. In some embodiments, the signal intensity changes by at least 10, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, or 90% or any derivable range therein when the Ca 2+ concentration is raised.
  • the methods of the disclosure determine the Ca 2+ concentration in a range of 10 nM to 10 mM, the range is inclusive of the recited Ca 2+ concentration.
  • a Ca 2+ concentration that is determined by the methods of the disclosure concentration is in a range of 10 nM to 1 mM, or 10 nM to 100 nM, or 10 nM to 500 nM, or 100 nM to 500 nM, or 100 nM to 1 pM, or 500 nM to 1 pM, or 500 nM to 50 pM, or 1 pM to 1 mM, or 1 pM to 10 mM, or 10 pM to 10 mM, or 100 pM to 1 mM, or 100 pM to 10 mM.
  • the recited Ca 2+ concentration is excluded.
  • the Ca 2+ concentration is determined by comparing the measured signal to a reference value.
  • the Ca 2+ concentration is determined by comparing the measured signal to a reference value.
  • the signal value and/or reference value is normalized.
  • the method further comprises creating a standard curve.
  • a standard curve can be created by measuring the signal intensity at different known Ca 2+ concentration values.
  • a curve can be plotted as signal intensity vs. Ca 2+ concentration. The signal intensity of an unknown Ca 2+
  • the signal intensity changes by at least twenty percent as the Cl- concentration is raised. In some embodiments, the signal intensity changes by at least 10, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, or 90% or any derivable range therein when the Cl- concentration is raised.
  • the methods of the disclosure determine the Cl- concentration in a range of 1 mM to 100 mM, the range is inclusive of the recited Cl- concentration.
  • a Cl- concentration that is determined by the methods of the disclosure concentration is in a range of 1 mM to 50 mM, 1 mM to 25 mM, or 1 mM to 10 mM, or 5 mM to 100 mM, or 5 mM to 50 mM, 5 mM to 25 mM, or 5 mM to 10 mM, or 10 mM to 25 mM, or 10 mM to 50 mM, or 10 mM to 75 mM, or 20 mM to 80 mM, or 30 mM to 70 mM.
  • the recited Cl- concentration is excluded.
  • the Cl- concentration is determined by comparing the measured signal to a reference value. In some embodiments, the Cl- concentration is determined by comparing the measured signal to a reference value. In some embodiments, the signal value and/or reference value is normalized. In some embodiments, the method further comprises creating a standard curve. A standard curve can be created by measuring the signal intensity at different known Cl- concentration values. A curve can be plotted as signal intensity vs. Cl- concentration. The signal intensity of an unknown Cl- concentration can then be determined by finding the corresponding reference value on the plot.
  • the signal intensity changes by at least twenty percent as the pH is raised from at least one of pH 4 to pH 5, pH 5 to pH 6, pH 6 to pH 7, and pH 7 to pH 8. In some embodiments, the signal intensity changes by at least 10, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, or 90% or any derivable range therein when the pH is raised from at least pH 4, 5, 6, or 7 to pH 5, 6, 7, or 8 (or any range derivable therein). [0130] In certain embodiments, when pH ranges or values are discussed herein, the range is inclusive of the recited pH. For example, a pH range from 4.0 to 5.0 or between 4.0 and 5.0 includes the pH of 4.0 and 5.0. In other embodiments, the recited pH is excluded.
  • the pH is determined by comparing the measured signal to a reference value. In some embodiments, the pH is determined by comparing the measured signal to a reference value. In some embodiments, the signal value and/or reference value is normalized. In some embodiments, the method further comprises creating a standard curve. A standard curve can be created by measuring the signal intensity at different known pH values. A curve can be plotted as signal intensity vs. pH. The signal intensity of an unknown pH can then be determined by finding the corresponding reference value on the plot.
  • the methods of the disclosure are suitable for measuring pH and concentration of Ca 2+ or Cl- in early endosome, late endosome, plasma membrane, lysosome, autophagolysosome, recycling endosome, cis Golgi network (CGN), trans Golgi network (TGN), endoplasmic reticulum (ER), peroxisomes, or secretory vesicles.
  • the methods of the disclosure are suitable for measuring pH and
  • any sample containing Ca 2+ or Cl- can be used in the methods of the disclosure.
  • the sample is a biological sample selected from a cell, cell extract, cell lysate, tissue, tissue extract, bodily fluid, serum, blood and blood product.
  • the sample is a live cell.
  • the sample is a biological sample from a patient.
  • nucleic acid complexes as described herein can be readily introduced into a host cell, e.g., a mammalian (optionally human), bacterial, parasite, yeast or insect cell by any method in the art.
  • a host cell e.g., a mammalian (optionally human), bacterial, parasite, yeast or insect cell
  • nucleic acids can be transferred into a host cell by physical, chemical or biological means. It is readily understood that the introduction of the nucleic acid molecules yields a cell in which the intracellular pH may be monitored. Thus, the method can be used to measure intracellular pH in cells cultured in vitro.
  • the nucleic acid complex of the disclosure can also be readily introduced into a whole organism to measure the pH in a cell or tissue in vivo.
  • nucleic acid complex of the disclosure can be transferred into an organism by physical, chemical or biological means, e.g., direct injection.
  • the methods for introducing nucleic acid complexes of the disclosure may be those disclosed in Chakraborty et ai,“Nucleic Acid-Based Nanodevices in Biological Imaging,” Annu. Rev. Biochem. 85:349-73 (2016), incorporated in its entirety by reference herein.
  • the use of lipid formulations is contemplated for the introduction of the nucleic acid complex of the disclosure into host cells (in vitro, ex vivo or in vivo).
  • the nucleic acid complex of the disclosure may be associated with a lipid.
  • the nucleic acid complex of the disclosure associated with a lipid may be encapsulated in the aqueous interior of a liposome, interspersed within the lipid bilayer of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the oligonucleotide(s), entrapped in a liposome, complexed with a liposome, dispersed in a solution containing a lipid, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with a micelle, or otherwise associated with a lipid.
  • the lipid, lipid/nucleic acid complex compositions are not limited to any particular structure in solution. For example, they may be present in a bilayer structure, as micelles, or with a“collapsed” structure. They may also simply be interspersed in a solution, possibly forming aggregates which are not uniform in either size or shape.
  • the one or more nucleic acid complexes of the disclosure are linked to a targeting sequence that directs the nucleic acid complex to a desired cellular compartment.
  • the methods, compositions, nucleic acid complexes, and kits of the disclosure can be used for the detection of diseases, the monitoring of diseases, and as a drug screening platform.
  • the disease is characterized as a lysosomal dysfunction disease.
  • the pathology of the disease includes lysosomal dysfunction.
  • Lysosomal dysfunction diseases include, for example, autosomal recessive osteopetrosis, Farber disease, Krabbe disease (infantile onset and late onset), Fabry disease (Alpha-galactosidase A), Schindler disease (Alpha-galactosidase B), Sandhoff disease (infantile, juvenile, or adult onset), Tay-Sachs, juvenile hexosaminidase A
  • Type C and D Neuronal Ceroid Lipofuscinoses: Type 1 Santavuori- Haltia disease / Infantile NCL (CLN1 PPT1), Type 2 Jansky-Bielschowsky disease / Late infantile NCL (CLN2/LINCL TPP1), Type 3 Batten-Shis meyer- Vogt disease / Juvenile NCL (CLN3), Type 4 Kufs disease / Adult NCL (CLN4), Type 5 Finnish Variant / Late Infantile (CLN5), Type 6 Late Infantile Variant (CLN6), Type 7 CLN7, Type 8 Northern Epilepsy (CLN8), Type 8 Turkish Late Infantile (CLN8), Type 9 German/Serbian Late Infantile
  • Type 10 Congenital Cathepsin D Deficiency (Unknown), Type 10 Congenital Cathepsin D Deficiency (CTSD); Wolman disease, alpha- mannosidosis, beta-mannosidosis, aspartylglucosaminuria, fucosidosis, lysosomal transport diseases, cystinosis, pycnodysostosis, salla disease / sialic acid storage disease, infantile free sialic acid storage disease (ISSD), glycogen storage diseases, Type II Pompe Disease, Type lllb Danon disease, and cholesteryl ester storage disease.
  • the disease is autosomal recessive osteopetrosis.
  • the disease is Niemann-Pick C disease.
  • the disclosure provides a detection kit useful for determining the pH and the presence, absence, or concentration of Ca 2+ in a sample, cell or region thereof.
  • the technology encompasses kits for measuring the pH and Ca 2+ of one or more cells in a sample.
  • the disclosure also provides a detection kit useful for determining the pH and the presence, absence, or concentration of Cl- in a sample, cell or region thereof.
  • the technology encompasses kits for measuring the pH and Cl- of one or more cells in a sample.
  • the kit can comprise a nucleic acid complex as described herein.
  • the methods described herein may be performed by utilizing pre-packaged diagnostic kits comprising the necessary reagents to perform any of the methods of the technology.
  • a kit would include a detection reagent for measuring the pH and Ca 2+ of a cell or region thereof, or a detection reagent for measuring the pH and CF of a cell or region thereof.
  • the detection reagents are the nucleic acid complexes of the disclosure. Oligonucleotides are easily synthesized and are stable in various formulations for long periods of time, particularly when lyophilized or otherwise dried to a powder form. In this form, they are easily reconstituted for use by those of skill in the art.
  • kits can also include buffers useful in the methods of the technology.
  • the kits may contain instructions for the use of the reagents and interpreting the results.
  • the technology provides a kit comprising at least one sample (e.g., a pH standard and/or a Ca 2+ concentration standard and/or a CF concentration standard) packaged in one or more vials for use as a control.
  • a sample e.g., a pH standard and/or a Ca 2+ concentration standard and/or a CF concentration standard
  • Each component of the kit can be enclosed within an individual container and all of the various containers can be within a single package, along with instructions for performing the assay and for interpreting the results of the assays performed using the kit.
  • the kit comprises a device for the measurement of pH and Ca 2+ in a sample. In some embodiments, the kit comprises a device for the measurement of pH and CF in a sample. In some embodiments, the device is for measuring pH and/or analyte in cell culture or in whole, transparent organisms (e.g., C. elegans).
  • a“fluorescent dye” or a“fluorophore” is a chemical group that can be excited by light to emit fluorescence. Some fluorophores may be excited by light to emit phosphorescence. Dyes may include acceptor dyes that are capable of quenching a fluorescent signal from a fluorescent donor dye.
  • crosslinked refers to a covalent connection between the nucleic acid molecule and another moiety of interest, such as the Ca 2+ fluorophore or the OG fluorophore.
  • the crosslink between the nucleic acid molecule and this moiety is water compatible.
  • the crosslink between the nucleic acid molecule and this moiety is stable under physiological conditions.
  • nucleic acid As used herein,“nucleic acid,”“nucleotide sequence,” or“nucleic acid sequence” refer to a nucleotide, oligonucleotide, polynucleotide, or any fragment thereof and to naturally occurring or synthetic molecules.
  • peptide nucleic acid or "PNA” as used herein generally refers to nucleic acid analogue in which the sugar phosphate backbone of natural nucleic acid has been replaced by a synthetic peptide backbone.
  • RNA equivalent in reference to a DNA sequence, is composed of the same linear sequence of nucleotides as the reference DNA sequence with the exception that all occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose. It is understood to be a molecule that has a sequence of bases on a backbone comprised mainly of identical monomer units at defined intervals. The bases are arranged on the backbone in such a way that they can enter into a bond with a nucleic acid having a sequence of bases that are complementary to the bases of the oligonucleotide. The most common oligonucleotides have a backbone of sugar phosphate units.
  • Oligonucleotides which do not have a hydroxyl group at the 2' position
  • Oligonucleotides also may include derivatives, in which the hydrogen of the hydroxyl group is replaced with organic groups, e.g., an allyl group.
  • An oligonucleotide is a nucleic acid that includes at least two nucleotides.
  • nucleic acid sequence may be“complementary” to a second nucleic acid sequence.
  • the terms“complementary” or“complementarity,” when used in reference to nucleic acids refer to sequences that are related by base-pairing rules. For natural bases, the base pairing rules are those developed by Watson and Crick. As an example, for the sequence“T-G-A”, the complementary sequence is“A-C-T.”
  • Complementarity can be “partial,” in which only some of the bases of the nucleic acids are matched according to the base pairing rules. Alternatively, there can be“complete” or“total” complementarity between the nucleic acids. The degree of complementarity between the nucleic acid strands has effects on the efficiency and strength of hybridization between the nucleic acid strands.
  • Oligonucleotides as described herein may be capable of forming hydrogen bonds with oligonucleotides having a complementary base sequence. These bases may include the natural bases such as A, G, C, T and U, as well as artificial bases. An oligonucleotide may include nucleotide substitutions. For example, an artificial or modified base may be used in place of a natural base such that the artificial base exhibits a specific interaction that is similar to the natural base.
  • oligonucleotide that is complementary to another nucleic acid will“hybridize” to the nucleic acid under suitable conditions (described below).
  • “hybridization” or“hybridizing” refers to the process by which an oligonucleotide single strand anneals with a complementary strand through base pairing under defined hybridization conditions.
  • “Specific hybridization” is an indication that two nucleic acid sequences share a high degree of complementarity. Specific hybridization complexes form under permissive annealing conditions and remain hybridized after any subsequent washing steps.“Hybridizing” sequences which bind under conditions of low stringency are those which bind under non- stringent conditions (6*SSC/50% formamide at room temperature) and remain bound when washed under conditions of low stringency (2*SSC, 42° C). Hybridizing under high stringency refers to the above conditions in which washing is performed at 2*SSC, 65° C (where SSC is 0.15M NaCI, 0.015M sodium citrate, pH 7.2).
  • HPLC purified oligonucleotides were dissolved in Milli-Q water to make 100 mM stock solutions and quantified using UV-spectrophotometer and stored at -20 °C.
  • Ethylene glycol-bis ⁇ -aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA) ampicillin, carbencillin, isopropyl b-D-l-thiogalactopyranoside (IPTG), nigericin and monensin were purchased from Sigma and ionomycin was obtained from Cayman Chemical (USA).
  • Calcium nitrate tetrahydrate, sodium nitrate, sodium phosphate dibasic, sodium phosphate monobasic, magnesium sulfate anhydrous, glycerol, bovine serum albumin (66 kDa), nigericin, monensin, tributyltin chloride (TBT-CI) and amitriptyline hydrochloride were obtained from Sigma, and sodium chloride was purchased from Alfa Aesar.
  • Magnesium nitrate tetrahydrate, sodium nitrate, sodium phosphate dibasic, sodium phosphate monobasic, magnesium sulfate anhydrous, glycerol, bovine serum albumin (66 kDa), nigericin, monensin, tributyltin chloride (TBT-CI) and amitriptyline hydrochloride were obtained from Sigma, and sodium chloride was purchased from Alfa Aesar.
  • CalipHluor Ly sample was diluted to 30 nM in pH clamping buffer [CaCI 2 (50 mM to 10 mM), HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI 2 (1 mM)] of desired pH and equilibrated for 30 minutes at room temperature. All the samples were excited at 495 nm and emission spectra was collected from 505 nm to 750 nm.
  • pH clamping buffer [CaCI 2 (50 mM to 10 mM), HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI 2 (1 mM)] of desired pH and equilibrated for 30 minutes at room temperature. All the samples were excited at 495 nm and emission spectra
  • CalipHluor Ly sample was diluted to 30 nM in Ca 2+ clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM),
  • Rhod-5F and Alexa 647 were excited at 545 nm and 630 nm respectively. Emission spectra for Rhod-5F (O) and Alexa 647 (R) were collected from 570-620 nm and 660-750 nm, respectively. Mean of O/R from two independent experiments and their S.E.M.
  • Rhod-5F Rhod-5F
  • X is free [Ca 2+ ]
  • Y is O/R ratio at given free [Ca 2+ ]
  • S is O/R ratio at low [Ca 2+ ]
  • E is O/R ratio at high [Ca 2+ ]
  • K d is dissociation constant and n is Hill coefficient.
  • Fold change response in O/R of CalipHluor Ly was calculated from ratio of O/R at high [Ca 2+ ] and O/R at low [Ca 2+ ]
  • CalipHluor trafficking in coelomocytes was done in transgenic strains expressing endosomal markers such as GFP::RAB-5 (EE), GFP::RAB-7 (LE) and LMP-1 ::GFP (Ly) as described previously (Surana et al. (2011) Nat. Commun. 2:340). Briefly, worms were injected with CalipHluor A6 47 (500 nM) and incubated for specific time points and transferred on to ice. Worms were anaesthetized using 40 mM of sodium azide in M9 solution.
  • RNAi experiments in C.elegans Bacteria from the Ahringer RNAi library expressing dsRNA against the relevant gene was fed to worms, and measurements were carried out in one-day old adults of the F1 progeny (Kamath and Ahringer, 2003). RNA knockdown was confirmed by probing mRNA levels of the candidate gene, assayed by RT-PCR. Briefly, total RNA was isolated using the Trizol-chloroform method; 2.5 pg of total RNA was converted to cDNA using oligo-dT primers. 5 pL of the RT reaction was used to set up a PCR using gene- specific primers. Actin mRNA was used as a control. PCR products were separated on a 1.5% agarose-TAE gel.
  • BHK-21 cells BHK-21 cells, Human dermal fibroblasts (HDF), human fibroblast cells harboring mutations in ATP13A2 and homozygous for
  • Fibroblasts AG01518, GM08429, GM00112, GM16195, GM13205, GM03252, GM03393, GM11097, GM18414, GM23162 and GM17910 were purchased from Coriell Institute (Camden, NJ) and cultured with the suggested protocols from Coriell Institute.
  • Lysosomes pre-labeled with TMR-Dex w utilized to examine the trafficking time scales for ChloropHore.
  • Pre-labeled cells were pulsed with 1 mM of ChloropHore and chased for indicated time and imaged.
  • Cross talk and bleed-through were recorded and disciver to be negligible between TMR channel and A647 channel.
  • Pearson's correlation coefficient measures the pixel-by-pixel covariance of two images while it range from 0-1 and 1 indicates complete colocalization .
  • PCC are examined by the tool in ImageJ/Fiji 2.0.0-rc-54/1.51h. Upon pixel shift, PCC values decrease significantly suggesting non-random colocalization.
  • Clamping buffers with various concentration of chloride ions were prepared by adding chloride positive buffer (150 m M KCI, 5 mM NaCI, 1 mM CaCI 2 , 1 mM MgCI 2 , 10 mM HEPES, MES, sodium acetate) to a chloride negative buffer (150 mM KN0 3 , 20 mM NaN0 3 , 1 mM Ca(N0 3 ) 2 , 1 mM Mg(N0 3 ) 2 , 10 mM HEPES, MES, sodium acetate) at the same indicated pH in different ratios.
  • the cells were then imaged in clamping buffer. pH and calcium clamping were carried out using CalipHluor" 1 ⁇ .
  • Fibroblast cells were pulsed for 1 hour and chased for 2 hours with 500nM CalipHluor mLy . Cells are then fixed with 200 ml_ 4% paraformaldehyde (PFA) for 15 minutes at room temperature, washed three times and retained in 1 * PBS.
  • PFA paraformaldehyde
  • endosomal calcium concentrations were equalized by incubating the previously fixed cells in the appropriate calcium clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI 2 (1 mM)] by varying amount of free [Ca 2+ ] from 1 mM to 10 mM and adjusted to different pH values.
  • the buffer also contained nigericin (50 pM), monensin (50 pM) and ionomycin (20 pM) and the cells were incubated for 2 hours at room temperature.
  • fibroblast cells are pulsed with 500nM of CalipHluor mLy for 1 hour, chased for 9 hours (8 hours for L0625 cells) and then washed with 1 * PBS and imaged in Hank’s Balanced Salt Solution (HBSS). Imaging was carried out on IX83 research inverted microscope (Olympus Corporation of the Americas, Center Valley, PA) using a 100*, 1.42 NA, DIC oil immersion objective (PLAPON, Olympus Corporation of the Americas, Center Valley, PA) and Evolve Delta 512 EMCCD camera (Photometries, USA).
  • DNA stability assay DNA stability assay was performed as described previously. Cells are pulsed with 1 mg/ml_ TMR-Dex for 1 h and chased for 16 h. The TMR-Dex labeled cells are pulsed with 2 pM of ChloropHore for 1 h, chased for indicated time points for imaging.
  • Fibroblast cells are pulsed with 2 pM of l mLY , ChloropHore or ChloropHore LY for 1 h, chased for 9 h (30 min pulse and 1 h chase for J774.A1, BHK-21 and T47D cells) and then washed with 1 * PBS and subjected for imaging.
  • NP-A/B Niemann Pick type A/B
  • NP-C type C
  • Niemann Pick type A/B and C models were prepared according to the previous reported method. Cells were incubated with 65 mM of the acid sphingomyelinase (ASM) inhibitor amitriptyline (AH) and 20 mM of the NPC1 inhibitor U18666A for 24 h to create a NP-A/B and NP-C models respectively.
  • ASM acid sphingomyelinase
  • AH amitriptyline
  • the cells are pulsed with 2 pM of ChloropHoreLY and either 65 pM AH or 20 pM U18666A for 1 h, chased with either 65 pM AH or 20 pM U18666A for 9 h and then washed with 1 * PBS and imaged.
  • fibroblasts from NP-C patient were incubated with 50 pM of o-Hydroxypropyl-b- cyclodextrin (b-CD) for 24 h.
  • the cells are pulsed with 2 pM of ChloropHore LY and 50 pM of b-CD for 1 h, chased with 50 pM of b-CD for 9 h and then washed with 1 * PBS and imaged.
  • Image acquisition was carried out on wide field IX83 inverted microscope (Olympus Corporation of the Americas, Center Valley, PA) using a 60*, 1.42 NA, phase contrast oil immersion objective (PLAPON, Olympus Corporation of the
  • FRET channel images were obtained using the 480/20 band pass excitation filter, 705/72 band pass emission filter and 89016-ET-FITC/Cy3/Cy5 dichroic filter.
  • Rhod-5F channel images (O) were obtained using 545/25 band pass excitation filter, 595/50 band pass emission filter and a 89016-ET-FITC/Cy3/Cy5 dichroic filter.
  • Confocal images were acquired on a Leica TCS SP5 II STED laser scanning confocal microscope (Leica
  • Alexa 488 was excited using an Argon ion laser for 488 nm excitation, Alexa 647 using He- Ne laser for 633 excitation and Rhod-5F using DPSS laser for 561 nm excitation with a set of dichroics, excitation, and emission filters suitable for each fluorophore.
  • Mean O/R of each distribution was plotted as a function of free [Ca 2+ ] to generate the in vivo Ca 2+ calibration curve.
  • Pseudo color pH and Ca 2+ images were obtained by measuring the D/A and O/R ratio per pixel, respectively.
  • worms were immersed in clamping buffer [CaCI 2 (50 mM to 10 mM), HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI 2 (1 mM)] of desired pH solutions containing the ionophores nigericin (50 pM), monensin (50 pM) and ionomycin (20 pM). Worm cuticle was perforated to facilitate the entry of buffer in to the body. After 75 min of incubations in clamping buffer, coelomocytes were imaged using wide-field microscopy. Three independent measurements, each with 10 worms, were made for each pH value.
  • Ca 2+ clamping measurements were carried out using CalipHluor Ly . Worms were injected with CalipHluor Ly (500 nM) and incubated at 22 °C for 2 hours. After 2 hours, worms were immersed in Ca 2+ clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI 2 (1 mM)] by varying amount of free [Ca 2+ ] from 1 pM to 10 mM and adjusted to different pH values (5.3-6.5). Three independent measurements, each with 10 worms, were made for Ca 2+ value.
  • Ca 2+ clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI 2 (1 mM)
  • K K 2 and pH 1 2 represent parameters derived from a Boltzmann fit of the in vivo pH calibration curve, and Y represents the D/A ratio in a given endosome.
  • Donor (D) and acceptor (A) images were background subtracted by drawing an ROI outside the worms.
  • Donor (D) image was duplicated and a threshold was set to create a binary mask. Background subtracted donor and acceptor images were then multiplied with the binary mask to get processed donor and acceptor images.
  • This processed donor (D) image was divided by the processed acceptor (A) image to get a pseudocolor D/A image, using Image calculator module of ImageJ.
  • the pH value was calculated by using the equation (2) formulated from in vivo and in vitro pH calibration plot.
  • K d of CalipHluor Ly is a function of pH and this relation is formulated by the K d calibration plot in vivo and in vitro using equation (3).
  • background was set to a non-zero value.
  • the K d image represents the affinity of CalipHluor Ly for calcium and thus compensating the calcium image (O/R) with K d would precisely represent the calcium levels at single endo-lysosomes.
  • the pH dependent K d compensation is performed according to equation (5), where 0/R max and 0/R min are calculated by incubating CalipHluor coated beads at 10mM and 1 mM respectively. Image calculation were done using image calculator module in ImageJ. This image is multiplied with binary image to bring the background value to zero.
  • the pH corrected K d image were obtained for various mutants for accurate comparison of calcium levels in lysosomes.
  • cup-5(ok1698)/mT1 [dpy-10(e128)] III nematode strain was used for this assay. Homozygous lethal deletion of cup-5 gene is balanced by dpy-10- marked translocation. Heterozygotes are superficially wildtype [cup5+/-], Dpys (mT1 homozygotes) are sterile, and cup-5(ok1698) homozygotes are lethal. cup5+/- L4 worms were placed on plates containing RNAi bacterial strains for L4440 empty vector (positive control), mrp-4, catp-6, catp-5 and clh-6.
  • arls37 [myo-3p::ssGFP + dpy-20(+)]
  • cup- 5(ar465) is transgenic nematode strain which secretes GFP from the body muscle cells and this is endocytosed by coelomocytes which show enlarged GFP labelled vesicles as a result of defective degradation caused by cup 5 mutation.
  • arls37 ; cup-5(ar465) L4 worms were placed on plates containing RNAi bacterial strains for empty vector (control), catp-6, catp-5 and mrp-4 (positive control). The worms lay eggs for 24 hours after which they are removed from the plates.
  • the eggs thus hatch and grow to adulthood after which they were imaged to check for lysosomal size differences.
  • Worms were imaged on a Leica TCS SP5 II STED laser scanning confocal microscope (Leica Microsystems, Inc., Buffalo Grove, IL) equipped with 63*, 1.4 NA, oil immersion objective upon excitation with Argon laser in the Alexa 488 channel. Lysosomal areas were measured using ImageJ. Out 100 lysosomes in arls37 worms, 7 lysosomes had an area in range of 7.0-9.5 pm 2 . Enlarged lysosomes are defined as those lysosomes whose diameter is > 33% of the diameter of the largest lysosome observed in normal N2 worms.
  • HEPES mM
  • MES mM
  • sodium acetate 10 mM
  • EGTA EGTA
  • KCI 140 mM
  • NaCI 500 mM
  • MgCI 2 MgCI 2
  • the amount of [Ca 2+ ] was varied from about OmM to 10 mM and adjusted the pH to either pH 4.6 or pH 5.1.
  • the beads were incubated in clamping buffer for 30 mins after which there were imaged on a slide on the IX83 inverted microscope in the G, O and R channels to obtain G/R (pH) and O/R (Ca 2+ ) images.
  • BAPTA-5F aldehyde (1) was synthesized according to previous reported procedure (Grynkiewicz et al. (1985) J. Biol. Chem. 260:3440-3450; Collot et al. (2015) eUfe
  • the crude product was diluted with a saturated solution of Na 2 S 2 0 3 to quench the unreacted iodine followed by extraction of the compound with ethyl acetate (3x50 ml_). This was dried over Na 2 S0 4 and the product 1-azido-3- iodopropane was used for further reactions without purification.
  • Rhod-5F-OMe (5 mg, 0.005 mmol) was dissolved in methanol and water mixture (1 :0.5 ml_) and KOH (3.5 mg, 0.063mmol) was added and allowed to stir for 8 hours at room temperature. After completion of the reaction, pH was adjusted to 6.0 and crude Rhod-5F-N 3 was extracted with dichloromethane (3x5 ml_). Product was purified by HPLC (1 : 1 acetonitrile: water, 0.1 % TFA). LCMS (ESI) m/z. [M] + calculated for C 4 H FN 10 OI I + 897.33, found: 897.5.
  • Rhod- 5F conjugation was confirmed by gel electrophoresis by running a native polyacrylamide gel containing 15% (19: 1 acrylamide: bis-acrylaimde) in 1 c TBE buffer (Tris HCI (100 mM), boric acid (89 mM), EDTA (2 mM), pH 8.3).
  • Rhod-5F conjugated strand was further confirmed by recording the gel in TMR channel, where the lower mobility band shows strong fluorescence.
  • Rhod-5F labeled strand was purified and hybridized with normalizing and pH sensing module as described in methods section.
  • 12% Native PAGE was run to characterize the formation of complete sensor.
  • 12% Acrylamide:bisacrylamide resolves duplex DNA from ssDNA.
  • Example 5 In vitro characterization of nucleic acid complexes of the disclosure
  • CalipHluor Ly is a 57-base pair DNA duplex comprising two strands D1 and D2 and bears three distinct domains ( Figure 1A, Table 1).
  • the first domain in CalipHluor Ly is a Ca 2+ - reporter domain that uses a novel small molecule that functions as a Ca 2+ indicator denoted Rhod-5F.
  • Rhod-5F In the absence of Ca 2+ , the rhodamine fluorophore in Rhod-5F is quenched by photoinduced electron transfer (PeT) from the BAPTA core. Upon Ca 2+ chelation quenching is relieved resulting in high fluorescence. Note that protonation of the amines in BAPTA also relieves PeT. Thus, the percentage change in signal as well as the dissociation constant (K d ) in Rhod-5F will be affected as a function of pH. The K d of Rhod- 5F for Ca 2+ binding is pH dependent and shown in Figure 6.
  • Rhod-5F is attached to the D2 strand bearing a dibenzocyclooctyne (DBCO) group using click chemistry. Conjugation to D2 did not change the K d of Rhod-5F in CalipHluor Ly ( Figure 1 B). In CalipHluor Ly Rhod-5F (O, orange diamond) shows a K d of 1.1 mM at pH 7.2 which increases as acidity increases ( Figure 1 B). CalipHluor Ly was characterized by gel electrophoresis ( Figure 7A-7B).
  • Alexa 647 was chosen for its negligible spectral overlap with Rhod-5F and insensitivity to pH, Ca 2+ and other ions (sphere, Figure 1A).
  • the fixed stoichiometry of Alexa 647 efficiently corrects for Rhod-5F intensity changes due to inhomogeneous probe distribution in cells, thus making the ratio of Rhod-5F (O) and Alexa 647 (R) intensities in CalipHluor probes proportional to pH and Ca 2+ .
  • the second domain (gray line) constitutes a DNA based pH-reporter domain as previously described, called the l-switch ( Figure 1A). This /- switch has been used to map pH in diverse endocytic organelles in living cells.
  • CalipHluor is comprises three strands 01 , 02, and 03 ( Figure 7C; Table 1). CalipHluor were characterized by gel electrophoresis ( Figure 7D-7E).
  • the third‘integration’ domain comprises a 30-mer duplex that integrates the pH and the Ca 2+ reporter domains into a single DNA assembly. One end is fused to the l-switch and the other is fused to the Ca 2+ sensor. This domain also helps in targeting, because its anionic nature aids recognition and trafficking by scavenger receptors in a DNA sequence independent manner (Surana et al. (2011) Nat. Cornmun. 2:340).
  • Example 6 In vivo performance of nucleic acid complexes of the disclosure
  • the worms were then imaged in four channels; (i) the donor channel (D or Alexa 488) (ii) the FRET acceptor channel (A), which corresponds to the intensity image of A647 fluorescence upon exciting A488, (iii) the orange channel (O or Rhod-5F), and (iv) the red channel (R) which
  • Figure 2A (i-iv) shows representative images of a CalipHluor Ly labeled coelomocyte imaged in the four channels.
  • the calibrated D/A and O/R ratios were calculated by measuring the intensity values at single lysosome resolution in all four channels, as described in methods. Plotting D/A against pH values, at different Ca 2+ concentration shows the insensitivity of pH sensing module towards Ca 2+ levels.
  • Figures 9A-9D show the comparison of D/A vs pH plots at different Ca 2+ levels between in vitro and in vivo. Non-significant change in fold change and pH 1 2 , shows the robustness of the nucleic acid complexes of the disclosure in biological systems.
  • Ca 2+ clamping of coelomocytes was performed in Ca 2+ clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI2 (1 mM)] by varying amount of free [Ca 2+ ] from 1 mM to 10 mM and adjusting pH (5.5-6.5) in presence of nigericin (50 mM), monensin (50 pM) and ionomycin (20 pM).
  • HEPES Ca 2+ clamping buffer
  • Endosomal maturation critical to both organelle function and cargo trafficking, is accompanied by progressive acidification of the organelle lumen (Figure 3A). Unlike pH, little is known about lumenal Ca 2+ changes as a function of endosomal maturation. The applicability of the probe was demonstrated across a range of acidic organelles by mapping lumenal Ca 2+ as a function of endosomal maturation. The time points at which CalipHluor Ly localized in the early endosome, the late endosome and the lysosome were determined in coelomocytes as described previously (Surana et al. (2011) Nat. Commun. 2:340) ( Figures 3B-3C).
  • Table 3 provides mean pH and free [Ca 2+ ] in EE, LE and Ly of wild type (N2) worms, lysosomes of catp-6, cup-5 7- and catp-6 RNAi in cup-5 7- worms using CalipHluor Ly . pH decreases progressively with endosomal maturation, with lumenal acidity showing a about 3- fold decrease at each endocytic stage. In contrast, Ca 2+ in the early endosome and the late endosome were comparable and fairly low i.e. , 0.3 mM.
  • n 15 cells, 50 endosomes; data represent the mean ⁇ S.E.M.
  • Example 8 Catp-6 is identified as a potential lysosomal Ca 2+ importer
  • P-type ATPases in the human lysosomal proteome were manually identified. It was found that the P5-ATPase ATP13A2 was described to transport cations like Mn 2+ , Zn 2+ ,
  • C. elegans has two homologs of ATP13A2 i.e., catp-5 and catp-6 ( Figure 4A).
  • catp-6 mediated lysosomal Ca 2+ accumulation
  • Figure 4B To test whether catp-6 mediated lysosomal Ca 2+ accumulation, it was investigated whether its knockdown would rescue a phenotype arising due to high lumenal Ca 2+ ( Figure 4B).
  • TRPML1 is a well-known lysosomal Ca 2+ release channel whose knockdown would be expected to elevate lysosomal Ca 2+ . Mutations in TRPML1 result in lysosomal dysfunction that leads to the lysosomal storage disease Mucopolysaccharidoses Type IV (MPS IV). In C. elegans, loss of cup-5, the C. elegans homolog of TRPML1 , results in lysosomal storage and embryonic lethality. Therefore it was also tested whether catp-6 knockdown a cup-5+/- genetic background could reverse cup-5 -I- lethality. In this strain, the homozygous lethal deletion of cup-5 is balanced by dpy-10 marked translocation. A survival assay was performed by knocking down specific genes in cup-5+/- worms and scoring for lethality ( Figure 4C and Figure 11).
  • Multidrug resistance protein-4 (MRP-4) is a versatile efflux transporter for drugs, toxins, peptides and lipids and is known to rescue cup-5-/- lethality. It is hypothesized that in the absence of cup-5, mrp-4 mis-localizes in endocytic compartments causing toxicity that is then alleviated upon its knockdown. RNAi knockdown of either catp-6 or catp-5 rescued cup- 5-/- lethality favorably compared to mrp-4 knockdown ( Figure 11). Knocking down clh-6, another lysosome-resident channel that regulates lumenal chloride, showed no such rescue.
  • Example 9 Catp-6 facilitates lysosomal Ca 2+ accumulation
  • RNAi knockdowns of catp-6 in these nematodes rescued lysosomal morphology ( Figures 4D-4E).
  • Knocking down either catp-5 or mrp-4 showed only a marginal recovery of phenotype.
  • mrp-4 is not a lysosome resident protein and its inability rescue the lysosomal phenotype suggests that mechanistically, its rescue of cup-5 -/- lethality is likely to be extra lysosomal, consistent with previous hypotheses.
  • Parkinson's disease (PD) susceptibility locus These mutations lead to the Kufor-Rakeb syndrome, a severe, early onset, autosomal recessive form of PD with dementia.
  • PD is strongly connected to Ca 2+ dysregulation as excessive cytosolic Ca 2+ causes excitotoxicity of dopaminergic neurons.
  • Overexpressing ATP13A2 suppresses toxicity and reduces cytosolic Ca 2+ .
  • loss of ATP13A2 function leads to neuronal ceroid lipofuscinosis, a lysosomal storage disorder, implicating the lysosome as its potential site of action.
  • CalipHluor mLy was localized in lysosomes of primary human dermal fibroblasts (HDF cells) obtained from punch-skin biopsies. CalipHluor mLy labels lysosomes in HDF cells by scavenger receptor mediated endocytosis ( Figures 5A-5B; Figure13). Briefly, a 1 hour pulse of 500 nM CalipHluor mLy followed by a 9 hour chase efficiently labels lysosomes in this cell type ( Figures 5A-5B).
  • Lysosomal Ca 2+ was measured in fibroblasts from normal individuals and L6025 primary fibroblasts isolated from male patients with Kufor Rakeb syndrome, that are homozygous for a OT mutation in 1550 of ATP13A2. This mutation results in ATP13A2 being unable to exit the ER and the lysosomes are devoid of ATP13A2.
  • a OT mutation results in ATP13A2 being unable to exit the ER and the lysosomes are devoid of ATP13A2.
  • Figures 5B-5D Lysosomes in KRS patients showed 14-fold lower Ca 2+ and about 2-fold lower [H + ] than normal ( Figures 5C-5E) confirming that ATP13A2 mediates lysosomal Ca 2+ accumulation.
  • ChloropHore is a 61-base pair DNA duplex comprising three strands C1 , C2 and P (Table 4) and bears three distinct domains ( Figure 14A). Two of these are fluorescent, ratiometric reporter domains that are previously reported, namely a CF reporter domain, Clensor and a pH-reporter domain called the l-switch. Each reporter domain is fused to either end of an“integration domain”, which comprises a 27-mer duplex, that serves to integrate the pH and the CF reporter domains into a single DNA assembly. This 27-mer duplex also helps in targeting, because its anionic nature aids recognition and trafficking by scavenger receptors in a DNA sequence independent manner.
  • ChloropHore Ly a variant that used modified 5'-bromocytosines in the C-rich region, was also made (Table 4). Sequences used for ChloropHore LY assembly are oligo P, oligo C1-Br and oligo C2.
  • CD circular dichroism
  • ChloropHore shows a positive peak at 272 nm and a negative peak at 248 nm in pH 7.5 characteristic of duplex DNA. However, at pH 4.0, a positive peak at 285 nm and a negative peak at 248 nm was observed. The difference spectra of ChloropHore at pH 4.0 and pH 7.5 showed a positive peak at 292 nm and a negative peak at 263 nm which consistent with the CD signature of an i-motif. This was also how i-motif formation was proved in the parent /- switch.
  • ChloropHore to various ions such as Mg(N0 3 ) 2 , Ca(N0 3 ) 2 , NaN0 3 , Na 2 HP0 4 , MgS0 4 , NaHC0 3 and CH 3 COONa and 30% glycerol was also investigated (Figure 20). This reveals that the sensitivity of ChloropHore to diverse biologically abundant ions including Na + , Ca 2+ , Mg 2+ , N0 3 - and P0 4 3- is negligible. BAC is also quenched by halides such as bromide and iodide (Brand I-).
  • the [CF] sensing characteristics of ChloropHore and ChloropHore Ly are insensitive to changes in physiological pH. This indicates that in ChloropHore and ChloropHore Ly , the sensing characteristics of the H + sensing module is unaffected by integration to the CF sensing module and vice versa thus enabling potential detection of these two ions in parallel.
  • ChloropHore and TMR- Dextran showed maximal co-localization at 9 h ( Figures 15F-15G, Figure 22). Stability measurements revealed that ChloropHore and ChloropHore Ly had a half-life of 20 hours and were stable for at least 10 h in HDF lysosomes ( Figure 22).
  • FIG. 16A shows representative fluorescence images of a cell clamped at the indicated pH and [CF] imaged in the D, A, R and G channels along with the corresponding D/A and R/G maps. Histograms of D/A and R/G values of 150 endosomes clamped at different pH and [Cl-] are shown in Figure 23.
  • ChloropHore showed a ⁇ 5.5 fold- change in D/A value from pH 4.0 to pH 7.0 across all tested values of [Cl-], with the in vitro 3D surface plot being quantitatively recapitulated in cells ( Figure 16B, Figure 24).
  • ChloropHore response in terms of R/G as a function of [CF] was performed at different fixed values of physiological pH ( Figure 16C).
  • Both the K sv and fold-change in R/G from 5 mM to 120 mM [CF] was constant from pH 4.0-7.0, with the in vitro 3D surface plot of R/G being quantitatively recapitulated in cells (Figure 16C, Figure 24). This revealed that ChloropHore and ChloropHore Ly can simultaneously report pH and [CF] with performance characteristics in cells that closely match their in vitro pH and [CF] sensing properties.
  • FIG. 16F-16H shows the 2-IM profile of 150 endosomes clamped at the same [CF] (100 mM) but different pH
  • Figures 16I-16K shows the 2-IM profile of 150 endosomes clamped at the same pH (pH 6.5), but different [CF]
  • Figure 16M shows a plot of individual endosomes in each clamped state, with black lines connecting the same endosome in either clamped state. It is clear that when [CF] was constant and pH changed, all the endosomes show increased D/A and negligible variation of R/G, moving parallel to the Y-axis.
  • Figure 16N(i— iv) shows the D/A and R/G maps of cells clamped at 100 mM CF and pH 6.5 that have subsequently been clamped at the same value of pH, but at 5 mM [CF]
  • pH was constant and [CF] was changed, individual endosomes moved from right to left, parallel to the X-axis ( Figure 160).
  • 2-IM provides information with single endosome resolution.
  • NP-A and NP-B Niemann Pick A and Niemann Pick B (NP-B) diseases arise due to defects in the enzyme acid sphingomyelinase (ASM), for which enzyme replacement therapy is available.
  • ASM acid sphingomyelinase
  • NP-C Niemann Pick C arises due to defective cholesterol transport due to mutations in any one of two key proteins, NPC1 or NPC2, and for which clinical trials using cyclodextrin derivatives are under way.
  • Three patient samples corresponding to NP-A and NP-C disease and two for NP-B were studied based on sample availability, common mutations and characterization by enzyme activity (Table 5). Table 5
  • lysosomal pH correlates with spatial position in certain cell types, no such dependence is available yet for lysosomal Cl-.
  • Peripheral lysosomes show lower acidity in C2C12 murine myoblasts, human adipose microvascular endothelial cells and HeLa cells, but not primary human dendritic cells or CHO cells.
  • 2-IM profiles in primary HDF cells showed that peripheral lysosomes had higher pH than perinuclear lysosomes ( Figure 17A(i), arrowhead).
  • rhASM human acid sphingomyelinase
  • NP-C does not arise from the deficiency of a degradative enzyme, but rather due to a transport defect, for which there is still no FDA approved therapeutic.
  • o-2- hydroxypropyl ⁇ -cyclodextrin (bq ⁇ ) treatment for 24 h has been shown to improve cholesterol transport from the lysosome to the endoplasmic reticulum thereby delivering cholesterol to the ER for esterification and reducing storage in the lysosome.
  • nucleic acid complex comprising
  • a first single-stranded nucleic acid molecule comprising a Ca 2+ fluorophore or a Cl- fluorophore crosslinked to the first strand;
  • nucleic acid complex further comprises a first label conjugated to the first single-stranded nucleic acid molecule or the second single- stranded nucleic acid molecule and the first label is capable of producing a signal, wherein the intensity of the signal is dependent on change in pH; measuring the intensity of the signal; and
  • Embodiment 2 provides the method of embodiment 1 , wherein the sample is a biological sample selected from a cell, cell extract, cell lysate, tissue, tissue extract, bodily fluid, serum, blood, and blood product.
  • Embodiment 3 provides the method of embodiment 1 , wherein the sample is a live cell.
  • Embodiment 4 provides the method of any of embodiments 1-3, wherein determining is in early endosome, late endosome, plasma membrane, lysosome, autophagolysosome, recycling endosome, cis Golgi network, trans Golgi network, endoplasmic reticulum, peroxisomes, or secretory vesicles.
  • Embodiment 5 provides the method of any of embodiments 1-4, wherein the nucleic acid complex comprises the Cl- fluorophore crosslinked to the first strand.
  • Embodiment 6 provides the method of embodiment 5, wherein the Cl- fluorophore comprises 10,10'-bis[3-carboxypropyl]-9,9'-biacridinium dinitrate.
  • Embodiment 7 provides the method of any of embodiments 1-4, wherein the nucleic acid complex comprises the Ca 2+ fluorophore crosslinked to the first strand.
  • Embodiment 8 provides the method of embodiment 7, wherein the Ca 2+ fluorophore is a single wavelength indicator.
  • Embodiment 9 provides the method of any of embodiment 7, wherein the Ca 2+ fluorophore comprises Rhod-5F, XRhod-5F, Rhod-FF, XRhod-FF, Oregon Green 488 BAPTA-6F, Fluo 5F, Fluo 4FF, Oregon Green BAPTA-5N, Fluo-5 N, or Mag-Fluo-4 indicator.
  • the Ca 2+ fluorophore comprises Rhod-5F, XRhod-5F, Rhod-FF, XRhod-FF, Oregon Green 488 BAPTA-6F, Fluo 5F, Fluo 4FF, Oregon Green BAPTA-5N, Fluo-5 N, or Mag-Fluo-4 indicator.
  • Embodiment 10 provides the method of any of embodiments 7-9, wherein the Ca 2+ fluorophore comprises Rhod-5F, XRhod-5F, or Rhod-FF indicator.
  • Embodiment 11 provides the method of any of embodiments 7-9, wherein the Ca 2+ fluorophore comprises Rhod-5F indicator.
  • Embodiment 12 provides the method of any of embodiments 7-11 , wherein the Ca 2+ fluorophore is crosslinked to the first strand through a triazole, thioether, or alkenyl sulfide group.
  • Embodiment 13 provides the method embodiment 12, wherein the Ca 2+ fluorophore further comprises a linker moiety configured to form the triazole, thioether, or alkenyl sulfide group through a reaction of an azide or thiol moiety on the Ca 2+ fluorophore and a alkyne or alkene moiety on the first strand.
  • a linker moiety configured to form the triazole, thioether, or alkenyl sulfide group through a reaction of an azide or thiol moiety on the Ca 2+ fluorophore and a alkyne or alkene moiety on the first strand.
  • Embodiment 14 provides the method of any of embodiments 7-13, wherein the crosslinked Ca 2+ fluorophore is:
  • Embodiment 15 provides the method of any of embodiments 7-14, wherein the first single-stranded nucleic acid molecule comprising a Ca 2+ fluorophore is of formula:
  • Embodiment 16 provides the method of any of embodiments 7-11 , wherein the Ca 2+ fluorophore is crosslinked to the first strand through a linker moiety stable under
  • Embodiment 17 provides the nucleic acid complex of any of embodiments 1-16, wherein the first label comprises Oregon green, FITC, or pHrhodo label.
  • Embodiment 18 provides the method of any of embodiments 1-16, wherein the intensity of the signal dependent on change in pH varies as a function of the conformation of the nucleic acid complex.
  • Embodiment 19 provides the method of embodiment 18, wherein the second single- stranded nucleic acid molecule comprising the sequence C n XC n YC n ZC n , (SEQ ID NO: 13), wherein C is cytosine; X, Y, and Z are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
  • Embodiment 20 provides the method of embodiment 19, wherein each of X, Y, and Z is independently AA or TAA.
  • Embodiment 21 provides the method of embodiment 18, wherein the second single- stranded nucleic acid molecule comprising the sequence C n XC n , wherein C is cytosine; X and Y are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
  • Embodiment 23 provides the method of any of embodiments 19-22, wherein each n is independently 3, 4, or 7.
  • Embodiment 24 provides the method of any of embodiments 19-23, wherein the modification is selected from one or more of a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group.
  • Embodiment 25 provides the method of any of embodiments 19-23, wherein the modification is a methyl or bromo group.
  • Embodiment 26 provides the method of any of embodiments 19-23, wherein the modification is at the 5' position of the cytosine.
  • Embodiment 27 provides the method of any of embodiments 1-26, wherein the nucleic acid further comprises a second label conjugated to the first single-stranded nucleic acid or the second single-stranded nucleic acid.
  • Embodiment 28 provides the method of embodiment 27, wherein the intensity of the signal varies as a function of at least one of the distance between the first label and second label and the relative orientation of the first label and second label.
  • Embodiment 29 provides the method of embodiment 27, wherein the first label and second label comprise a donor and acceptor pair.
  • Embodiment 31 provides the method of any of embodiments 1-30, wherein the signal is measured at 2 different wavelengths.
  • Embodiment 33 provides the method of any of embodiments 27-32, wherein the Ca 2+ concentration is determined from the measured signal from the Ca 2+ fluorophore with the measured signal from the second label.
  • Embodiment 34 provides the method of any of embodiments 27-32, wherein the CF concentration is determined from the measured signal from the CF fluorophore with the measured signal from the first label or the second label.
  • Embodiment 36 provides the method of any of embodiments 1-34, wherein the second single-stranded nucleic acid is capable of forming an intramolecular complex comprising two parallel-stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
  • Embodiment 37 provides the method of any of embodiments 1-36, wherein the nucleic acid complex further comprises a third single-stranded nucleic acid molecule that is partially complementary to the first single-stranded molecule.
  • Embodiment 38 provides the method of embodiment 37, wherein the second single- stranded nucleic acid molecule comprising the sequence C n XC n YC n ZC n , (SEQ ID NO: 13), wherein C is cytosine; X, Y, and Z are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
  • Embodiment 39 provides the method of embodiment 38, wherein each of X, Y, and Z is independently AA or TAA.
  • Embodiment 40 provides the method of embodiment 37, wherein the third single- stranded nucleic acid molecule comprising the sequence C n XC n , wherein C is cytosine; X and Y are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
  • Embodiment 41 provides the method of embodiment 37, wherein each of X and Y are independently AA.
  • Embodiment 42 provides the method of embodiment 37 or 04, wherein each n is independently 3, 4, or 7.
  • Embodiment 43 provides the method of any of embodiments 37-42, wherein the modification is selected from one or more of a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group.
  • Embodiment 44 provides the method of any of embodiments 37-42, wherein the modification is a methyl or bromo group.
  • Embodiment 46 provides the method of any of embodiments 37-45, wherein the first label is on the second single-stranded nucleic acid.
  • Embodiment 49 provides the method of any of embodiments 37-47, wherein the second single-stranded nucleic acid or the third single-stranded nucleic acid is capable of forming an intramolecular complex comprising two parallel-stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
  • Embodiment 50 provides the method of any of embodiments 27-49, wherein the first label and the second label are independently selected from the group consisting of an Atto dye, an Alexa Flour® dye, a Cy® dye, and a BODIPY dye.
  • Embodiment 51 provides the method of any of embodiments 27-50, wherein the first label and the second label comprise a donor fluorophore and an acceptor quencher.
  • Embodiment 52 provides the method of embodiment 51 , wherein the donor fluorophore and an acceptor quencher pair are: FITC and TRITC, or Cy3 and Cy5, or Alexa- 488 and Alexa-647.
  • Embodiment 53 provides the method of any of embodiments 1-52, wherein the nucleic acid complex further comprises a targeting moiety.
  • Embodiment 55 provides the method of embodiment 53, wherein the targeting moiety has a cognate artificial protein receptor.
  • Embodiment 56 provides the method of any of embodiments 53-55, wherein the targeting moiety is encoded on the same nucleic acid strand as the first single-stranded nucleic acid molecule, the second single-stranded nucleic acid molecule, the third single- stranded nucleic acid molecule, or any combination thereof.
  • Embodiment 57 provides the method of any of embodiments 53-55, wherein the targeting moiety is selected from an aptamer, a duplex domain targeted to an artificial protein receptor, a nucleic acid sequence that binds an anionic-ligand binding receptor, and an endocytic ligand.
  • Embodiment 58 provides the method of any of embodiments 53-55, wherein the targeting moiety comprises a peptide directly or indirectly conjugated to the nucleic acid molecule.
  • Embodiment 59 provides the method of any of embodiments 53-55, wherein the targeting moiety comprises one or more of a fusogenic peptide, a membrane-permeabilizing peptide, a sub-cellular localization sequence, or a cell-receptor ligand.
  • Embodiment 60 provides the method of embodiment 59, wherein the sub-cellular localization sequence targets the nucleic acid complex to a region of a cell where spatial localization of a targeted protein is present.
  • Embodiment 61 provides the method of embodiment 60, wherein the sub-cellular localization sequence targets the nucleic acid complex to a region of the cell selected from the group consisting of: the cytosol, the endoplasmic reticulum, the mitochondrial matrix, the chloroplast lumen, the medial trans-Golgi cistemae, the lumen of lysosome, the lumen of an endosome, the peroxisome, the nucleus, and a specific spatial location on the plasma membrane.
  • Embodiment 62 provides the method of any of embodiments 1-61 , wherein the first and/or second single-stranded nucleic acid molecule is less than 200 nucleotides; or less than 100 nucleotides; or less than 50 nucleotides.
  • Embodiment 63 provides the method of any of embodiments 1-62, wherein the determined Ca 2+ concentration is in a range of 10 nM to 10 mM.
  • Embodiment 64 provides the method of any of embodiments 1-62, wherein the determined Ca 2+ concentration is in a range of 10 nM to 1 mM.
  • Embodiment 65 provides the method of any of embodiments 1-62, wherein the determined Ca 2+ concentration is in a range of 1 pM to 10 mM.
  • Embodiment 66 provides the method of any of embodiments 1-62, wherein the determined Cl- concentration is in a range of 10 nM to 10 mM.
  • Embodiment 67 provides the method of any of embodiments 1-62, wherein the determined Cl- concentration is in a range of 10 nM to 1 pM.
  • Embodiment 68 provides the method of any of embodiments 1-62, wherein the determined Cl- concentration is in a range of 1 pM to 10 mM.
  • Embodiment 69 provides the method of any of embodiments 1-68, wherein the determined pH is less than pH 5.5.
  • Embodiment 70 provides the method of any of embodiments 1-68, wherein the determined pH is more than pH 7.0.
  • Embodiment 71 provides a nucleic acid complex comprising: a first single-stranded nucleic acid molecule comprising a Ca 2+ fluorophore or a CF fluorophore crosslinked to the first strand; and
  • nucleic acid complex further comprises a first label conjugated to the first single-stranded nucleic acid molecule or the second single-stranded nucleic acid molecule and the first label is capable of producing a signal.
  • Embodiment 72 provides the method of embodiment 71 , wherein the nucleic acid complex comprises the Cl- fluorophore crosslinked to the first strand.
  • Embodiment 73 provides the method of embodiment 72, wherein the Cl- fluorophore comprises 10,10'-bis[3-carboxypropyl]-9,9'-biacridinium dinitrate.
  • Embodiment 74 provides the method of embodiment 71 , wherein the nucleic acid complex comprises the Ca 2+ fluorophore crosslinked to the first strand.
  • Embodiment 75 provides the nucleic acid complex of embodiment 74, wherein the Ca 2+ fluorophore is a single wavelength indicator.
  • Embodiment 77 provides the nucleic acid complex of embodiment 74, wherein the Ca 2+ fluorophore comprises Rhod-5F, XRhod-5F, or Rhod-FF indicator.
  • Embodiment 78 provides the nucleic acid complex of embodiment 74, wherein the Ca 2+ fluorophore comprises Rhod-5F indicator.
  • Embodiment 80 provides the nucleic acid complex of any of embodiments 74-79, wherein the crosslinked Ca 2+ fluorophore is:
  • Embodiment 81 provides the nucleic acid complex of any of embodiments 74-80, wherein the first single-stranded nucleic acid molecule comprising a Ca 2+ fluorophore is of formula:
  • R is a linker
  • Embodiment 82 provides the nucleic acid complex of any of embodiments 71-81 , wherein the first label comprises Oregon green, FITC, or pHrhodo label.
  • Embodiment 84 provides the nucleic acid complex of embodiment 83, wherein each of X and Y are independently AA or TAA.
  • Embodiment 85 provides the nucleic acid complex of any of embodiments 71-81 , wherein the second single-stranded nucleic acid molecule comprising the sequence C n XC n YC n ZC n , (SEQ ID NO: 13), wherein C is cytosine; X, Y, and Z are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
  • Embodiment 86 provides the nucleic acid complex of embodiment 85, wherein each of X, Y, and Z are independently AA or TAA.
  • Embodiment 89 provides the nucleic acid complex of any of embodiments 83-87, wherein the modification is a methyl or bromo group.
  • Embodiment 90 provides the nucleic acid complex of any of embodiments 83-89, wherein the modification is at the 5' position of the cytosine.
  • Embodiment 91 provides the nucleic acid complex of any of embodiments 83-90, further comprising a second label capable of producing a signal conjugated to the first single-stranded nucleic acid or the second single-stranded nucleic acid.
  • Embodiment 92 provides the nucleic acid complex of embodiment 91 , wherein the first label and second label comprise a donor and acceptor pair.
  • Embodiment 93 provides the nucleic acid complex of any of embodiments 83-92, wherein the first single-stranded nucleic acid molecule and second single-stranded nucleic acid are configured to form an i-motif under acidic conditions.
  • Embodiment 94 provides the nucleic acid complex of any of embodiments 83-92, wherein the second single-stranded nucleic acid is configured to form an intramolecular complex comprising two parallel-stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
  • Embodiment 95 provides the nucleic acid complex of any of embodiments 83-94 further comprising a third single-stranded nucleic acid molecule that is partially
  • Embodiment 96 provides the nucleic acid complex of embodiment 95, wherein the third single-stranded nucleic acid molecule comprises the sequence C n XC n YC n ZC n , (SEQ ID NO: 13), wherein C is cytosine; X, Y, and Z are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
  • Embodiment 99 provides the nucleic acid complex of embodiment 98, wherein each of X and Y are independently AA.
  • Embodiment 100 provides the nucleic acid complex of embodiment 96-99, wherein each n is independently 3, 4, or 7.
  • Embodiment 101 provides the nucleic acid complex of any of embodiments 96-100, wherein the modification is selected from one or more of a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group.
  • Embodiment 102 provides the nucleic acid complex of any of embodiments 96-100, wherein the modification is a methyl or bromo group.
  • Embodiment 103 provides the nucleic acid complex of any of embodiments 96-102, wherein the modification is at the 5' position of the cytosine.
  • Embodiment 104 provides the nucleic acid complex of any of embodiments 96-103, wherein the first label is on the second single-stranded nucleic acid.
  • Embodiment 105 provides the nucleic acid complex of any of embodiments 96-104, wherein the nucleic acid complex further comprises the second label conjugated to the third single-stranded nucleic acid.
  • Embodiment 107 provides the nucleic acid complex of any of embodiments 96-105, wherein the second single-stranded nucleic acid or the third single-stranded nucleic acid is configured to form an intramolecular complex comprising two parallel-stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
  • Embodiment 108 provides the nucleic acid complex of any of embodiments 71-107, wherein the first label and the second label are independently selected from the group consisting of an Atto dye, an Alexa Flour® dye, a Cy® dye, and a BODIPY dye.
  • Embodiment 109 provides the nucleic acid complex of any of embodiments 91-108, wherein the first label and the second label comprise a donor fluorophore and an acceptor quencher.
  • Embodiment 110 provides the nucleic acid complex of embodiment 109, wherein the donor fluorophore and an acceptor quencher pair are: FITC and TRITC, or Cy3 and Cy5, or Alexa-488 and Alexa-647.
  • Embodiment 111 provides the nucleic acid complex of any of embodiments 71-107, wherein the nucleic acid complex further comprises a targeting moiety.
  • Embodiment 113 provides the nucleic acid complex of embodiment 111 , wherein the targeting moiety has a cognate artificial protein receptor.
  • Embodiment 114 provides the nucleic acid complex of any of embodiments 111-113, wherein the targeting moiety is encoded on the same nucleic acid strand as the first single- stranded nucleic acid molecule, the second single-stranded nucleic acid molecule, the third single-stranded nucleic acid molecule, or any combination thereof.
  • Embodiment 116 provides the nucleic acid complex of embodiment 115, wherein the targeting moiety comprises a peptide directly or indirectly conjugated to the nucleic acid molecule.
  • Embodiment 118 provides the nucleic acid complex of embodiment 117, wherein the sub-cellular localization sequence targets the nucleic acid complex to a region of a cell where spatial localization of a targeted protein is present.
  • Embodiment 119 provides the nucleic acid complex of embodiment 1 18, wherein the sub-cellular localization sequence targets the nucleic acid complex to a region of the cell selected from the group consisting of: the cytosol, the endoplasmic reticulum, the mitochondrial matrix, the chloroplast lumen, the medial trans-Golgi cistemae, the lumen of lysosome, the lumen of an endosome, the peroxisome, the nucleus, and a specific spatial location on the plasma membrane.
  • Embodiment 120 provides the nucleic acid complex of embodiment 71 , wherein the first strain has the sequence 5’-the Ca 2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and
  • the second strain has the sequence 5’-the first label-CCC CTA ACC CCT AAC CCC TAA CCC CAT ATA TAT CCT AGA ACG ACA GAC AAA CAG TGA GTC-3’(SEQ ID NO:08).
  • Embodiment 121 provides the nucleic acid complex of embodiment 71 , wherein the first strain has the sequence 5’-TTA TAG GAT CCT GCG GTC GG/the Ca 2+ fluorophore/ GGC ACC AGG CGT AAA ATG TA-3’(SEQ ID NO:09);
  • the second strain has the sequence: 5’-the first label-CCC CAA CCC CAA TAC ATT TTA CGC CTG GTG CC-3’ (SEQ ID NO: 10);
  • the second strain has the sequence 5'- the first label - AT AAC ACA TAA CAC ATAACAAAA TAT ATA TCC TAG AAC GAC AGA CAA ACA GTG AGT C - 3' (SEQ ID NO: 12).
  • Embodiment 123 provides the nucleic acid complex of embodiment 71 , wherein the first strain has the sequence 5’-the Cl- fluorophore-ATC AAC ACT GCA-Lys-COOH
  • the second strain has the sequence 5'-TAT TGT GTA TTG TGT ATT GTT TTA TAT AT /the first label/ A TAG GAT CTT GCT GTC TGG TGT GCA GTG TTG AT-3'(SEQ ID NO:23); and
  • the third strain has the sequence: 5’-CAC CAG ACA GCA AGA TCC TAT ATA TAT ACC CCA ATC CCC AAT CCC CAA TCC CC-the second label-3' (SEQ ID NO:24).
  • Embodiment 124 provides the method of embodiment 1 , wherein the first strain has the sequence 5’-the Ca 2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and
  • the second strain has the sequence 5’-the first label-CCC CTA ACC CCT AAC CCC TAA CCC CAT ATA TAT CCT AGA ACG ACA GAC AAA CAG TGA GTC-3’(SEQ ID NO:08).
  • Embodiment 125 provides the method of embodiment 1 , wherein
  • the second strain has the sequence: 5’-the first label-CCC CAA CCC CAA TAC ATT TTA CGC CTG GTG CC-3’ (SEQ ID NO: 10);
  • the third strain has the sequence: 5’-CCG ACC GCA GGA TCC TAT AAA ACC CCA ACC CC-the second label-3 (SEQ ID NO: 11).
  • Embodiment 126 provides the method of embodiment 1 , wherein
  • the first strain has the sequence 5’-the Ca 2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07);
  • the second strain has the sequence 5'- the first label - AT AAC ACA TAA CAC ATAACAAAA TAT ATA TCC TAG AAC GAC AGA CAA ACA GTG AGT C - 3' (SEQ ID NO: 12).
  • Embodiment 127 provides the method of embodiment 1 , wherein
  • the second strain has the sequence 5'-TAT TGT GTA TTG TGT ATT GTT TTA TAT AT /the first label/ A TAG GAT CTT GCT GTC TGG TGT GCA GTG TTG AT-3'(SEQ ID NO:23); and
  • the third strain has the sequence: 5’-CAC CAG ACA GCA AGA TCC TAT ATA TAT ACC CCA ATC CCC AAT CCC CAA TCC CC-the second label-3' (SEQ ID NO:24).

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Food Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Inorganic Chemistry (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Investigating, Analyzing Materials By Fluorescence Or Luminescence (AREA)

Abstract

This disclosure relates to methods for determining pH and also calcium (Ca2+) concentration or chloride (Cl-) concentration in biological samples. More particularly, this disclosure relates to methods capable of simultaneously determining pH and Ca2+ concentration, or pH and Cl- concentration using nucleic acid complexes.

Description

METHODS OF DETERMINING PH AND CALCIUM OR CHLORIDE
CONCENTRATION IN SAMPLES
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of priority of U.S. Provisional Patent Application No. 62/774,314, filed December 2, 2018, all of which is incorporated by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] This invention was made with United States government support awarded by National Center for Advancing Translational Sciences (NCATS) of the National Institutes of Health (NIH) Grant No. 1 UL1TR002389-01. The United States government has certain rights in this invention.
BACKGROUND OF DISCLOSURE
Field of Disclosure
[0003] This disclosure relates to methods for determining pH and also calcium (Ca2+) concentration or chloride (Cl-) concentration in biological samples. More particularly, this disclosure relates to methods capable of simultaneously determining pH and Ca2+ concentration, or pH and Cl- concentration using nucleic acid complexes.
Technical Background
[0004] Lysosomes are highly fusogenic organelles that regulate cellular processes such as innate immunity by fusion with the phagosome, cell membrane repair through fusion with the plasma membrane, autophagy by fusion with the autophagosome and nutrient sensing through the mTOR pathway. Lysosome dysfunction is central to the pathology of common neurological disorders such as Alzheimer’s disease, Parkinson’s disease as well as about 60 rare, largely untreatable genetic diseases called lysosomal storage diseases. It has been challenging to deconvolute how each of its multiple roles are affected in the diverse pathophysiologies associated with lysosome-related diseases.
[0005] Lysosomes are, by and large, regarded as a single population while assaying for a specific lysosomal function. However, recent promising studies have considered that sub populations of lysosomes might perform sub-sets of tasks. Indeed, many cell types have evolved specialized lysosomes that perform distinct functions. For instance in addition to lysosomes, skin cells have melanosomes, neutrophils have azurophil granules, cytotoxic T- cells have secretory lysosomes while every cell has autolysosomes. Functional imaging based on physical parameters such as lysosome movement, morphology or spatial position within cells have revealed sub-populations that exhibit different behaviors and functions. For example, autolysosomes and lysosomes adopt tubulovesicular and vesicular morphologies respectively. Lysosomes have also been sorted into two populations based on how actively they move within the cell. Spatial positioning of lysosomes is emerging as a correlate of lysosome function. Nevertheless, the capacity to chemically discriminate between lysosome populations in live cells would significantly aid the understanding of lysosome biology by providing the ability to quantitatively correlate chemotypes with function. For example, in 1960s electron microscopy and bright field imaging could only distinguish up to three stages in melanosome maturation based on morphology and melanin content respectively.
However, when protein markers were used to chemotype melanosomes, it revealed four stages in melanosome maturation. Chemical resolution revealed the colorless, stage I melanosome that had eluded identification till then due to its high physical and morphological similarity with lysosomes. However, there are still no methods to chemically resolve lysosome populations.
[0006] Specialized lysosomes have a different protein composition from normal lysosomes to enable the distinct biochemistries within their lumens. This lumenal biochemistry is facilitated by an optimal chemical milieu, of which key components are high concentrations of specific ions homeostatically maintained by the lysosome protein composition.
[0007] H+ and Cl- are two highly abundant ions in the lysosome that are critical to its function. In fact, no other organelle has a greater concentration of either ion. Lysosomal pH is critical to lysosome maturation, cargo degradation and recycling of degraded material.
High lumenal CF in the lysosome is required for the activity of certain lysosome-resident hydrolases. Unlike other organelles, however, lumenal Cl- level in the lysosome is independent of lumenal pH.
[0008] Ca2+ regulates diverse cellular functions upon its controlled release from different intracellular stores that initiates signaling cascades. Lysosomes have recently been recognized as“acidic Ca2+ stores”, and lumenal Ca2+ is central to its diverse functions. For example, risk genes for Parkinson's disease such as LRRK2, ATP6AP2, ATP13A2, and genetic risk associated GBA1 gene, are predicted to act in lysosomal pathways.
[0009] Although electrophysiology has enabled the discovery of several channels that release lysosomal Ca2+, mediators of lysosomal Ca2+ import have not yet been identified. Lysosomal Ca2+ release channels are amenable to investigation because Ca2+ release can be tracked using cytosolic Ca2+ dyes or genetically encoded Ca2+ indicators anchored to the cytoplasmic face of the lysosome. Upon Ca2+ release, these probes indicate cytosolic Ca2+ in the area surrounding lysosomes. In contrast, lumenal Ca2+ cannot be quantitated, impeding the study of lysosomal Ca2+ importers. Consequently, lysosomal Ca2+ importers have not yet been identified in animals, with the closest evidence being that the Xenopus CAX gene localizes in lysosomes upon overexpression.
[0010] The inability to quantify Ca2+ in acidic organelles arises because all Ca2+ indicators function by coordinating Ca2+ through carboxylate groups that get protonated at acidic pH. This changes probe affinity to Ca2+ ions. Further, organellar pH is coupled to lumenal Ca2+ entry and exit. Thus, it is non-trivial to deconvolute the contribution of Ca2+ to the observed fluorescence changes of any Ca2+ indicator. Previous attempts used endocytic tracers bearing either pH or Ca2+ sensitive dyes to serially measure population-averaged pH and apparent Ca2+ in different batches of cells thus, scrambling information from individual endosomes. Given the broad pH distribution in endocytic organelles, this approach does not provide the resolution needed to study Ca2+ import.
SUMMARY OF THE DISCLOSURE
[0011] The inventors have determined that the novel nucleic acid complexes of the disclosure can efficiently and accurately determine pH in addition to Ca2+ concentration or CF concentration in samples. In certain embodiments, the novel nucleic acid complexes of the disclosure simultaneously determine pH and Ca2+ concentration, or pH and CF concentration in samples.
[0012] Thus, one aspect of the disclosure provides nucleic acid complexes including:
a first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore or a CF fluorophore crosslinked to the first strand; and
a second single-stranded nucleic acid molecule that is partially or fully
complementary to the first single-stranded molecule,
wherein the nucleic acid complex further comprises a first label conjugated to the first single-stranded nucleic acid molecule or the second single-stranded nucleic acid molecule and the first label is capable of producing a signal.
[0013] Another aspect of the disclosure provides methods of simultaneously determining 1) pH, and 2) Ca2+ concentration or CF concentration in samples using the nucleic acid complexes of the disclosure as provided herein. In general, such methods include providing a nucleic acid complex of the disclosure comprising a Ca2+ fluorophore or CF fluorophore and a first label capable of producing a signal; measuring the intensity of the signal; and determining the pH, and Ca2+ or Cl- concentration from the measured signal. Thus, in one embodiment, the methods of the disclosure include:
providing a nucleic acid complex comprising
a first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore or a Cl- fluorophore crosslinked to the first strand; and
a second single-stranded nucleic acid molecule that is partially or fully complementary to the first single-stranded molecule,
wherein the nucleic acid complex further comprises a first label conjugated to the first single-stranded nucleic acid molecule or the second single- stranded nucleic acid molecule and the first label is capable of producing a signal, wherein the intensity of the signal is dependent on change in pH; measuring the intensity of the signal; and
determining the pH, and Ca2+ or Cl- concentration from the measured signal.
[0014] Another aspect of the disclosure relates to a cell comprising a nucleic acid complex described herein.
[0015] Another aspect of the disclosure relates to a methods for screening a candidate drug in a model cell or organism, the method including delivering the nucleic acid complex of the disclosure to the cell or organism; contacting the cell or organism with the candidate drug, measuring the intensity of the signal; and determining the pH, and Ca2+ or Cl- from the measured signal. In some embodiments, the model cell or organism is a model for a lysosomal storage disease. In some embodiments, the disease is a lysosomal storage disease.
[0016] Another aspect of the disclosure relates to a method for detecting the severity of a disease, the progression of the disease, or the presence of a disease, the method including delivering the nucleic acid complex of the disclosure to a sample; measuring the intensity of the signal; and determining the pH, and Ca2+ or Cl- from the measured signal.
[0017] Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. BRIEF DESCRIPTION OF THE DRAWINGS
[0018] The accompanying drawings are included to provide a further understanding of the methods and materials of the disclosure, and are incorporated in and constitute a part of this specification. The drawings illustrate one or more embodiment(s) of the disclosure and, together with the description, serve to explain the principles and operation of the disclosure.
[0019] Figures 1A-1D illustrate design and characterization of a nucleic acid complex according to one embodiment of the disclosure, CalipHluorLy. Figure 1 A provides a working principle of CalipHluorLy. pH-induced FRET between Alexa488 (donor, D, sphere) and Alexa647 (acceptor, A, star) reports on pH ratiometrically. A Ca2+ sensitive dye (Rhod-5F, diamond, Aex=560 nm) and Alexa647 (Aex=650 nm) report Ca2+ ratiometrically by direct excitation. Figure 1 B shows fluorescence emission spectra of CalipHluorLy corresponding to Rhod-5F (left, O) and Alexa647 (right, R) with increasing [Ca2+] at pH = 7.2. Figure 1C illustrates 3D-surface plot of donor to acceptor ratio (D/A) or pH response of CalipHluorLy as a function of pH and [Ca2+] Figure 1D illustrates 3D-surface plot of the Rhod-5F to Alexa647 ratio (O/R) or Ca2+ response of CalipHluorLy as a function of pH and [Ca2+]
[0020] Figures 2A-2J illustrate in vivo sensing characteristics of a nucleic acid complex according to one embodiment of the disclosure, CalipHluorLy. Figure 2A illustrates representative CalipHluorLy labelled coelomocytes imaged in the donor (D,i), acceptor (A,ii), Rhod-5F (O, iii) and Alexa647 (R, iv) channels. D/A (v) and O/R (vi) are the corresponding pixel-wise pseudocolor images. Figure 2B illustrates representative pseudo colored D/A and O/R maps of coelomocytes clamped at indicated pH and free [Ca2+] Figure 2C shows distribution of D/A ratios of > 50 endosomes clamped at the indicated pH (n = 10 cells). Figure 2D shows distribution of O/R ratios of > 50 endosomes clamped at different indicated free [Ca2+] (n = 10 cells). Comparison of fold change of D/A ratios from pH 4 to 6.5 (Figure 2E) and pH1 2 from pH 4 to 6.5 (Figure 2F) of CalipHluorLy at different [Ca2+] obtained in vitro (light gray) and in vivo (dark gray). Figure 2G shows dissociation constant Kd (mM) and
Figure 2H shows fold change of O/R as a function of pH. Comparison of fold change in O/R ratio from 1 pM to 10 mM [Ca2+] (Figure 2I) and dissociation constant Kd (pM) (Figure 2J) of CalipHluorLy at the indicated pH obtained in vitro (light gray) and in vivo (dark gray). Scale bars, 5 pm. Data represent mean ± S.E.M. * Error is obtained from the non-exponential fit. Experiments were repeated thrice independently with similar results.
[0021] Figures 3A-3M illustrate the pH and [Ca2+] maps accompanying endosomal maturation of a nucleic acid complex according to one embodiment of the disclosure, CalipHluor. Figure 3A illustrates CalipHluor marks the indicated organelles in coelomocytes time dependently, by scavenger receptor mediated endocytosis. Figure 3B illustrates colocalization of CalipHluor and GFP-tagged markers of endocytic organelles at indicated time points post-injection in nematodes. Figure 3C illustrates quantification of colocalization provided in Figure 3B (n = 10 cells, > 50 endosomes). Figures 3D-3H illustrate pseudo color images of CalipHluorLy labelled lysosomes where the D/A map (3D) is converted to the corresponding pH map (3E), the pH map is converted into a Kd map (3F) where the value of Kd is encoded pixel-wise according to the pH at that pixel; the Kd map (3F) is multiplied by the O/R map (3G) to yield the Ca2+ map (3H). Figure 3I illustrates representative
pseudocolor pH and Ca2+ maps of early endosomes (EE), late endosomes (LE) and lysosomes (Ly) labelled with CalipHluor and CalipHluorLy. Figures 3J-3K illustrate distributions of D/A and O/R ratios of EE, LE and Ly from n = 15 cells, 50 endosomes.
Figure 3L illustrates mean endosomal pH of EE, LE and Ly. Figure 3M illustrates mean endosomal [Ca2+] in EE, LE and Ly. Data represented as the mean ± S.E.M. Experiments were repeated thrice independently with similar results.
[0022] Figures 4A-4H illustrate Catp-6 facilitates lysosomal Ca2+ accumulation. Figure 4A illustrates P-type ATPases in human lysosomes obtained from the human Lysosome Gene Database (hLGDB). Figure 4B illustrates functional connectivity between catp-6 and cup-5. Figure 4C illustrates number of adult cup-5 +/- progeny where the indicated proteins are knocked down by RNAi. Data represents mean ± S.E.M. of three independent trials. Figure 4D illustrates representative fluorescence images of arls37[myo-3p::ssGFP + dpy-20 (+)]! and arls37; cup-5(ar465) upon RNAi knockdown of the indicated proteins. Figure 4E illustrates percentage area occupied by enlarged lysosomes in the indicated genetic background (n = 15 cells, > 100 lysosomes). Figure 4F illustrates pH and Ca2+ maps in CalipHluorLy- 1 a be led lysosomes in coelomocytes in indicated genetic backgrounds. Figure 4G illustrates mean lysosomal pH in the indicated genetic backgrounds; Figure 4H
illustrates mean lysosomal [Ca2+] in the indicated genetic backgrounds. Data represent the mean ± S.E.M. Scale bar 5 pm. Experiments were repeated thrice independently with similar results.
[0023] Figures 5A-5D illustrate lysosomal Ca2+ of a nucleic acid complex according to one embodiment of the disclosure, CalipHluor171^, in human cells. Figure 5A illustrates representative images of lysosomes in fibroblast cells from normal individuals and Kufor Rakeb syndrome patients (L6025S) labelled with TMR dextran (TMR; middle panels) and CalipHluo mLy (Alexa647, left panels). Figure 5B illustrates Pearson’s correlation coefficient (PCC) of colocalization between CalipHluor mLy and lysosomes as a function of time (n = 20 cells). Figure 5C illustrates pseudocolor pH and Ca2+ maps of lysosomes in normal and L6025 fibroblasts. Figure 5D illustrates mean lysosomal pH, and Figure 5E illustrates mean lysosomal [Ca2+] in normal and L6025 fibroblasts (n = 5 cells; 50 endosomes) Scale bar: 10 pm. Data represent mean ± S.E.M. Experiments were repeated thrice independently with similar results.
[0024] Figure 6A illustrates fluorescence emission spectra of Rhod-5F (left) and Alexa 647 (right) with increasing [Ca2+] upon exciting Rhod-5F and Alexa647 at 560 nm and 650 nm respectively. Figure 6B illustrates normalized O/R ratio of Rhod-5F/Alexa 647 with increasing [Ca2+] at pH 7.2 and 5.5. Error bar represents ± S.E.M of three independent experiments.
[0025] Figures 7A-7G illustrate characterization of nucleic acid complexes according to certain embodiments of the disclosure, CalipHluorCy and CalipHluor. Figure 7A illustrates gel showing the conjugation of Rhod-5F to D2-DBCO strand. Gels were visualized in EtBr and TMR channels. Figure 7B illustrates native PAGE showing formation of CalipHluorLy. Gels were visualized in Alexa 488, TMR and Alexa 647 channels. Figure 7C is a schematic of working principle of CalipHluor. A pH-induced FRET changes between Alexa488 (donor, sphere) and Alexa647 (acceptor, star) is used to report pH ratiometrically. A Ca2+ sensitive fluorophore (Rhod-5F, diamond) and Alexa647 report Ca2+ (at a given pH) ratiometrically by direct excitation of each dye. Figure 7D illustrates gel showing the conjugation of Rhod-5F to 03-DBCO strand. Gels were visualized in EtBr and TMR channels. Figure 7E illustrates native PAGE showing formation of CalipHluor. Gels were visualized in Alexa 488, TMR and Alexa 647 channels. Figure 7F shows emission spectra of CalipHluor at pH values ranging from 7.5 to 5.0 upon excitation at 488 nm. Figure 7G shows normalized ratio of fluorescence intensity of donor to that of acceptor (D/A) of CalipHluor as a function of pH. (D Aex = 495 nm, Aem = 520 nm; A Aex = 495 nm, Aem = 665 nm). Error bar represents ± S.E.M. of three independent experiments.
[0026] Figures 8A-8B illustrate in vivo performance of a nucleic acid complex according to one embodiments of the disclosure, CalipHluorLy. Figure 8A provides representative pseudo color images of coelomocytes labeled with CalipHluorLy and clamped at the indicated pH. Figure 8B provides representative pseudo color images of coelomocytes labeled with CalipHluorLy and clamped at different free [Ca2+] at pH 5.5. Scale bar 5 pm. Experiments were repeated thrice independently with similar results.
[0027] Figures 9A-9H illustrate comparison of in vitro and in vivo pH and Ca2+ calibration profile of a nucleic acid complex according to one embodiments of the disclosure,
CalipHluorLy. Figures 9A-9D provide D/A ratios of CalipHluorLy as a function of pH clamped at different amounts of added [Ca2+] Figures 9E-9H provide normalized O/R ratios of CalipHluorLy as a function of free [Ca2+] clamped at different pH points. For in vivo n=10 worms; 15 cells and 50 endosomes were considered; in vitro n=2. Error bar represents mean ± S.E.M.
[0028] Figures 10A-10C illustrate endocytic trafficking of a nucleic acid complex according to one embodiments of the disclosure, CaiipHiuor^ 7 in coelomocytes. Representative confocal images taken 5 min, 17 min, and 60 min following injection of CaiipHiuor^ 7 in worms expressing GFP::RAB-5, GFP::RAB-7 and LMP-1 ::GFP. Scale bar 5 pm.
Experiment was performed once in n=10 worms.
[0029] Figure 11A illustrates catp-6 rescues the lethality of cup-5 +/-. Representative images showing the number of progeny of cup-5 +/- worms in plates containing RNAi bacteria of mrp-4 (positive control), clh-6, catp-6, catp-5 and e.v. (control). Experiments were repeated twice independently with similar results. Figure 11 B illustrates RT-PCR analysis of total RNA isolated from C. elegans pre- and post- RNAi. Lanes correspond to PCR-amplified cDNA of the indicated gene product isolated from wild type without RNAi treatment (denoted by gene name) and the corresponding dsRNA-fed worms (denoted as.
‘— gene name) Figure 11C shows representative images of worms expressing LMP-1 ::GFP (the left panels) in the background of various indicated RNAi, which were injected with CalipHluorLyA647 (the middle panels) and imaged 60 mins post-injection. Scale bar: 5 pm. Figure 11 D illustrates quantification of colocalization between the CalipHluorLyA647 and GFP in LMP-1 ::GFP worms. n=10 cells; error bars represent mean ± S.E.M.
[0030] Figures 12A-12H illustrate design and characterization of a nucleic acid complex according to one embodiment of the disclosure, CalipHluormLy. Figure 12A is a schematic of working principle of CalipHluorwLy. An Oregon Green based pH sensor (sphere) and ion insensitive Alexa647 (acceptor, star) and a Ca2+ sensitive fluorophore (Rhod-5F, diamond). Figures 12B and 12C provide calibration curves comparing in vitro (solid gray) and on beads (light gray) calibration at pH 4.6 and pH 5.1 , respectively. Figures 12D and 12E provide comparison of Kd and fold change (FC), respectively, in O/R of CalipHluorLy (solid) and CalipHluorwLy (hatched). Figure 12F provides representative images. Figures 12G and 12H provide comparison of fold change (FC) in O/R and Kd, respectively, of CalipHluormLy in vitro (middle gray), on beads (light gray) and in cellulo (dark grey) (n = 5 cells; 30 endosomes; n=60 beads). Experiments were performed thrice independently. *Error is obtained from Hill equation fit. Error bars represent mean ± S.E.M. Scale bar: 10pm.
[0031] Figure 13A provides images and Figure 13B graphs CalipHluormLy internalization by primary human skin fibroblasts is competed out by excess maleylated BSA (mBSA, 10 pM), revealing uptake is by scavenger receptors. Cells are imaged in Alexa647 channel. AF: autofluorescence. Scale bar: 10pm . Experiments were performed in triplicate. Error bars indicate the mean of three independent experiments ± S.E.M. (n = 25 cells)
[0032] Figures 14A-14E illustrate design and characterization of ChloropHore. Figure 14A is a schematic of the working principle: A pH-induced change in FRET between Alexa546 (donor, sphere) and Alexa647 (acceptor, star) reports pH ratiometrically. A Cl- sensitive fluorophore (BAC, triangle) and Alexa647 report Cl- ratiometrically. Figure 14B shows a pH and Cl- response profiles of ChloropHore : Normalized fluorescence intensity ratio (D/A) of donor (D) and acceptor (A) upon donor excitation in vitro as a function of pH and 50 mM Cl- (bottom, light gray). Normalized fluorescence intensity ratio (R/G) of Alexa 647 (R) and BAC (G) as a function of Cl- concentration at pH 7 (top, dark gray). Values were normalized 5 mM Cl- for R/G or pH 4 for D/A. Figure 14C shows a performance of the pH sensing module at different [Cl-] and Cl- sensing module at different pH. Fold changes in D/A (light gray hatched bars, on the right) or R/G (dark gray hatched bars, in the middle) for the pH and Cl- sensing modules are shown. Stern Volmer's constant (KSv, solid gray bars) for Cl- sensing at each pH. Calibration surface plot of the fluorescence intensity ratios of (Figure 14D) D/A and (Figure 14E) R/G of ChloropHore as a function of Cl- and pH. Error bars indicate the mean ± s.e.m. of three independent measurements.
[0033] Figures 15A-15G illustrate trafficking pathway of ChloropHore in human dermal fibroblasts. Figure 15A shows trafficking of ChloropHore along the scavenger receptor- mediated endocytic pathway. Figures 15B and 15C show ChloropHore uptake in primary skin fibroblasts (HDF cells) is competed out by excess maleylated BSA (mBSA, 10 mM).
Cells are imaged in Alexa647 channel. AF: autofluorescence. Figures 15D and 15G show ChloropHore labels lysosomes in HDF cells. Figure 15D are representative images of co localization between lysosomes of HDF cells labeled with FITC Dextran (middle panel) and LAMP-1 RFP (left panel) with the corresponding Pearson’s correlation coefficient (Figure 15E). Figure 15F are representative images of lysosomes of HDF cells labelled with TMR dextran (TMR; middle panel) and ChloropHore (Alexa647, left panel). Figure 15G is a Pearson’s correlation coefficient of colocalization between ChloropHore and lysosomes as a function of ChloropHore chase times. Experiments were performed in triplicate. Error bars indicate the mean of three independent experiments ± s.e.m. (n = 20 cells).
[0034] Figures 16A-160 illustrate intracellular calibration of ChloropHore and
ChloropHoreLy. Figure 16A are fluorescence images of primary HDF cells labeled with ChloropHore , clamped at the indicated pH and [Cl-], imaged in the donor (D), acceptor (A), reference (R), BAC (G) channel and the corresponding pseudocolour D/A (pH) and R/G (Cl-) maps. Figures 16B and 16C show in cell calibration surface corresponding to the pH and Cl- response profiles of the sensing modules in ChloropHore at various [Cl-] and pH values respectively. Figure 16D is a representative scatter plot of D/A versus the R/G values of endosomes in primary HDF cells from a normal individual clamped at the indicated pH and [Cl-]. Each data point corresponds to a single endosome. Figure 16E is the scatter plot in Figure 16D, represented as a density plot in pseudocolour where red and blue correspond to populations with higher and lower frequencies of occurrence, i.e., a 2-IM profile. Figures 16F and 16H show 2-IM profiles of HDF cells clamped in varying pH and fixed [Cl-]. Figures 161 and 16K show 2-IM profiles of HDF cells at fixed pH and increasing [Cl-]. In Figure 16A- 16K, experiments were performed in duplicate (n = 15 cells, n = 150 endosomes). Figure 16L is a single endosome clamping in HDF cells. ChloropHore labeled cells clamped at indicated pH (i) and Cl- (ii) were clamped to a different indicated pH and same Cl- (iv)
Figure 16M shows 2D-scatter plots and their projected histograms on a single axis of each clamping step of the same endosomes are shown. Gray arrow represents the direction of change in D/A (pH) values for each endosome. Figure 16N is ChlorophoreLY labeled HDF clamped at indicated pH (i) and Cl- (ii) were clamped to the same pH (iii) but different [Cl-] (iv) Figure 160 shows 2D-scatter plots and their projected histograms on a single axis for each clamping step. Gray arrow indicates direction of change in R/G (Cl-) values for each endosome. Scale bars, 10 pm. In Figures 16L-160, experiments were performed in duplicate (n = 30 endosomes).
[0035] Figures 17A-17G illustrate that 2-IM chemically resolves lysosome populations. Figure 17A is a respective pseudocolour D/A and R/G map of HDF cells derived from normal individuals 1 , NP-A patient 1 and NP-C patient 1 labeled with ChlorophoreLY. 2-IM profiles and the histograms of D/A, R/G ratios of (Figures 17B, 171) normal individual (N.l.) and in presence of bafilomycin A1 (Figure 17C) or NPPB (Figure 17D). 2-IM profiles of lysosomes of primary HDF cells from the same normal individual showing three replicates (Figure 17B, ii-vi), two different normal individuals 2-3, (Figure 17E) NP-A, NP-B, NPC patients. Figure 17F are scatter plots of lysosome sizes versus their R/G or D/A values in a normal individual and an NP-A patient. Figure 17G is a 2-IM profile and corresponding histograms of D/A, R/G ratios of HDF cells treated with 65 pM amitriptyline (AH) or 20 pM U18666A (h) 2-IM profiles of NP-A, NP-B patient fibroblasts in the presence of 5 pg of acid sphingomyelinase (ASM) and NP-C patient fibroblasts in the presence of 50 pM of b-CD. Experiments were performed in duplicate (n = 550 lysosome, = 55 cells). Scale bars 10 pm.
[0036] Figure 18 illustrates formation of ChloropHore. Gel mobility shift assay showing the formation of ChloropHore. 12% Native PAGE run in 1 * TBE buffer (pH 8.3) at 4 °C. Lane 1 : C1 2: C2, 3: C2+P 4: C1+C2+P ( ChloropHore ), in ethidium bromide, Alexa 546 and Alexa 647 channel. Experiments were performed in triplicate. [0037] Figure 19 illustrates characterization of ChloropHore by CD spectroscopy (a) CD spectra of ChloropHore at pH 7.5 (light gray), pH 4.0 (dark gray) and difference spectra (pH 4.0 - pH 7.5) is shown in black. Thermal denaturation was carried out on ChloropHore at (b) pH 4.0 as well as at (c) pH 7.5 to demonstrate i-motif formation. Experiments were performed in triplicate.
[0038] Figure 20 illustrates response of ChloropHore (200 nM) given by the fold change in R/G from ~ 0 mM to 100 mM of all indicated ions and 30% glycerol. *2 mM. Error bars indicate the mean ± s.e.m. of three independent measurements.
[0039] Figures 21A-21E illustrates design and characterization of ChloropHoreLy. Figure 21 A is a schematic of the working principle of ChloropHoreLy in the“open state” (low FRET) at high pH and in the“closed” state (high FRET) at low pH. Cl sensitive fluorophore BAC displays high fluorescence in the absence of Cl ion whereas the fluorescence has been quenched by increasing concentration of Cl ions. Both pH and Cl sensing modules are independent of each other. ChloropHoreLy incorporating brominated cytosines that has a more acidic pH sensing range Figure 21 B shows normalized fluorescence intensity ratio (D/A) of donor (D) and acceptor (A) in vitro as a function of pH. In vitro Cl calibration profile of ChloropHoreLy showing the normalized fluorescence intensity ratio (R/G) of Alexa 647 (R) and BAC (G) against increasing concentration of chloride ions. R/G values at different chloride concentrations were normalized to the value at 5 mM chloride. Figure 21 C shows the pH sensing module of ChloropHoreLy, is insensitive to Cl concentration and Cl sensing module, is insensitive to pH. Performance of the pH and Cl sensing modules are in light gray and dark gray, respectively. Plot of fold change of pH sensing modules in indicated Cl concentration (light gray hatched bars, on the right). Fold change (dark gray hatched bars, in the middle) and Ksv (solid gray) for Cl sensing module versus pH. Fluorescence intensity ratio (Figure 21 D) D/A and (Figure 21 E) R/G of ChloropHoreLy as a function of Cl and pH. Error bars indicate the mean ± s.e.m. of three independent measurements.
[0040] Figure 22 illustrates stability of ChloropHore in HDF cells (a) Representative images of HDF cells at indicated time points. Plots showing the (b) mean R/O ratio, (c) Alexa 647 labelled ChloropHore in the (R) channel and (d) TMR-Dex (O) channel whole cell intensity. Experiments were performed in triplicate. Error bars indicate the mean of cell intensity ± s.e.m. (n = 60 cells). Scale bar = 10 pm (e) Plot showing R/G ratios indicating increase of lumenal [Cl-] (dark gray trace) and D/A ratios indicating increase of lumenal acidity (light gray trace) as a function of chase time. Error bars indicate the mean of three independent experiments ± s.e.m. (n = 25 cells, 250 endosomes) [0041] Figure 23 illustrates intracellular calibration profile of ChloropHore. (a) Fluorescence images of primary human skin fibroblasts labeled with ChloropHore and clamped at the indicated pH and [Cl-] = 100 mM, imaged in the donor channel and the corresponding pseudocolour pH map (b) Histograms of D/A ratios of endosomes at each pH (c)
Fluorescence images of ChloropHore labeled fibroblasts clamped at the indicated [Cl-], pH 5 shown in the Alexa 647 channel and respective pseudocolour [Cl-] map. Scale bar = 10 pm and inset scale bar = 5 pm (d) Histograms of R/G ratios of endosomes at each [Cl-].
Experiments were performed in triplicate (n = 15 cells, 150 endosomes).
[0042] Figure 24 illustrates intracellular calibration profile of ChloropHore. (a) D/A and (b) R/G ratio of ChloropHore as a function of pH and Cl- in endosomes of human dermal fibroblasts each showing the intracellular calibration profile in the specified pH/CI- clamped condition (color dots). Error bars indicate the mean of three independent experiments ± s.e.m. (n = 15 cells, 150 endosomes).
[0043] Figure 25 illustrates performance of /mLYin fibroblast (a) Schematic of the working principle of lmLY at low and high pH. pH indicator - Oregon green (OG) was conjugated to DNA and hybridize with its Alexa 647 labeled complementary strand (b) In vitro (dark gray) and in cellulo (light gray) calibration curve of lmLY. Histograms showing spread of G/R ratios of lysosomes of cells from a normal individual (c) without or (d) 500 nM bafilomycin A1. (e) Bar graphs showing the mean lysosomal pH (G/R) ratios obtained from lysosomes of normal individual cells treated with or without 500 nM bafilomycin. Experiments were performed in triplicate. Error bars indicate the mean of three independent experiments ± s.e.m. (n = 15 cells, 150 lysosomes)
[0044] Figure 26 illustrates quantitative performance of lmLY within single lysosome. (a) Respective pseudocolour G/R map of normal individual skin fibroblast labeled with lmLY and clamped at the indicated pH and [Cl-] = 100 mM. Scale bar = 10 pm. (b) G/R change of lysosomes of cells that clamped with pH 7.5 and pH 4.5. Experiments were performed in triplicate (n = 15 lysosomes).
[0045] Figure 27 illustrates labeling lysosomes of BHK-21 cells (a-b) ChloropHore internalization by BHK-21 cells is competed out by excess maleylated BSA (mBSA, 10 pM), revealing uptake is by scavenger receptors. Cells are imaged in Alexa647 channel (c)
Representative images of lysosomes of HDF cells labelled with TMR dextran (TMR; G) by fluid phased endocytosis and by scavenger receptor mediated endocytosis of ChloropHore (Alexa647). (d) Pearson’s correlation coefficient of colocalization between ChloropHore and lysosomes as a function of ChloropHore labeling chase times. Scale bar = 10 pm. Experiments were performed in triplicate. Error bars indicate the mean of four independent experiments ± s.e.m. (n = 20 cells).
[0046] Figure 28 illustrates labeling lysosome of T47D cells (a-b) ChloropHore
internalization by T47D cells is competed out by excess maleylated BSA (mBSA, 10 mM), revealing uptake is by scavenger receptors. Cells are imaged in Alexa647 channel (c)
Representative images of lysosomes of T47D cells labelled with TMR dextran (TMR; G) by fluid phased endocytosis and by scavenger receptor mediated endocytosis of ChloropHore (Alexa647) and the corresponding Pearson’s correlation coefficient for the colocalization is shown in (d). Scale bar = 10 pm. Experiments were performed in triplicate. Error bars indicate the mean of three independent experiments ± s.e.m. (n = 20 cells).
[0047] Figure 29 illustrates 2-IM profiles of lysosomes in various cell types. 2-IM profiles of lysosomes in (a) human skin fibroblasts, (b) Baby hamster kidney fibroblasts, BHK-21 , (c) murine macrophages J774A.1 and (d) T47D human breast cancer cell lines. Experiments were performed in duplicate (n = 55 cells, = 550 lysosomes)
[0048] Figure 30 illustrates lysosome labeling of endocytosed ChloropHore in fibroblasts. Representative images of lysosomes of (a) normal individual skin fibroblasts, (c) amitriptyline (AH) induced NP-A/B model, (e) U18666A induced NP-C model, (g) NP-A patient derived skin fibroblasts, (i) NP-B patient derived skin fibroblasts and (k) NP-C patient derived skin fibroblasts labelled with TMR dextran (TMR-Dex) and ChloropHore (Alexa647) upon 1 h pulse and 9 h chase of ChloropHore. Pearson's correlation coefficient (PCC) for the colocalization between ChloropHore and lysosome marker - TMR Dextran in (b) normal individual skin fibroblasts, (d) amitriptyline (AH) induced NP-A/B model, (f) U18666A induced NP-C model, (h) NP-A patient derived skin fibroblasts, (j) NP-B patient derived skin fibroblasts and (I) NP-C patient derived skin fibroblasts at indicated chasing times. Scale bar = 10 pm and inset scale bar = 5 pm. Experiments were performed in triplicate. Error bars indicate the mean of three independent experiments ± s.e.m. (n = 25 cells).
DETAILED DESCRIPTION
[0049] Before the disclosed methods and materials are described, it is to be understood that the aspects described herein are not limited to specific embodiments, and as such can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only and, unless specifically defined herein, is not intended to be limiting. In view of the present disclosure, the methods described herein can be configured by the person of ordinary skill in the art to meet the desired need. [0050] Small molecules as well as genetically encodable Ca2+ indicators have profoundly impacted biology. However, their pH sensitivity has restricted their use to the cytoplasm or the endoplasmic reticulum, where the pH is neutral and fairly constant. Ca2+ mapping of acidic microenvironments has therefore not been previously possible. In general, the disclosed methods provide improvements in measurement of pH and Ca2+ concentration.
The inventors have found that a combination reporter for pH and Ca2+ can map both ions in parallel in the same endosome with single endosome addressability, achieving highly accurate measures of lumenal Ca2+. For example, using the pH reporter module of the combination reporter, the pH is determined in individual endosomes. The affinity of the Ca2+ sensitive module, for example, dissociation constant (Kd) and changes with pH, a Kd correction factor suited to the lumenal pH of each endosome may be applied to compute the true value of lumenal Ca2+ with single-endosome resolution. For example, the Kd was computed at every pixel in the pH map to generate a Kd map. From the Kd map and the O/R map, the true Ca2+ map of the acidic organelle can be constructed.
[0051] DNA nanodevices are versatile chemical reporters that can quantitatively map second messengers in real time, in living systems. The modularity of DNA allows integration of distinct functions in precise stoichiometries into a single assembly. These include, for example, (i) a module to fluorescently sense a given ion (ii) a normalizing module for ratiometric quantitation, and/or (iii) a targeting module to localize the reporter in a specific organelle. In certain embodiments, a nucleic acid complex according to one embodiment of the disclosure, CalipHluor, is used in the methods of the disclosure to map organellar pH and Ca2+ simultaneously and with single organelle addressability. For example, by targeting CalipHluor to the scavenger receptor-mediated endocytic pathway, lumenal Ca2+ was mapped as a function of endosomal maturation in nematodes. Ca2+ is fairly low in early and late endosomes, followed by a about 35 fold surge in lumenal Ca2+ in lysosomes - implicating the existence of lysosome-specific Ca2+ import mechanisms. The P5 Ca2+ATPase ATP13A2 was identified as a potential candidate given its similarity to a well-known Ca2+ importer in the endoplasmic reticulum. ATP13A2 (a risk gene for Parkinson’s disease) transports divalent ions such as Mg2+, Mn2+, Cd2+, Zn2+ yet, has not been tested for its ability to transport Ca2+. The C. elegans homolog of ATP13A2, catp-6, was shown to function in opposition to the well-known lysosomal Ca2+ release channel, cup-5. It reversed cup-5 phenotypes at three different levels - a whole organism phenotype, a sub-cellular phenotype and an intra-lysosomal phenotype. The human homolog, ATP13A2 was also shown herein to facilitate lysosomal Ca2+ entry by measuring lysosomal Ca2+ in fibroblasts derived from patients with Kufor Rakeb Syndrome. This constitutes the first example of a lysosomal Ca2+ importer in the animal kingdom. [0052] The ability to map pH and Ca2+ or with single organelle addressability is important to discriminate between lysosomal hypo-acidification and Ca2+ dysregulation. The nucleic acid complexes of the disclosure, in certain embodiments, can be used to map lumenal Ca2+ changes in diverse organelles. As a result, the nucleic acid complexes of the disclosure, for example, can provide new insights into organellar Ca2+ regulation.
[0053] The inventors have also shown that by measuring the pH and Cl- - simultaneously in the same lysosome (referred herein as“2-IM”) and retaining this information with single lysosome addressability, one can resolve lysosomal sub-populations quantitatively in live cells. Methods such as 2-IM have proved elusive to realize thus far for several reasons. CP- sensitive small molecule probes offer the necessary chemical selectivity, molar brightness and long wavelength excitation, but not the required spatial addressability or organelle targetability. Genetically encoded CP sensors offer stable spatial localization, but the response of these reporters to CP is pH sensitive. This complicates analysis of most organelles as lumenal CP entry is coupled to their acidification. Fluorescent proteins label organelles with lower specificity than endocytic tracers and have lower dynamic range compared to DNA-based nanodevices. DNA nanodevices comprise a range of biologically interfacable, quantitative imaging probes that unite the photophysical advantages of small molecules, the stable localization provided by proteins along with the precision of organelle targeting that is accessible to endocytic tracers. Using nucleic acid complexes of the disclosure that can ratiometrically image pH and (CP) simultaneously with single lysosome addressability, lysosome chemotypes on a two-dimensional map that correlates lumenal pH with lumenal (CP) can be discriminated. Lysosome profiles of cells obtained from healthy individuals revealed a high chloride, high acidity population that was absent in cells derived from patients afflicted with Niemann Pick A, B or C diseases. Interestingly, treating these cultured patient cells with the known therapeutic for these diseases led to a reemergence of the high chloride high acidity population.
[0054] In certain embodiments, nucleic acid complexes of the disclosure as describe herein comprises the 1 :1 stoichiometry of DNA hybridization to integrate four functions with stoichiometric precision onto a single probe: (i) a pH sensing function (ii) a CP sensing function (iii) an internal standard for simultaneous ratiometric quantitation of both CP & pH and (iv) a lysosome targeting function for addressability. For example, 2-IM is a highly sensitive method that chemically resolved a high-chloride, high acidity lysosome population in human fibroblasts isolated from skin biopsies of normal, healthy humans. The significance of this high-chloride, high-acidity population was revealed upon 2-IM investigation of fibroblasts derived from skin biopsies of patients afflicted with three variants of Niemann Pick disease, where this population was lost, resulting in highly monodisperse 2-IM profiles. Replenishing cells with the relevant therapeutic, i.e., the defective enzyme, recovered the high-chloride, high-acidity population. Treatment with a molecule documented to have a limited therapeutic efficacy showed marginal recovery of this population. In certain embodiments, 2-IM profiling of lysosomes can be used to screen for potential lead compounds for Niemann Pick diseases, for example to potentially identify suitable patient cohorts for clinical trials in an unbiased way, monitor therapeutic efficacy, or track disease progression.
Nucleic Acid Complexes of the Disclosure
[0055] As provided above, one aspect of the disclosure includes nucleic acid complexes. The nucleic acid complexes of the disclosure as described herein include a Ca2+ fluorophore or a Cl- fluorophore crosslinked to the first single-stranded nucleic acid molecule.
[0056] In certain embodiments, the nucleic acid complexes of the disclosure as describe herein include a Ca2+ fluorophore crosslinked to the first single-stranded nucleic acid molecule. Such Ca2+ fluorophores may be single wavelength indicators or ratiometric indicators. In one embodiment, the Ca2+ fluorophore is a single wavelength indicator.
Numerous Ca2+ fluorophore known in the art may be used in the complexes and methods of the disclosure. Some examples include, but are not limited to, Rhod-5F, XRhod-5F, Rhod- FF, XRhod-FF, Oregon Green 488 BAPTA-6F, Fluo 5F, Fluo 4FF, Oregon Green BAPTA- 5N, Fluo-5 N, and Mag-Fluo-4 indicator. Additional Ca2+ fluorophores may be selected from the labels disclosed herein. In certain embodiments of the disclosure, the Ca2+ fluorophore includes Rhod-5F, XRhod-5F, or Rhod-FF indicator. In certain embodiments of the disclosure, the Ca2+ fluorophore Rhod-5F indicator. Rhod-5F molecule has the following formula:
Figure imgf000017_0001
wherein any available position may be functionalized as to allow for crosslinking to the first strand. For example, one of skill in the art recognizes that a hydrogen atom on the Rhod-5F molecule provided above may be replaced by a functional group that is configured to crosslink to the first strand. [0057] The Ca2+ fluorophore as described herein can be crosslinked to the first single- stranded nucleic acid molecule using linkers and methods known in the art. For example, the Ca2+ fluorophore can be crosslinked using peptide chemistry, click chemistry, by forming ester, ether, thioether, disulfide, amine reactive N-Hydroxysuccinimidyl (NHS) esters, isocyanates, and isothiocyanates bonds, etc. In general, the Ca2+ fluorophore is crosslinked to the first strand through a linker moiety stable under physiological conditions.
[0058] In certain embodiments, the present inventors have determined that the Ca2+ fluorophore can be crosslinked to the first single-stranded nucleic acid molecule using click chemistry. Thus, in certain embodiments, the Ca2+ fluorophore is crosslinked to the first strand through a triazole, thioether, or alkenyl sulfide group. For example, the triazole, thioether, or alkenyl sulfide group can be formed from an azide or thiol moiety on the Ca2+ fluorophore and a alkyne or alkene moiety on the first strand. In another example, the triazole, thioether, or alkenyl sulfide group can be formed from an azide or thiol moiety on the first strand and a alkyne or alkene moiety on the Ca2+ fluorophore.
[0059] In certain embodiments, the Ca2+ fluorophore of the disclosure as described herein includes the following formula:
Figure imgf000018_0001
[0060] In certain embodiments of the disclosure, the first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore is of formula:
Figure imgf000019_0001
[0061] In certain embodiments, the nucleic acid complexes of the disclosure as describe herein include a Cl- fluorophore crosslinked to the first single-stranded nucleic acid molecule.
[0062] Numerous Cl- fluorophores known in the art may be used in the complexes and methods of the disclosure. Some examples include, but are not limited to, 6-methoxy-1-(3- sulfonatopropyl) quinolinium (SPQ), 6-methoxy-N-ethylquinolium Cl- (MEQ), and N-(6- methoxyquinolyl)-acetoethyl ester (MQAE). Additional Cl- fluorophores may be selected from the labels disclosed herein. In certain embodiments of the disclosure, the Cl- fluorophore includes 10,10'-bis[3-carboxypropyl]-9,9'-biacridinium dinitrate (BAC) fluorophore. In certain embodiments of the disclosure, the Cl- fluorophore includes a fluorophore derived from lucigenin.
[0063] The oligonucleotides and nucleic acid molecules in the compositions and methods described herein may include one or more labels. Nucleic acid molecules can be labeled by incorporating moieties detectable by one or more means including, but not limited to, spectroscopic, photochemical, biochemical, immunochemical, or chemical assays. The method of linking or conjugating the label to the nucleotide or oligonucleotide depends on the type of label(s) used and the position of the label on the nucleotide or oligonucleotide.
[0064] As used herein,“labels” are chemical or biochemical moieties useful for labeling a nucleic acid.“Labels” include, for example, fluorescent agents, chemiluminescent agents, chromogenic agents, quenching agents, radionucleotides, enzymes, substrates, cofactors, inhibitors, nanoparticles, magnetic particles, and other moieties known in the art. Labels are capable of generating a measurable signal and may be covalently or noncovalently joined to an oligonucleotide or nucleotide. [0065] In some embodiments, the nucleic acid molecules may be labeled with a“fluorescent dye” or a“fluorophore.” Dyes that may be used in the disclosed methods include, but are not limited to, the following dyes sold under the following trade names: 1 ,5 IAEDANS; 1 ,8-ANS; 4-methylumbelliferone; 5-carboxy-2,7-dichlorofluorescein; 5-carboxyfluorescein (5-FAM); 5- carboxytetramethylrhodamine (5-TAMRA); 5-hydroxy tryptamine (HAT); 5-ROX (carboxy-X- rhodamine); 6-carboxyrhodamine 6G; 6-JOE; 7-amino-4-methylcoumarin; 7- aminoactinomycin D (7-AAD); 7-hydroxy-4-methylcoumarin; 9-amino-6-chloro-2- methoxyacridine; ABQ; Acid Fuchsin; ACMA (9-amino-6-chloro-2-methoxyacridine); Acridine Orange; Acridine Red; Acridine Yellow; Acriflavin; Acriflavin Feulgen SITSA; Alexa Fluor 350™; Alexa Fluor 430™; Alexa Fluor 488™; Alexa Fluor 532™; Alexa Fluor 546™; Alexa Fluor 568™; Alexa Fluor 594™; Alexa Fluor 633™; Alexa Fluor 647™; Alexa Fluor 660™; Alexa Fluor 680™; Alizarin Complexon; Alizarin Red; Allophycocyanin (APC); AMC; AMCA- S; AMCA (Aminomethylcoumarin); AMCA-X; Aminoactinomycin D; Aminocoumarin;
Aminomethylcoumarin (AMCA); Anilin Blue; Anthrocyl stearate; APC (Allophycocyanin); APC-Cy7; APTS; Astrazon Brilliant Red 4G; Astrazon Orange R; Astrazon Red 6B; Astrazon Yellow 7 GLL; Atabrine; ATTO-TAG™ CBQCA; ATTO-TAG™ FQ; Auramine;
Aurophosphine G; Aurophosphine; BAO 9 (Bisaminophenyloxadiazole); Berberine Sulphate; Beta Lactamase; BFP blue shifted GFP (Y66H); Blue Fluorescent Protein; BFP/GFP FRET; Bimane; Bisbenzamide; Bisbenzimide (Hoechst); Blancophor FFG; Blancophor SV;
BOBO™-1 ; BOBO™-3; Bodipy 492/515; Bodipy 493/503; Bodipy 500/510; Bodipy 505/515; Bodipy 530/550; Bodipy 542/563; Bodipy 558/568; Bodipy 564/570; Bodipy 576/589; Bodipy 581/591 ; Bodipy 630/650-X; Bodipy 650/665-X; Bodipy 665/676; Bodipy FL; Bodipy FL ATP; Bodipy Fl-Ceramide; Bodipy R6G SE; Bodipy TMR; Bodipy TMR-X conjugate; Bodipy TMR- X, SE; Bodipy TR; Bodipy TR ATP; Bodipy TR-X SE; BO-PRO™-1 ; BO-PRO™-3; Brilliant Sulphoflavin FF; Calcein; Calcein Blue; Calcium Crimson™; Calcium Green; Calcium Orange; Calcofluor White; Cascade Blue™; Cascade Yellow; Catecholamine; CCF2
(GeneBlazer); CFDA; CFP— Cyan Fluorescent Protein; CFP/YFP FRET; Chlorophyll;
Chromomycin A; CL-NERF (Ratio Dye, pH); CMFDA; Coelenterazine f; Coelenterazine fcp; Coelenterazine h; Coelenterazine hep; Coelenterazine ip; Coelenterazine n; Coelenterazine O; Coumarin Phalloidin; C-phycocyanine; CPM Methylcoumarin; CTC; CTC Formazan; Cy2™; Cy3.18; Cy3.5™; Cy3™; Cy5.18; Cy5.5™; Cy5™; Cy7™; Cyan GFP; cyclic AMP Fluorosensor (FiCRhR); Dabcyl; Dansyl; Dansyl Amine; Dansyl Cadaverine; Dansyl Chloride; Dansyl DHPE; Dansyl fluoride; DAPI; Dapoxyl; Dapoxyl 2; Dapoxyl 3; DCFDA; DCFH (Dichlorodihydrofluorescein Diacetate); DDAO; DHR (Dihydorhodamine 123); Di-4- ANEPPS; Di-8-ANEPPS (non-ratio); DiA (4- Di- 16-ASP); Dichlorodihydrofluorescein
Diacetate (DCFH); DiD— Lipophilic Tracer; DiD (DilC18(5)); DIDS; Dihydorhodamine 123 (DHR); Dil (DilC18(3)); Dinitrophenol; DiO (DiOC18(3)); DiR; DiR (DilC18(7)); DNP; Dopamine; DsRed; DTAF; DY-630-NHS; DY-635-NHS; EBFP; ECFP; EGFP; ELF 97; Eosin; Erythrosin; Erythrosin ITC ; Ethidium Bromide; Ethidium homodimer-1 (EthD-1); Euchrysin; EukoLight; Europium (III) chloride; EYFP; Fast Blue; FDA; Feulgen (Pararosaniline); Flazo Orange; Fluo-3; Fluo-4; Fluorescein (FITC); Fluorescein Diacetate; Fluoro-Emerald; Fluoro- Gold (Hydroxystilbamidine); Fluor-Ruby; FluorX; FM 1-43™; FM 4-46; Fura Red™; Fura RedTm/Fluo-3; Fura-2; Fura-2/BCECF; Genacryl Brilliant Red B; Genacryl Brilliant Yellow 10GF; Genacryl Pink 3G; Genacryl Yellow 5GF; GeneBlazer (CCF2); GFP (S65T); GFP red shifted (rsGFP); GFP wild type, non-UV excitation (wtGFP); GFP wild type, UV excitation (wtGFP); GFPuv; Gloxalic Acid; Granular Blue; Haematoporphyrin; Hoechst 33258; Hoechst 33342; Hoechst 34580; HPTS; Hydroxycoumarin; Hydroxystilbamidine (FluoroGold);
Hydroxytryptamine; lndo-1 ; Indodicarbocyanine (DiD); Indotricarbocyanine (DiR); Intrawhite Cf; JC-1 ; JO-JO-1 ; JO-PRO-1 ; Laurodan; LDS 751 (DNA); LDS 751 (RNA); Leucophor PAF; Leucophor SF; Leucophor WS; Lissamine Rhodamine; Lissamine Rhodamine B;
Calcein/Ethidium homodimer; LOLO-1 ; LO-PRO-1 ; Lucifer Yellow; Lyso Tracker Blue; Lyso Tracker Blue-White; Lyso Tracker Green; Lyso Tracker Red; Lyso Tracker Yellow;
LysoSensor Blue; LysoSensor Green; LysoSensor Yellow/Blue; Mag Green; Magdala Red (Phloxin B); Mag-Fura Red; Mag-Fura-2; Mag-Fura-5; Mag-lndo-1 ; Magnesium Green;
Magnesium Orange; Malachite Green; Marina Blue; Maxilon Brilliant Flavin 10 GFF; Maxilon Brilliant Flavin 8 GFF; Merocyanin; Methoxycoumarin; Mitotracker Green FM; Mitotracker Orange; Mitotracker Red; Mitramycin; Monobromobimane; Monobromobimane (mBBr-GSH); Monochlorobimane; MPS (Methyl Green Pyronine Stilbene); NBD; NBD Amine; Nile Red; NED™; Nitrobenzoxadidole; Noradrenaline; Nuclear Fast Red; Nuclear Yellow; Nylosan Brilliant lavin E8G; Oregon Green; Oregon Green 488-X; Oregon Green™; Oregon Green™ 488; Oregon Green™ 500; Oregon Green™ 514; Pacific Blue; Pararosaniline (Feulgen); PBFI; PE-Cy5; PE-Cy7; PerCP; PerCP-Cy5.5; PE-TexasRed [Red 613]; Phloxin B (Magdala Red); Phorwite AR; Phorwite BKL; Phorwite Rev; Phorwite RPA; Phosphine 3R;
Phycoerythrin B [PE]; Phycoerythrin R [PE]; PKH26 (Sigma); PKH67; PMIA; Pontochrome Blue Black; POPO-1 ; POPO-3; PO-PRO-1 ; PO-PRO-3; Primuline; Procion Yellow;
Propidium lodid (PI); PYMPO; Pyrene; Pyronine; Pyronine B; Pyrozal Brilliant Flavin 7GF; QSY 7; Quinacrine Mustard; Red 613 [PE-TexasRed]; Resorufin; RH 414; Rhod-2;
Rhodamine; Rhodamine 110; Rhodamine 123; Rhodamine 5 GLD; Rhodamine 6G;
Rhodamine B; Rhodamine B 200; Rhodamine B extra; Rhodamine BB; Rhodamine BG; Rhodamine Green; Rhodamine Phallicidine; Rhodamine Phalloidine; Rhodamine Red;
Rhodamine WT; Rose Bengal; R-phycocyanine; R-phycoerythrin (PE); RsGFP; S65A; S65C; S65L; S65T; Sapphire GFP; SBFI; Serotonin; Sevron Brilliant Red 2B; Sevron Brilliant Red 4G; Sevron Brilliant Red B; Sevron Orange; Sevron Yellow L; sgBFP™; sgBFP™ (super glow BFP); sgGFP™; sgGFP™ (super glow GFP); SITS; SITS (Primuline); SITS (Stilbene Isothiosulphonic Acid); SNAFL calcein; SNAFL-1 ; SNAFL-2; SNARF calcein; SNARF1 ; Sodium Green; Spectru Aqua; SpectrumGreen; Spectru Orange; Spectrum Red; SPQ (6- methoxy-N-(3-sulfopropyl)quinolinium); Stilbene; Sulphorhodamine B can C;
Sulphorhodamine G Extra; SYTO 11 ; SYTO 12; SYTO 13; SYTO 14; SYTO 15; SYTO 16; SYTO 17; SYTO 18; SYTO 20; SYTO 21 ; SYTO 22; SYTO 23; SYTO 24; SYTO 25; SYTO 40; SYTO 41 ; SYTO 42; SYTO 43; SYTO 44; SYTO 45; SYTO 59; SYTO 60; SYTO 61 ; SYTO 62; SYTO 63; SYTO 64; SYTO 80; SYTO 81 ; SYTO 82; SYTO 83; SYTO 84; SYTO 85; SYTOX Blue; SYTOX Green; SYTOX Orange; TET™; Tetracycline;
Tetramethylrhodamine (TRITC); Texas Red™; Texas Red-X™ conjugate;
Thiadicarbocyanine (DiSC3); Thiazine Red R; Thiazole Orange; Thioflavin 5; Thioflavin S; Thioflavin TON; Thiolyte; Thiozole Orange; Tinopol CBS (Calcofluor White); TMR; TO-PRO- 1 ; TO-PRO-3; TO-PRO-5; TOTO-1 ; TOTO-3; Tricolor (PE-Cy5); TRITC
TetramethyIRodaminelsoThioCyanate; True Blue; TruRed; Ultralite; Uranine B; Uvitex SFC; VIC®; wt GFP; WW 781 ; X-Rhodamine; XRITC; Xylene Orange; Y66F; Y66H; Y66W; Yellow GFP; YFP; YO-PRO-1 ; YO-PRO-3; YOYO-1 ; YOYO-3; pHrodo™ (available from Thermo Fischer Scientific, Inc. Waltham, MA), and salts thereof.
[0066] Fluorescent dyes or fluorophores may include derivatives that have been modified to facilitate conjugation to another reactive molecule. As such, fluorescent dyes or fluorophores may include amine-reactive derivatives such as isothiocyanate derivatives and/or succinimidyl ester derivatives of the fluorophore.
[0067] The nucleic acid molecules of the disclosed compositions and methods may be labeled with a quencher. Quenching may include dynamic quenching (e.g., by FRET), static quenching, or both. Illustrative quenchers may include Dabcyl. Illustrative quenchers may also include dark quenchers, which may include black hole quenchers sold under the tradename“BHQ” (e.g., BHQ-0, BHQ-1 , BHQ-2, and BHQ-3, Biosearch Technologies, Novato, Calif.). Dark quenchers also may include quenchers sold under the tradename “QXL™” (Anaspec, San Jose, Calif.). Dark quenchers also may include DNP-type non- fluorophores that include a 2,4-dinitrophenyl group.
[0068] In some embodiments, it may be useful to include interactive labels on two or more oligonucleotides with due consideration given for maintaining an appropriate spacing of the labels on the nucleic acid molecules to permit the separation of the labels during a conformational change in the nucleic acid complex. One type of interactive label pair is a quencher-dye pair, which may include a fluorophore and a quencher. The ordinarily skilled artisan can select a suitable quencher moiety that will quench the emission of the particular fluorophore. In an illustrative embodiment, the Dabcyl quencher absorbs the emission of fluorescence from the fluorophore moiety. [0069] In some embodiments, the proximity of the two labels can be detected using fluorescence resonance energy transfer (FRET) or fluorescence polarization. FRET is a distance-dependent interaction between the electronic excited states of two dye molecules in which excitation is transferred from a donor molecule to an acceptor molecule without emission of a photon. Examples of donor/acceptor dye pairs for FRET are known in the art and may include fluorophores and quenchers described herein such as
Fluorescein/Tetramethyl-rhodamine, lAEDANS/Fluorescein (Molecular Probes, Eugene, Oreg.), EDANS/Dabcyl, Fluorescein/Fluorescein (Molecular Probes, Eugene, Oreg.), BODIPY FUBODIPY FL (Molecular Probes, Eugene, Oreg.), BODIPY TMR/ALEXA 647, ALEXA-488/ALEXA-647, and Fluorescein/QSY7™.
[0070] The labels can be conjugated to the nucleic acid molecules directly or indirectly by a variety of techniques. Depending upon the precise type of label used, the label can be located at the 5' or 3' end of the oligonucleotide, located internally in the oligonucleotide's nucleotide sequence, or attached to spacer arms extending from the oligonucleotide and having various sizes and compositions to facilitate signal interactions. Using commercially available phosphoramidite reagents, one can produce nucleic acid molecules containing functional groups (e.g., thiols or primary amines) at either terminus, for example by the coupling of a phosphoramidite dye to the 5' hydroxyl of the 5' base by the formation of a phosphate bond, or internally, via an appropriately protected phosphoramidite.
[0071] Nucleic acid molecules may also incorporate functionalizing reagents having one or more sulfhydryl, amino or hydroxyl moieties into the nucleic acid sequence. For example, a 5' phosphate group can be incorporated as a radioisotope by using polynucleotide kinase and [y32P] ATP to provide a reporter group. Biotin can be added to the 5' end by reacting an aminothymidine residue, introduced during synthesis, with an N-hydroxysuccinimide ester of biotin. Labels at the 3' terminus, for example, can employ polynucleotide terminal transferase to add the desired moiety, such as for example, cordycepin, 35S-dATP, and biotinylated dUTP.
[0072] Oligonucleotide derivatives are also available as labels. For example, etheno-dA and etheno-A are known fluorescent adenine nucleotides which can be incorporated into a reporter. Similarly, etheno-dC is another analog that can be used in reporter synthesis. The reporters containing such nucleotide derivatives can be hydrolyzed to release much more strongly fluorescent mononucleotides by the polymerase's 5' to 3' nuclease activity as nucleic acid polymerase extends a primer during PCR.
[0073] In some embodiments, a first label is conjugated to the second single-stranded nucleic acid molecule. [0074] In certain embodiments of the disclosure, a second label is conjugated to the first single-stranded nucleic acid molecule or the second single-stranded nucleic acid molecule.
In some embodiments, a first label is conjugated to the second single-stranded nucleic acid molecule and the second label is conjugated to the first single-stranded nucleic acid molecule, wherein the first label is capable of producing a signal, and wherein the intensity of the signal varies as a function of the conformation of the nucleic acid complex.
[0075] In certain embodiments of the disclosure, a second label is conjugated to the third single-stranded nucleic acid. In some embodiments, the first label is conjugated to the second single-stranded nucleic acid molecule and the second label is conjugated to the third single-stranded nucleic acid, wherein the first label is capable of producing a signal, and wherein the intensity of the signal varies as a function of the conformation of the nucleic acid complex.
[0076] In certain embodiments, the intensity of the signal is irrelevant of the distance between the first and second labels and/or the relative orientation of the first and second labels.
[0077] In certain embodiments, the intensity of the signal varies as a function of at least one of the distance between the first and second labels and the relative orientation of the first and second labels.
[0078] In some embodiments, the first and second labels comprise a donor and acceptor pair. In some embodiments, the signal is measured using a FRET technique. For example, the signal can be measured at 2 different wavelengths. In another example, the signal can be measured at 4 different wavelengths. In some embodiments, at least one label is selected from the group consisting of an Atto dye, an Alexa Flour® dye, a Cy® dye, and a BODIPY dye. In some embodiments, the donor and acceptor pair are FITC and TRITC, Cy3 and Cy5, or Alexa-488 and Alexa-647. In some embodiments, the donor and acceptor pair are labels described herein. In some embodiments, the first and second label comprise a donor fluorophore and an acceptor quencher.
[0079] In some embodiments, the signal and label is directionally dependent (anisotropy). Non-limiting examples of such labels include Atto dyes, BODipy dyes, Alexa dyes,
TMR/TAMRA dyes, or Cy dyes.
[0080] A provided above, the nucleic acid complexes of the disclosure include a first single- stranded molecule and a second single-stranded nucleic acid molecule that is partially or fully complementary to the first single-stranded molecule. In certain embodiments, the nucleic acid complexes of the disclosure include a first single-stranded molecule, a second single-stranded nucleic acid molecule that is partially complementary to the first single- stranded molecule, and a third single-stranded nucleic acid molecule that is partially complementary to the first single-stranded molecule.
[0081] In certain embodiments of the methods and nucleic acid molecule and complexes described herein, the second nucleic acid strand and/or the third nucleic acid strand is one that is only partially complementary to the first nucleic acid. A nucleic acid strand is fully complementary when all bases are capable of forming conventional Watson-Crick base pairing (e.g. G-C and A-T base pairing). A nucleic acid strand is partially complementary when at least one of the base pairs is not complementary to the opposing strand. In some embodiments, the second single nucleic acid strand comprises at least 4 non
complementary nucleic acid bases. In some embodiments, the second single nucleic acid strand comprises at least, at most, or exactly 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 (or any derivable range therein) non-complementary nucleic acid bases. In some
embodiments, the second nucleic acid strand comprises 8 non-complementary nucleic acid bases.
[0082] In certain embodiments of the nucleic acid complexes of the disclosure, each of the first single-stranded nucleic acid molecule the second single-stranded nucleic acid molecule, and/or the third single-stranded nucleic acid molecule is independently less than 200 nucleotides. In some embodiments, each of the first single-stranded nucleic acid molecule the second single-stranded nucleic acid molecule, and/or the third single-stranded nucleic acid molecule is independently less than, at least, or exactly 20, 30, 40, 60, 80, 100, 125, 150, 175, 200, 225, 250, 275, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 900, or 1000 nucleotides in length, or any derivable range therein.
[0083] The nucleic acid complexes described herein are useful as Ca2+ concentration sensors, and have high sensitivity without a substantial change in cooperativity. In certain embodiments, the nucleic acid complexes described herein are capable of determining the Ca2+ concentration in a range of 10 nM to 10 mM, the range is inclusive of the recited Ca2+ concentration. For example, in certain embodiments, the nucleic acid complexes described herein are capable of determining the Ca2+ concentration in a range of 10 nM to 1 mM, or 10 nM to 100 nM, or 10 nM to 500 nM, or 100 nM to 500 nM, or 100 nM to 1 pM, or 500 nM to 1 pM, or 500 nM to 50 pM, or 1 pM to 1 mM, or 1 pM to 10 mM, or 10 pM to 10 mM, or 100 pM to 1 mM, or 100 pM to 10 mM. In other embodiments, the recited Ca2+ concentration is excluded.
[0084] The nucleic acid complexes described herein are useful as CF concentration sensors, and have high sensitivity without a substantial change in cooperativity. In certain embodiments, the nucleic acid complexes described herein are capable of determining the Cl- concentration in a range of 1 mM to 100 mM, the range is inclusive of the recited CF concentration. For example, in certain embodiments, the nucleic acid complexes described herein are capable of determining the CF concentration in a range of 1 mM to 50 mM, 1 mM to 25 mM, or 1 mM to 10 mM, or 5 mM to 100 mM, or 5 mM to 50 mM, 5 mM to 25 mM, or 5 mM to 10 mM, or 10 mM to 25 mM, or 10 mM to 50 mM, or 10 mM to 75 mM, or 20 mM to 80 mM, or 30 mM to 70 mM. In other embodiments, the recited CFconcentration is excluded.
[0085] The nucleic acid complexes described herein are also useful as pH sensors, and have high sensitivity without a substantial change in cooperativity. In certain embodiments, the nucleic acid complexes described herein are capable of determining the pH of is less than 5.5 or more than 7.0. In certain embodiments, the nucleic acid complexes described herein are capable of determining the pH of less than or exactly pH 5.8, 5.7, 5.6, 5.5, 5.4,
5.3, 5.2, 5.0, 4.8, 4.6, 4.4, 4.2, or 4.0 (or any derivable range therein). In certain
embodiments, the nucleic acid complexes described herein are capable of determining the pH of more than or exactly pH 6.8, 6.9, 7.0, 7.1 , 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 8.0, 8.2, 8.4, 8.6, 8.8 or 9.0 (or any derivable range therein). In certain embodiments, the nucleic acid complexes described herein are capable of determining the pH in the range of 5.5 to 7. In certain embodiments, the nucleic acid complexes described herein are capable of determining the pH in the range of 5.8 and 6.8, or 5.8 to 7, or 5.6 to 6.8, or 5.4 to 6.8.
[0086] Cytosine rich DNA sequences are found in human genomes such as in telomeres and in promoters of several oncogenes, e.g., c-myc. In certain embodiments, the nucleic acid complexes of the disclosure can form a special tetraplex structure under slightly acidic condition where two parallel duplexes paired through OCH+ pairs intercalated with each other in head to tail orientation called the i-motif. The“i-motif” is a nucleic acid (DNA and/or RNA) containing complex characterized by the presence of cytosine-rich stretches or stretches rich in cytosine derivatives, including two parallel-stranded duplexes in which the cytosines or derivatives thereof form base pairs, and the two duplexes are associated anti parallel to one another. The pairs of cytosine or derivatives thereof of one duplex are intercalated with those of the other duplex.
[0087] The structure of an i-motif differs from that of the usual DNA duplex because the base pairing scheme involves hemiprotonated cytosines which result in the formation of OC+ base pairs. Specifically, one of the cytosines contained in each pair is protonated. The i-motif may also exist as a tetramer formed by the association of two duplexes as described above.
[0088] In certain embodiments, the nucleic acid complexes of the disclosure may be synthesized from oligonucleotide sequences including a stretch of at least two, at least three, or at least four consecutive cytosines. By modifying the number of cytosines, as well as the degree of complementarity between both strands, it is possible to modulate the response time of the nucleic acid complexes of the disclosure and to the pH sensing range. When more cytosines contribute to the i-motif, the stability of the motif is increased. Moreover, this motif may be formed by the interaction of stretches containing different numbers of cytosines. Furthermore, a cytosine-rich stretch may contain one or two non-cytosine base(s) in between the cytosines. However, this may reduce the stability of the i-motif. The cytosine stretches which comprise the i-motif may belong to different strands of nucleic acids;
however, any two of them may also be linked together covalently or non-covalently. Also, any two of them may be part of a single nucleic acid strand wherein they are separated by a stretch of specified bases.
[0089] In certain embodiments, the second single-stranded nucleic acid molecule of the nucleic acid complexes of the disclosure comprises the sequence CnXCnYCnZCn (SEQ ID NO. 13), wherein C is cytosine; X, Y and Z are each one or more adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues of the nucleic acid molecule are modified. In some embodiments, at least or exactly 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, or 18 (or any derivable range therein) cytosine residues of the second single-stranded nucleic acid molecule are modified. In some embodiments, the number of cytosine residues of the second single-stranded nucleic acid molecule that are modified is in the range of 2 to 18, or 4 to 18, or 10 to 18, or 14 to 18, or 2 to 14, or 4 to 14, or 10 to 14, or 2 to 10, or 4 to 10, or 8 to 16.
[0090] In some embodiments, each of X, Y, and Z is independently AA or TAA. In certain embodiments, each of X, Y, and Z is independently TAA. In some embodiments, n is 3, 4, or 7. In further embodiments, n is at least, at most, or exactly 3, 4, 5, 6, 7, 8, or 9 (or any derivable range therein). In some embodiments, n is 4. In some embodiments, the modification is a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group. In some embodiments, the cytosine is modified with a methyl or bromo group. In some
embodiments, the modification is at the 5’ position of the cytosine. In some embodiments, all the cytosines in the second nucleic acid molecule are modified with the same
modification. In some embodiments, all the cytosines in the second nucleic acid molecule are modified with a negatively charged modification. In some embodiments, all the cytosines in the second nucleic acid molecule are modified with a positively charged modification.
[0091] In some embodiments, the second single-stranded molecule comprises the sequence ((Ca)nX(C )nY(Cc)nZ(Cd)n (SEQ ID NO. 14) wherein Ca, Cb, Cc, and Cd are equal to n number of consecutive cytosine residues; X, Y, and Z are one or more adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 3. In some embodiments, each of Ca, C , Cc, and Cd comprise at least one modified cytosine. In some embodiments, each of Ca, Cb, Cc, and Cd comprise at least, at most, or exactly 1 , 2, 3, 4, or 5 modified cytosines (or any derivable range therein). In some embodiments, the modified cytosine is the first or last consecutive cytosine in each of Ca, C , Cc, and Cd. In some embodiments, n = 3 and the modified cytosine is the second consecutive cytosine in each of Ca, C , Cc, and Cd. In some embodiments, n = 4 and the modified cytosine is the second or third consecutive cytosine in each of Ca, C , Cc, and Cd. In some embodiments, each of C and Cc comprise at least two modified cytosines and each of Ca and Cd consist of unmodified cytosine. In some embodiments, Ca or Cd consists of modified cytosine residues. In some embodiments, Ca, C , Cc, and/or Cd consist of or comprise of at least, at most, or exactly 1 ,
2, 3, 4, 5, 6, or 7 modified cytosine residues, or any derivable range therein.
[0092] In certain embodiments, the second single-stranded nucleic acid molecule of the nucleic acid complexes of the disclosure excludes the sequence CnXCnYCnZCn (SEQ I D NO. 13) or ((Ca)nX(Cb)nY(Cc)nZ(Cd)n (SEQ ID NO. 14) as provided herein.
[0093] In certain embodiments, the second single-stranded nucleic acid molecule and/or the third single-stranded nucleic acid molecule of the nucleic acid complexes of the disclosure comprises the sequence CnXCn, wherein C is cytosine; X and Y are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified. In some embodiments, at least or exactly 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, or 18 (or any derivable range therein) cytosine residues of the second single-stranded nucleic acid molecule are modified. In some embodiments, the number of cytosine residues of the second single-stranded nucleic acid molecule that are modified is in the range of 2 to 18, or 4 to 18, or 10 to 18, or 14 to 18, or 2 to 14, or 4 to 14, or 10 to 14, or 2 to 10, or 4 to 10, or 8 to 16.
[0094] In some embodiments, X and Y are independently AA or TAA. In certain
embodiments, X and Y are independently AA. In some embodiments, n is 3, 4, or 7. In further embodiments, n is at least, at most, or exactly 3, 4, 5, 6, 7, 8, or 9 (or any derivable range therein). In some embodiments, n is 4. In some embodiments, the modification is a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group. In some embodiments, the cytosine is modified with a methyl or bromo group. In some embodiments, the modification is at the 5’ position of the cytosine. In some embodiments, all the cytosines in the second nucleic acid molecule are modified with the same modification. In some embodiments, all the cytosines in the second nucleic acid molecule are modified with a negatively charged modification. In some embodiments, all the cytosines in the second nucleic acid molecule are modified with a positively charged modification. [0095] The nucleic acid complexes described herein are useful as pH sensors, and have high sensitivity (as evidenced by fold change of D/A ratio) without a substantial change in cooperativity. In some embodiments, the method further comprises calculating a D/A ratio from the signal intensity values. In some embodiments, the D/A ratio is a normalized value. In some embodiments, the fold change of the D/A ratio is at least 4.1 , 5, 6, 7, or 7.5. In some embodiments, the fold change of the D/A ratio is between 4.1 and 7.5, between 5 and 7.5, between 6 and 7.5, between 7 and 7.5, between 4.1 and 7, between 5 and 7, between 6 and 7, between 4.1 and 6, between 5 and 6, or between 4.1 and 5. In some embodiments, the fold change of the D/A ratio is at least or exactly 4.5, 5, 6, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11 , 11.5, 12, 12.5, 13, or 14 (or any derivable range therein). In some embodiments, the cooperativity, compared to the unmodified nucleic acid complexes, is changed less than 2 fold, or less than 1.75, 1.5, 1.25. 1 , 0.75. 0.5, 0.25, 0.2, 0.1 , fold or any derivable range therein. In some embodiments, the cooperativity is less than 5, 10, 15, 20, 25, 30, 35, 40,
45, or 50% different than the un-modified nucleic acid complexes. In some embodiments, the fold change of the D/A ratio is at least or exactly 4.5, 5, 6, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11 , 11.5, 12, 12.5, 13, or 14 (or any derivable range therein) and the cooperativity, compared to the unmodified nucleic acid complexes, is changed by less than 1.75, 1.5, 1.25. 1 , 0.75. 0.5, 0.25, 0.2, or 0.1 fold or any derivable range therein. In some embodiments, the pHha|f is altered without substantially increasing the cooperativity. In some embodiments, the pHha|f is at least, at most, or exactly 5.0, 5.2, 5.4, 5.6, 5.8, 6.0, 6.2, 6.4, 6.6, 6.8, 7.0, 7.2, 7.4, 7.6,
7.8, 8.0, 8.2, 8.4, 8.6, 8.8, or 9.0 (or any derivable range therein). In some embodiments, the pH haif, compared to the un-modified nucleic acid complexes, is at least, at most, or exactly 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1 , 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 2.9, or 3.0 pH units different (or any range derivable therein). In some embodiments, the pHhaif, compared to the un-modified nucleic acid complexes, is at least, at most, or exactly 3, 5, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 24,
26, 28, or 30% different (or any derivable range therein). In some embodiments, the pHhaif, compared to the un-modified nucleic acid complexes, is at least, at most, or exactly 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1 , 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 2.9, or 3.0 pH units different or is at least, at most, or exactly 3, 5, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 24, 26, 28, or 30% different (or any derivable range therein) and the cooperativity, compared to the unmodified nucleic acid complexes, is changed by less than 1.75, 1.5, 1.25. 1 , 0.75. 0.5, 0.25, 0.2, or 0.1 fold or any derivable range therein. In some embodiments, the measured value described herein (i.e. , signal intensity, pHha|f, fold change, or cooperativity) is a normalized value. [0096] In some embodiments, the first and second single-stranded nucleic acid molecules are capable of forming an i-motif under acidic conditions. In some embodiments, the first nucleic acid strand is capable of forming an intramolecular complex comprising two parallel- stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
[0097] The nucleic acid molecules and complexes of the disclosure, in some embodiments, comprise a targeting moiety, such as a nucleic acid, small molecule, or polypeptide that has an affinity for a certain target or, by virtue of its chemical makeup, is targeted to a particular location in the cell. The targeting moiety can act as a handle to target the nucleic acid complexes of the disclosure to different subcellular locations. The targeting moiety may be a nucleic acid that binds to a receptor protein, and the receptor protein may be one that is intracellularly targeted or conjugated to a protein that is intracellularly targeted. The targeting moiety or receptor protein may be a targeting nucleic acid or a protein such as a plasma membrane protein that is endocytosable, any proteins that possess a natural receptor, a protein that traffics between intracellular locations via the plasma membrane, toxins, viruses and viral coat proteins, cell penetrating peptides, signal sequences, intracellular targeting sequences, small organic molecules, endocytic ligands and trafficking proteins. In some embodiments, the targeting moiety is an aptamer, a duplex domain targeted to an artificial protein receptor, a nucleic acid sequence that binds an anionic-ligand binding receptor, or an endocytic ligand. The targeting moiety may also be a G4 core sequence or ribozyme.
[0098] In some embodiments, the targeting moiety is a nucleic acid sequence. In some embodiments, the targeting moiety has a cognate artificial protein receptor. The artificial receptor may be, for example, a single chain variable fragment (scFv), transcription factor, Zn-fingered protein, leucine zipper, or DNA binding immunoglobulin, In some embodiments, the targeting moiety is encoded on the same nucleic acid strand as the first and/or second single-stranded nucleic acid molecule. In some embodiments, the targeting moiety is selected from an aptamer, a duplex domain targeted to an artificial protein receptor, a nucleic acid sequence that binds an anionic-ligand binding receptor, and an endocytic ligand. In some embodiments, the targeting moiety comprises a peptide directly or indirectly conjugated to the nucleic acid molecule. In some embodiments, the targeting moiety peptide comprises one or more of a fusogenic peptide, a membrane-permeabilizing peptide, a sub- cellular localization sequence, or a cell-receptor ligand. In some embodiments, the sub- cellular localization sequence targets the nucleic acid complex to a region of the cell where spatial localization of a targeted protein is present. In some embodiments, the sub-cellular localization sequence targets the nucleic acid complex to a region of the cell selected from the group consisting of: the cytosol, the endoplasmic reticulum, the mitochondrial matrix, the chloroplast lumen, the medial trans-Golgi cistemae, the lumen of lysosome, the lumen of an endosome, the peroxisome, the nucleus, and a specific spatial location on the plasma membrane. In some embodiments, the sub-cellular organelle is one that exchanges membrane directly or indirectly with the plasma membrane.
[0099] In some embodiments, the nucleic acid is a peptide nucleic acid (PNA). The strand is conjugated to Ca2+ fluorophore or Cl- fluorophore. In some embodiments, the second label is Cl-- or Ca2+-sensitive fluorophore, for example, a CP-sensitive fluorophore such as BAC conjugated to PNA. In some embodiments, the first strain comprises PNA and CP fluorophore. In some embodiments, the second label is insensitive fluorophore, for example, a chloride ion insensitive fluorophore such as Alexa 647, conjugated to DNA sequence that is complementary to PNA of the sensing module.
[0100] The current methods, nucleic acids, and nucleic acid complexes may be used in combination with additional nucleic acid based sensors, such as those described in
International Patent Publication No, WO 2015/159122, which is herein incorporated by reference.
[0101] In the present disclosure, PNA or PNA strand or PNA sequence is used
interchangeably and has the same scope or meaning. In the present disclosure, DNA or DNA strand or DNA sequence is used interchangeably and has the same scope or meaning. In the present disclosure, RNA or RNA strand or RNA sequence is used interchangeably and has the same scope or meaning.
[0102] In some embodiments, the nucleic acid complex of the present disclosure self assembles two or all three strands through Watson-Crick base pairing, which is stable under physiological conditions.
[0103] In embodiments of the present disclosure, two types of targeting moiety are used: A) DNA only and B) a combination of DNA and RNA. The targeting moiety comprising only DNA hybridizes to normalizing module to form the dsDNA domain required for intracellular targeting via an anionic ligand binding receptor (ALBR). The RNA sequence used in combination with DNA in the targeting moiety is used to achieve targeting to Transferrin pathway.
[0104] In some embodiments, a DNA strand is used as first strand and/or the second strand. In an embodiment of the present disclosure, the nucleic acid complex has a dsDNA part (minimum 15 bp sequence) resulting from the hybridization of the first strand and the second strand, or the first strand and the third strand, or the first strand with the second strand and the third strand. In certain embodiments, the nucleic acid complex comprises d(AT)4 sequence and hence is targeted to any given compartment in any cell that expresses scFv tagged protein of choice.
[0105] In an exemplary, non-limiting embodiment, the nucleic acid complex as described herein comprises the first strain has the sequence 5’-the Ca2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and the second strain has the sequence 5’-the first label-CCC CTA ACC CCT AAC CCC TAA CCC CAT ATA TAT CCT AGA ACG ACA GAC AAA CAG TGA GTC-3’(SEQ ID NO:08).
[0106] In an exemplary, non-limiting embodiment, the nucleic acid complex as described herein comprises the first strain has the sequence 5’-TTA TAG GAT CCT GCG GTC GG/the Ca2+ fluorophore/ GGC ACC AGG CGT AAA ATG TA-3’(SEQ ID NO:09); the second strain has the sequence: 5’-the first label-CCC CAA CCC CAA TAC ATT TTA CGC CTG GTG CC- 3’ (SEQ ID NO:10); and the third strain has the sequence: 5’-CCG ACC GCA GGA TCC TAT AAA ACC CCA ACC CC-the second label-3 (SEQ ID NO: 11).
[0107] In an exemplary, non-limiting embodiment, the nucleic acid complex as described herein comprises the first strain has the sequence 5’-the Ca2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and the second strain has the sequence 5'- the first label - AT AAC ACA TAA CAC ATAACAAAA TAT ATA TCC TAG AAC GAC AGA CAA ACA GTG AGT
C - 3' (SEQ ID NO:12).
[0108] In an exemplary, non-limiting embodiment, the nucleic acid complex as described herein comprises the first strain has the sequence 5’-the Cl- fluorophore-ATC AAC ACT GCA-Lys-COOH (SEQ ID NO:22); the second strain has the sequence 5'-TAT TGT GTA TTG TGT ATT GTT TTA TAT AT /the first label/ A TAG GAT CTT GCT GTC TGG TGT GCA GTG TTG AT-3'(SEQ ID NO:23); and the third strain has the sequence: 5’-CAC CAG ACA GCA AGA TCC TAT ATA TAT ACC CCA ATC CCC AAT CCC CAA TCC CC-the second label-3' (SEQ ID NO:24).
Detection of pH and Ca2+ or Cl~ concentration in samples
[0109] The methods described herein may be used to monitor the pH changes and/or Ca2+ or Cl- concentration in real-time during cellular processes. In some embodiments, the methods are for monitoring endocytosis. While not wishing to be limited by theory, acidification plays a major role in facilitating cargo dissociation from receptors or in mediating cellular entry of toxins and viruses during endocytosis. In certain embodiments, the nucleic acid complex exhibits a pH response inside cells illustrated by the capture of spatiotemporal pH changes associated with endocytosis in living cells. [0110] Fluorescence in the sample can be measured in a variety of ways, such as using a fluorometer or fluorescence microscopy. In general, excitation radiation, from an excitation source having a first wavelength, passes through excitation optics. The excitation optics cause the excitation radiation to excite the sample. In response, labels in the sample emit radiation which has a wavelength that is different from the excitation wavelength. The device can include a temperature controller to maintain the sample at a specific temperature while it is being scanned. If desired, a multi-axis translation stage can be used to move a microtiter plate holding a plurality of samples in order to position different wells to be exposed. The multi-axis translation stage, temperature controller, auto-focusing feature, and electronics associated with imaging and data collection can be managed by an appropriately programmed digital computer. The computer also can transform the data collected during the assay into another format for presentation.
[0111] In some embodiments, the detecting includes measuring the magnitude of the signal generated, wherein the magnitude indicates the pH of the cell or region thereof or the Ca2+ or CF concentration in the cell or region thereof. In certain embodiments, the emission from the acceptor fluorophore increases as the nucleic acid complex forms a closed state, i.e. , as the i-motif is formed when the pH decreases. Likewise, the emission from the acceptor fluorophore decreases as the nucleic acid complex assumes an open state, i.e., as the i- motif dissociates when the pH increases. For fluorescence quenching, the emission from the fluorophore decreases as the nucleic acid complex forms a closed state, i.e., as the i-motif is formed when the pH decreases. Likewise, the emission from the fluorophore increases as the nucleic acid complex forms an open state, i.e., as the i-motif dissociates when the pH increases. In certain embodiments, the emission from the fluorophore is independent of the conformation of the nucleic acid complex.
[0112] As used herein, an“increase” (or“decrease”) in a signal from the nucleic acid complex refers to the change in a signal in the sample compared to a reference sample. The reference sample may be a control sample (e.g., an untreated population of cells where the effects of a drug or agent are being examined), or it may be the same sample at a different period of time, for instance, where the intracellular pH and/or Ca2+ or CF concentration is being monitored to follow one or more cellular processes.
[0113] As used herein, the term“detectable” refers to a property of the nucleic acid complex that allows one to determine the pH and/or Ca2+ or CF concentration of a biological sample by detecting activity, e.g., fluorescence activity, possessed by the nucleic acid complex under certain conditions. In some embodiments, the signal from the nucleic acid complex is normalized by plotting the donor/acceptor (D/A) signal ratio as a function of pH in a standard reference sample. pH variation on a doubly-labeled nucleic acid complex changes the ratio between its closed and open states thereby resulting in different ratios of the donor and acceptor intensities (D/A) because of FRET in the closed state due to i-motif formation.
[0114] In one embodiment, a pH calibration curve and/or Ca2+ or CF concentration calibration curve may be generated to which test samples may be compared and normalized. An intracellular calibration curve may be generated according to methods described in U.S. Patent Application Publication No.: 2010/0304370, which is herein incorporated by reference. Briefly, for the pH calibration curve, cells are pulsed, washed, incubated with an ionophore in buffers at a given pH and then mildly fixed. Donor and acceptor FRET images are acquired from which D/A ratios are obtained. The mean D/A of individual cells or regions thereof at each pH are plotted as a function of pH for the intracellular pH calibration curve. The D/A ratio of the test sample can be compared to the calibration curve. Similar approach may be used to generate Ca2+ concentration calibration curve and CF concentration calibration curve. Related methods are also described in the Examples of the application.
[0115] In some embodiments, intracellular pH and/or Ca2+ or CF concentration may be monitored for the purposes of examining cellular phenomena and/or screening the effects of various compounds, wherein the level of the signal from a nucleic acid complex (e.g., increased or decreased signal) in a test sample at a first time point is determined and compared with the level found in a test sample obtained at a later time point. The change in signal may reflect a relative change in pH and/or Ca2+ or CF concentration between the two samples. For example, where a FRET pair is used as a label, an increase in signal from one time point to another may indicate an increase in pH between the two time points. Likewise, a decrease in signal from one point to another may indicate a decrease in pH or decrease in Ca2+ or CF concentration. The absolute level of signal may be compared to a reference sample of known standards or reference samples in order to determine the precise pH and/or Ca2+ or CF concentration of the sample. The sample can be classified or assigned to a particular pH value based on how similar the measured levels were compared to the control levels for a given group.
[0116] As one of skill in the art will understand, there will be a certain degree of uncertainty involved in making this determination. Therefore, the standard deviations of the control group levels can be used to make a probabilistic determination and the method of this disclosure are applicable over a wide range of probability-based determinations. Thus, for example, and not by way of limitation, in one embodiment, if the measured level of signal falls within 2.5 standard deviations of the mean of any of the control groups, then that sample may be assigned to that group. In another embodiment if the measured level of signal falls within 2.0 standard deviations of the mean of any of the control groups then that sample may be assigned to that group. In still another embodiment, if the measured level of signal falls within 1.5 standard deviations of the mean of any of the control groups then that sample may be assigned to that group. In yet another embodiment, if the measured level of signal is 1.0 or less standard deviations of the mean of any of the control groups levels then that sample may be assigned to that group. Thus, this process allows determination, with various degrees of probability, in which group a specific sample should be placed.
[0117] Statistical methods can also be used to set thresholds for determining when the signal intensity in a test sample can be considered to be different than or similar to the reference level. In addition, statistics can be used to determine the validity of the difference or similarity observed between a test sample's signal intensity and the reference level.
Useful statistical analysis methods are described in L. D. Fisher & G. vanBelle, Biostatistics: A Methodology for the Health Sciences (Wiley-lnterscience, NY, 1993). For instance, confidence (“p”) values can be calculated using an unpaired 2-tailed t test, with a difference between groups deemed significant if the p value is less than or equal to 0.05.
[0118] The nucleic acid complexes described herein are useful as pH and Ca2+
concentration sensors and may vary in their respective pKa, and the differences in pKa can be used to select the most suitable nucleic acid complex for a particular application. In general, a sensor should be used whose pKa is close to the pH of the sample to be measured. For example, the pKa may be within 1.5 pH unit, within 1.0 pH unit, or within 0.5 pH units of the sample.
[0119] The nucleic acid complexes described herein are useful as pH and Cl- concentration sensors. In certain embodiments, the signal of the Cl- fluorophore is linearly dependent on CF concentration with constant Stern-Volmer quenching constant (Ksv). In certain embodiments, the signal of the Cl- fluorophore is insensitive to physiological change in pH and to cations, non-halide anions (nitrate, phosphate, bicarbonate, sulfate), and albumin.
[0120] To minimize artefactually low fluorescence measurements that occur due to cell movement or focusing, the fluorescence of the nucleic acid complex can be compared to the fluorescence of a second sensor, e.g., a second nucleic acid complex that is also present in the measured sample. The second nucleic acid complex should have an emission spectra distinct from the first nucleic acid complex so that the emission spectra of the two sensors can be distinguished. Because experimental conditions such as focusing and cell movement will affect fluorescence of the second sensor as well as the first sensor, comparing the relative fluorescence of the two sensors may allow for the normalization of fluorescence. A convenient method of comparing the samples is to compute the ratio of the fluorescence of the first fluorescent protein sensor to that of the second fluorescent protein sensor. [0121] In some embodiments, circular dichroism spectroscopy may be used to detect changes in the secondary structure of the nucleic acid complex in response to changes in pH and/or Ca2+ or Cl- concentration. Circular Dichroism (CD) is observed when optically active matter absorbs left and right hand circular polarized light slightly differently. It is measured with a CD spectropolarimeter. In another embodiment, change in intracellular pH may be detected by observing Raman band changes in the nucleic acid complex. In this embodiment, the nucleic acid complex contains a gold nanoparticle label and a Raman tag. The Raman band changes may be detected when the gold nanoparticle is brought close to a Raman tag.
[0122] In some embodiments, FLIM is used to measure the conformational change upon i- motif formation. In some embodiments, anisotropy imaging is used to measure the conformational change. Fluorescence-lifetime imaging microscopy (FLIM) is an imaging technique for producing an image based on the differences in the exponential decay rate of the fluorescence from a fluorescent sample. It can be used as an imaging technique in confocal microscopy, two-photon excitation microscopy, and multiphoton tomography. The lifetime of the fluorophore signal, rather than its intensity, is used to create the image in FLIM. In some embodiments, FLIM is used to gain pH information, as one of the photo physical properties of the dyes that would change when the nucleic acid complex changes conformation due to a change in the pH. In some embodiments, the dye is an Atto dye, BODIPY dye, Alexa dye, TMR/TAMRA dye, or Cy dye. Anisotropic imaging and FLIM are further described in Ekta Makhija, et al.,“Probing Chromatin Structure and Dynamics Using Fluorescence Anisotropy Imaging” CRC Handbook, Imaging Biological Mechanics (2014) and Levitt et al.,“Fluorescence lifetime and polarization-resolved imaging in cell biology” Current Opinion in Biotechnology 20(1): 28-36 (2009), which are herein incorporated by reference for all purposes.
[0123] In some embodiments, the signal intensity changes by at least twenty percent as the Ca2+ concentration is raised. In some embodiments, the signal intensity changes by at least 10, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, or 90% or any derivable range therein when the Ca2+ concentration is raised.
[0124] In certain embodiments, the methods of the disclosure determine the Ca2+ concentration in a range of 10 nM to 10 mM, the range is inclusive of the recited Ca2+ concentration. For example, a Ca2+ concentration that is determined by the methods of the disclosure concentration is in a range of 10 nM to 1 mM, or 10 nM to 100 nM, or 10 nM to 500 nM, or 100 nM to 500 nM, or 100 nM to 1 pM, or 500 nM to 1 pM, or 500 nM to 50 pM, or 1 pM to 1 mM, or 1 pM to 10 mM, or 10 pM to 10 mM, or 100 pM to 1 mM, or 100 pM to 10 mM. In other embodiments, the recited Ca2+ concentration is excluded. [0125] In some embodiments, the Ca2+ concentration is determined by comparing the measured signal to a reference value. In some embodiments, the Ca2+ concentration is determined by comparing the measured signal to a reference value. In some embodiments, the signal value and/or reference value is normalized. In some embodiments, the method further comprises creating a standard curve. A standard curve can be created by measuring the signal intensity at different known Ca2+ concentration values. A curve can be plotted as signal intensity vs. Ca2+ concentration. The signal intensity of an unknown Ca2+
concentration can then be determined by finding the corresponding reference value on the plot.
[0126] In some embodiments, the signal intensity changes by at least twenty percent as the Cl- concentration is raised. In some embodiments, the signal intensity changes by at least 10, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, or 90% or any derivable range therein when the Cl- concentration is raised.
[0127] In certain embodiments, the methods of the disclosure determine the Cl- concentration in a range of 1 mM to 100 mM, the range is inclusive of the recited Cl- concentration. For example, a Cl- concentration that is determined by the methods of the disclosure concentration is in a range of 1 mM to 50 mM, 1 mM to 25 mM, or 1 mM to 10 mM, or 5 mM to 100 mM, or 5 mM to 50 mM, 5 mM to 25 mM, or 5 mM to 10 mM, or 10 mM to 25 mM, or 10 mM to 50 mM, or 10 mM to 75 mM, or 20 mM to 80 mM, or 30 mM to 70 mM. In other embodiments, the recited Cl- concentration is excluded.
[0128] In some embodiments, the Cl- concentration is determined by comparing the measured signal to a reference value. In some embodiments, the Cl- concentration is determined by comparing the measured signal to a reference value. In some embodiments, the signal value and/or reference value is normalized. In some embodiments, the method further comprises creating a standard curve. A standard curve can be created by measuring the signal intensity at different known Cl- concentration values. A curve can be plotted as signal intensity vs. Cl- concentration. The signal intensity of an unknown Cl- concentration can then be determined by finding the corresponding reference value on the plot.
[0129] In some embodiments, the signal intensity changes by at least twenty percent as the pH is raised from at least one of pH 4 to pH 5, pH 5 to pH 6, pH 6 to pH 7, and pH 7 to pH 8. In some embodiments, the signal intensity changes by at least 10, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, or 90% or any derivable range therein when the pH is raised from at least pH 4, 5, 6, or 7 to pH 5, 6, 7, or 8 (or any range derivable therein). [0130] In certain embodiments, when pH ranges or values are discussed herein, the range is inclusive of the recited pH. For example, a pH range from 4.0 to 5.0 or between 4.0 and 5.0 includes the pH of 4.0 and 5.0. In other embodiments, the recited pH is excluded.
[0131] In some embodiments, the pH is determined by comparing the measured signal to a reference value. In some embodiments, the pH is determined by comparing the measured signal to a reference value. In some embodiments, the signal value and/or reference value is normalized. In some embodiments, the method further comprises creating a standard curve. A standard curve can be created by measuring the signal intensity at different known pH values. A curve can be plotted as signal intensity vs. pH. The signal intensity of an unknown pH can then be determined by finding the corresponding reference value on the plot.
[0132] As provided above, one aspect of the disclosure provides methods of determining pH and Ca2+ concentration in samples. Such methods include providing a nucleic acid complex including a Ca2+ fluorophore and a first label capable of producing a signal; measuring the intensity of the signal; and determining the pH and Ca2+ concentration from the measured signal. Another aspect of the disclosure provides methods of determining pH and Cl- concentration in samples. Such methods include providing a nucleic acid complex including a Cl- fluorophore and a first label capable of producing a signal; measuring the intensity of the signal; and determining the pH and Cl- concentration from the measured signal.
[0133] The methods of the disclosure, in certain embodiments, are suitable for measuring pH and concentration of Ca2+ or Cl- in early endosome, late endosome, plasma membrane, lysosome, autophagolysosome, recycling endosome, cis Golgi network (CGN), trans Golgi network (TGN), endoplasmic reticulum (ER), peroxisomes, or secretory vesicles. In certain embodiments, the methods of the disclosure, are suitable for measuring pH and
concentration of Ca2+ or Cl- in early endosome, late endosome, plasma membrane, lysosome, autophagolysosome, recycling endosome, or TGN.
[0134] In general, any sample containing Ca2+ or Cl- can be used in the methods of the disclosure. In some embodiments, the sample is a biological sample selected from a cell, cell extract, cell lysate, tissue, tissue extract, bodily fluid, serum, blood and blood product. In some embodiments, the sample is a live cell. In some embodiments, the sample is a biological sample from a patient.
[0135] The nucleic acid complexes as described herein can be readily introduced into a host cell, e.g., a mammalian (optionally human), bacterial, parasite, yeast or insect cell by any method in the art. For example, nucleic acids can be transferred into a host cell by physical, chemical or biological means. It is readily understood that the introduction of the nucleic acid molecules yields a cell in which the intracellular pH may be monitored. Thus, the method can be used to measure intracellular pH in cells cultured in vitro. The nucleic acid complex of the disclosure can also be readily introduced into a whole organism to measure the pH in a cell or tissue in vivo. For example, nucleic acid complex of the disclosure can be transferred into an organism by physical, chemical or biological means, e.g., direct injection.
[0136] In certain embodiments, the methods for introducing nucleic acid complexes of the disclosure may be those disclosed in Chakraborty et ai,“Nucleic Acid-Based Nanodevices in Biological Imaging,” Annu. Rev. Biochem. 85:349-73 (2016), incorporated in its entirety by reference herein.
[0137] Chemical means for introducing a polynucleotide into a host cell include colloidal dispersion systems, such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. One colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (i.e. , an artificial membrane vesicle). The preparation and use of such systems is well known in the art.
[0138] In some embodiments, the use of lipid formulations is contemplated for the introduction of the nucleic acid complex of the disclosure into host cells (in vitro, ex vivo or in vivo). In some embodiments, the nucleic acid complex of the disclosure may be associated with a lipid. The nucleic acid complex of the disclosure associated with a lipid may be encapsulated in the aqueous interior of a liposome, interspersed within the lipid bilayer of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the oligonucleotide(s), entrapped in a liposome, complexed with a liposome, dispersed in a solution containing a lipid, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with a micelle, or otherwise associated with a lipid. The lipid, lipid/nucleic acid complex compositions are not limited to any particular structure in solution. For example, they may be present in a bilayer structure, as micelles, or with a“collapsed” structure. They may also simply be interspersed in a solution, possibly forming aggregates which are not uniform in either size or shape.
[0139] In some embodiments, the one or more nucleic acid complexes of the disclosure are linked to a targeting sequence that directs the nucleic acid complex to a desired cellular compartment.
Diseases Detection and Monitoring
[0140] The methods, compositions, nucleic acid complexes, and kits of the disclosure can be used for the detection of diseases, the monitoring of diseases, and as a drug screening platform. In some embodiments of the disclosure, the disease is characterized as a lysosomal dysfunction disease. In some embodiments of the disclosure, the pathology of the disease includes lysosomal dysfunction.
[0141] Lysosomal dysfunction diseases include, for example, autosomal recessive osteopetrosis, Farber disease, Krabbe disease (infantile onset and late onset), Fabry disease (Alpha-galactosidase A), Schindler disease (Alpha-galactosidase B), Sandhoff disease (infantile, juvenile, or adult onset), Tay-Sachs, juvenile hexosaminidase A
deficiency, chronic hexosaminidase A deficiency, glucocerebroside, Gaucher disease (Type I, II, and III), lysosomal acid lipase deficiency (early onset and late onset), Niemann-Pick disease (Type A and B), sulfatidosis, metachromatic leukodystrophy (MLD), saposin B deficiency, multiple sulfatase deficiency, mucopolysaccharidoses: MPS I Hurler Syndrome, MPS I S Scheie Syndrome, MPS I H-S Hurler-Scheie Syndrome, Type II (Hunter syndrome), Type III (Sanfilippo syndrome), MPS III A (Type A), MPS III B (Type B), MPS III C (Type C), MPS MI D (Type D), Type IV (Morquio), MPS IVA (Type A), MPS IVB (Type B), Type VI (Maroteaux-Lamy syndrome), Type VII Sly Syndrome, Type IX (Hyaluronidase Deficiency); Mucolipidosis: Type I (Sialidosis), Type II (l-cell disease), Type III (Pseudo-Hurler
Polydystrophy / Phosphotransferase Deficiency), Type IV (Mucolipidin 1 deficiency);
Niemann-Pick disease (Type C and D), Neuronal Ceroid Lipofuscinoses: Type 1 Santavuori- Haltia disease / Infantile NCL (CLN1 PPT1), Type 2 Jansky-Bielschowsky disease / Late infantile NCL (CLN2/LINCL TPP1), Type 3 Batten-Spiel meyer- Vogt disease / Juvenile NCL (CLN3), Type 4 Kufs disease / Adult NCL (CLN4), Type 5 Finnish Variant / Late Infantile (CLN5), Type 6 Late Infantile Variant (CLN6), Type 7 CLN7, Type 8 Northern Epilepsy (CLN8), Type 8 Turkish Late Infantile (CLN8), Type 9 German/Serbian Late Infantile
(Unknown), Type 10 Congenital Cathepsin D Deficiency (CTSD); Wolman disease, alpha- mannosidosis, beta-mannosidosis, aspartylglucosaminuria, fucosidosis, lysosomal transport diseases, cystinosis, pycnodysostosis, salla disease / sialic acid storage disease, infantile free sialic acid storage disease (ISSD), glycogen storage diseases, Type II Pompe Disease, Type lllb Danon disease, and cholesteryl ester storage disease. In some embodiments, the disease is autosomal recessive osteopetrosis. In some embodiments, the disease is Niemann-Pick C disease.
Kits
[0142] The materials and components described for use in the methods may be suited for the preparation of a kit. Thus, the disclosure provides a detection kit useful for determining the pH and the presence, absence, or concentration of Ca2+ in a sample, cell or region thereof. Specifically, the technology encompasses kits for measuring the pH and Ca2+ of one or more cells in a sample. The disclosure also provides a detection kit useful for determining the pH and the presence, absence, or concentration of Cl- in a sample, cell or region thereof. Specifically, the technology encompasses kits for measuring the pH and Cl- of one or more cells in a sample. For example, the kit can comprise a nucleic acid complex as described herein.
[0143] In some embodiments, the methods described herein may be performed by utilizing pre-packaged diagnostic kits comprising the necessary reagents to perform any of the methods of the technology. For example, such a kit would include a detection reagent for measuring the pH and Ca2+ of a cell or region thereof, or a detection reagent for measuring the pH and CF of a cell or region thereof. In one embodiment of such a kit, the detection reagents are the nucleic acid complexes of the disclosure. Oligonucleotides are easily synthesized and are stable in various formulations for long periods of time, particularly when lyophilized or otherwise dried to a powder form. In this form, they are easily reconstituted for use by those of skill in the art. Other reagents and consumables required for using the kit could be easily identified and procured by those of skill in the art who wish to use the kit. The kits can also include buffers useful in the methods of the technology. The kits may contain instructions for the use of the reagents and interpreting the results.
[0144] In some embodiments, the technology provides a kit comprising at least one sample (e.g., a pH standard and/or a Ca2+ concentration standard and/or a CF concentration standard) packaged in one or more vials for use as a control. Each component of the kit can be enclosed within an individual container and all of the various containers can be within a single package, along with instructions for performing the assay and for interpreting the results of the assays performed using the kit.
[0145] In some embodiments, the kit comprises a device for the measurement of pH and Ca2+ in a sample. In some embodiments, the kit comprises a device for the measurement of pH and CF in a sample. In some embodiments, the device is for measuring pH and/or analyte in cell culture or in whole, transparent organisms (e.g., C. elegans).
Definitions
[0146] Throughout this specification, unless the context requires otherwise, the word “comprise” and“include” and variations (e.g.,“comprises,”“comprising,”“includes,” “including”) will be understood to imply the inclusion of a stated component, feature, element, or step or group of components, features, elements or steps but not the exclusion of any other integer or step or group of integers or steps.
[0147] As used in the specification and the appended claims, the singular forms“a,”“an” and“the” include plural referents unless the context clearly dictates otherwise. [0148] As used herein, a“fluorescent dye” or a“fluorophore” is a chemical group that can be excited by light to emit fluorescence. Some fluorophores may be excited by light to emit phosphorescence. Dyes may include acceptor dyes that are capable of quenching a fluorescent signal from a fluorescent donor dye.
[0149] As used herein,“crosslinked” refers to a covalent connection between the nucleic acid molecule and another moiety of interest, such as the Ca2+ fluorophore or the OG fluorophore. In certain embodiments, the crosslink between the nucleic acid molecule and this moiety is water compatible. In certain embodiments, the crosslink between the nucleic acid molecule and this moiety is stable under physiological conditions.
[0150] As used herein,“nucleic acid,”“nucleotide sequence,” or“nucleic acid sequence” refer to a nucleotide, oligonucleotide, polynucleotide, or any fragment thereof and to naturally occurring or synthetic molecules. The term "peptide nucleic acid" or "PNA" as used herein generally refers to nucleic acid analogue in which the sugar phosphate backbone of natural nucleic acid has been replaced by a synthetic peptide backbone. The term“RNA equivalent” in reference to a DNA sequence, is composed of the same linear sequence of nucleotides as the reference DNA sequence with the exception that all occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose. It is understood to be a molecule that has a sequence of bases on a backbone comprised mainly of identical monomer units at defined intervals. The bases are arranged on the backbone in such a way that they can enter into a bond with a nucleic acid having a sequence of bases that are complementary to the bases of the oligonucleotide. The most common oligonucleotides have a backbone of sugar phosphate units. A distinction may be made between oligodeoxyribonucleotides, which do not have a hydroxyl group at the 2' position, and oligoribonucleotides, which have a hydroxyl group in this position. Oligonucleotides also may include derivatives, in which the hydrogen of the hydroxyl group is replaced with organic groups, e.g., an allyl group. An oligonucleotide is a nucleic acid that includes at least two nucleotides.
[0151] One nucleic acid sequence may be“complementary” to a second nucleic acid sequence. As used herein, the terms“complementary” or“complementarity,” when used in reference to nucleic acids (i.e. , a sequence of nucleotides such as an oligonucleotide or a target nucleic acid), refer to sequences that are related by base-pairing rules. For natural bases, the base pairing rules are those developed by Watson and Crick. As an example, for the sequence“T-G-A”, the complementary sequence is“A-C-T.” Complementarity can be “partial,” in which only some of the bases of the nucleic acids are matched according to the base pairing rules. Alternatively, there can be“complete” or“total” complementarity between the nucleic acids. The degree of complementarity between the nucleic acid strands has effects on the efficiency and strength of hybridization between the nucleic acid strands.
[0152] Oligonucleotides as described herein may be capable of forming hydrogen bonds with oligonucleotides having a complementary base sequence. These bases may include the natural bases such as A, G, C, T and U, as well as artificial bases. An oligonucleotide may include nucleotide substitutions. For example, an artificial or modified base may be used in place of a natural base such that the artificial base exhibits a specific interaction that is similar to the natural base.
[0153] An oligonucleotide that is complementary to another nucleic acid will“hybridize” to the nucleic acid under suitable conditions (described below). As used herein,“hybridization” or“hybridizing” refers to the process by which an oligonucleotide single strand anneals with a complementary strand through base pairing under defined hybridization conditions.
“Specific hybridization” is an indication that two nucleic acid sequences share a high degree of complementarity. Specific hybridization complexes form under permissive annealing conditions and remain hybridized after any subsequent washing steps.“Hybridizing” sequences which bind under conditions of low stringency are those which bind under non- stringent conditions (6*SSC/50% formamide at room temperature) and remain bound when washed under conditions of low stringency (2*SSC, 42° C). Hybridizing under high stringency refers to the above conditions in which washing is performed at 2*SSC, 65° C (where SSC is 0.15M NaCI, 0.015M sodium citrate, pH 7.2).
EXAMPLES
[0154] Certain aspects of the disclosure are illustrated further by the following examples, which are not to be construed as limiting the disclosure in scope or spirit to the specific methods and materials described in them.
Materials and Methods
[0155] Reagents. All the chemicals were purchased from Sigma (USA) and Alfa Aesar (USA). 1H-NMR and 13C-NMR were recorded on Bruker AVANCE II+, 500MHz NMR spectrophotometer in CDCI3 and DMSO-d6 and tetramethylsilane (TMS) used as an internal stranded. Mass spectra were recorded in Agilent 6224 Accurate-Mass TOF LC/MS. All fluorescently labeled oligonucleotides were purchased from IDT (USA) and IBA-GmBh (Germany). HPLC purified oligonucleotides were dissolved in Milli-Q water to make 100 mM stock solutions and quantified using UV-spectrophotometer and stored at -20 °C. Ethylene glycol-bis^-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA), ampicillin, carbencillin, isopropyl b-D-l-thiogalactopyranoside (IPTG), nigericin and monensin were purchased from Sigma and ionomycin was obtained from Cayman Chemical (USA). Calcium nitrate tetrahydrate, sodium nitrate, sodium phosphate dibasic, sodium phosphate monobasic, magnesium sulfate anhydrous, glycerol, bovine serum albumin (66 kDa), nigericin, monensin, tributyltin chloride (TBT-CI) and amitriptyline hydrochloride were obtained from Sigma, and sodium chloride was purchased from Alfa Aesar. Magnesium nitrate
hexahydrate, sodium acetate anhydrous, sodium bicarbonate was purchased from Fisher Scientific (USA). 2-Hydroxypropyl^-cyclodextrin (b-CD) and U18666A were purchased from Cayman Chemical (USA). CellLight® Reagents* BacMam 2.0, DMEM and FBS were purchased from molecular probes from Life Technologies (USA). Maleylated BSA (mBSA) was maleylated according to an established protocol (Modi et al. (2009) Nat. Nanotechnol. 4:325-330). Monodisperse Silica Microspheres were obtained from Cospheric (USA).
[0156] Gel electrophoresis. Native polyacrylamide gels containing 12% acrylamide [19: 1 acrylamide/ bisacrylamide] were used for gel electrophoresis. Gels were run in 1 * TBE buffer (100 mM Tris - HCI, 89 mM boric acid, and 2 mM EDTA, pH 8.3) at 4°C. Gels were observed under Biorad Universal Hood II Gel Doc System (Bio-Rad Laboratories, Inc.) using Image Lab™ Software 6.0.0 for image acquisition for Alexa 647 and Alexa 546 channel.
After measurement, gels were stained with ethidium bromide (1 pg/mL) and observed.
[0157] In vitro fluorescence measurements. Fluorescence spectra were measured on a FluoroMax-4 spectrophotometer (Horiba Scientific, Edison, NJ) using previously established protocols (Modi et al. (2009)). 200 nM of lMLY, ChloropHore or ChloropHoreLY in UB4 buffer (20 mM HEPES, MES and sodium acetate, 150 mM KN03, 5 mM NaN03, 1 mM Ca(N03)2 and Mg(N03)2) of indicated pH were mixed and incubated at 37°C for 30 min after which measurements were performed at 25°C. For in vitro pH measurements, CalipHluorLy sample was diluted to 30 nM in pH clamping buffer [CaCI2 (50 mM to 10 mM), HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI2 (1 mM)] of desired pH and equilibrated for 30 minutes at room temperature. All the samples were excited at 495 nm and emission spectra was collected from 505 nm to 750 nm. The ratio of donor (D) emission intensity at 520 nm to acceptor (A) emission intensity at 665 nm was plotted as a function of pH to generate the pH calibration curve. Mean of D/A from two independent experiments and their S.E.M were plotted for each pH. Fold change in D/A of CalipHluorLy was calculated from the ratios of D/A at pH 4.0 and pH 6.5. pH1 2 of CalipHluorLy at different [Ca2+] values were derived from pH calibration curve by fitting to Boltzmann sigmoid.
[0158] For in vitro [Ca2+] measurements, CalipHluorLy sample was diluted to 30 nM in Ca2+ clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM),
KCI (140 mM), NaCI (5 mM) and MgCI2 (1 mM)]. The amount of [Ca2+] was varied from 0 mM to 20 mM and adjusted to different pH values (4.5-7.20). The amount of free [Ca2+] at a given pH was calculated based on Maxchelator software ( axchelator.stanford.edu). Rhod-5F and Alexa 647 were excited at 545 nm and 630 nm respectively. Emission spectra for Rhod-5F (O) and Alexa 647 (R) were collected from 570-620 nm and 660-750 nm, respectively. Mean of O/R from two independent experiments and their S.E.M. were plotted for each [Ca2+] Similar experiments were performed with 50nM CalipHluorwLy at pH 4.6 and pH 5.1. In vitro calcium binding affinity ( d) of Rhod-5F was obtained by plotting ratios of Rhod-5F (O) emission intensity at 580 nm to Alexa 647 (R) emission intensity at 665 nm as a function of free [Ca2+] and fitted using sigmoidal growth Hill (1) equation.
Y = S + (E - S) x (Xn/(K% + Xn )) (1)
X is free [Ca2+], Y is O/R ratio at given free [Ca2+], S is O/R ratio at low [Ca2+], E is O/R ratio at high [Ca2+], Kd is dissociation constant and n is Hill coefficient. Fold change response in O/R of CalipHluorLy was calculated from ratio of O/R at high [Ca2+] and O/R at low [Ca2+]
[0159] Determination of Stern-Volmer quenching constant Ksv. In vitro OG calibration curve of ChloropHore and ChloropHoreLY revealing the intensity ratio A647 and BAC (R/G) against [Cl-]. R/G at different chloride concentrations were normalized to the value at 5 mM chloride. R/G vs [OF] plot is equivalent to F0/F vs [OF] plot or the Stern-Volmer plot.
[0160] Circular Dichroism spectroscopy. CD scans were carried out on Jasco J-1500 CD Spectrometer equipped with a temperature controller. 7 mM unlabeled ChloropHore at pH 4.0 and 7.5 were prepared at 80 mM potassium phosphate buffer. Samples were then measured and reveal as an average of three scans.
[0161] C.elegans methods and strains. Standard methods were followed for the maintenance of C. elegans. Wild type strain used was the C. elegans isolate from Bristol, strain N2 (Brenner, 1974). Strains used in the study were provided by the Caenorhabditis Genetics Center (CGC), and are RRID:WB-STRAIN:RB2510 W08D2.5(ok3473) and RRI D: WBSTRAI N : VC1242 [+/mT1 II; cup-5(ok1698)/mT1 [dpy-10(e128)J lll\. Transgenics used in this study, also provided by the CGC, are RRID:WB-STRAIN:NP1 129 cdls131
[pcd::GFP::rab-5 + unc-119(+) + myo-2p::GFP], a transgenic strain that express GFP- fused early endosomal marker RAB-5 inside coelomocytes, RRID:WB-STRAIN:NP871 cdls66 [pcd::GFP::rab-7 + unc-119(+) + myo-2p::GFP], a transgenic strain that express GFP-fused late endosomal / lysosomal marker RAB-7 inside coelomocytes RRID:WB- STRAIN:RT258 pwls50 [Imp-1 ::GFP + Cbr-unc-119(+)], a transgenic strain expressing GFP- tagged lysosomal marker LMP-1 and arls37[myo-3p::ssGFP + dpy-20(+)]l, a transgenic strain that express ssGFP in the body muscles which secreted in pseudocoelom and endocytosed by coelomocytes and arls37[myo-3p::ssGFP + dpy-20(+)]lcup5(ar465) a transgenic strain with enlarged GFP containing vesicles in coelomocytes due to defective degradation. Gene knocked down was performed using Ahringer library-based RNAi methods. The RNAi clones used were: L4440 empty vector control, catp-6 (W08D2.5, Ahringer Library), catp-5 (K07E3.7, Ahringer Library) and mrp-4 (F21G4.2, Ahringer Library).
[0162] CalipHluor trafficking in coelomocytes. CalipHluor trafficking in coelomocytes was done in transgenic strains expressing endosomal markers such as GFP::RAB-5 (EE), GFP::RAB-7 (LE) and LMP-1 ::GFP (Ly) as described previously (Surana et al. (2011) Nat. Commun. 2:340). Briefly, worms were injected with CalipHluorA647 (500 nM) and incubated for specific time points and transferred on to ice. Worms were anaesthetized using 40 mM of sodium azide in M9 solution. Worms were then imaged on Leica TCS SP5 II STED laser scanning confocal microscope (Leica Microsystems, Inc., Buffalo Grove, IL) using an Argon ion laser for 488 nm excitation and He-Ne laser for 633nm excitations with a set of filters suitable for GFP and Alexa 647 respectively. Colocalization of GFP and CalipHluorA 647 was determined by counting the number CalipHluorA 647 positive puncta that colocalize with GFP- positive puncta and quantified as a percentage of total number of CalipHluorA 647 positive puncta (Surana et al. (2011) Nat. Commun. 2:340). In order to confirm lysosomal labeling in a given genetic background, the same procedure was performed on the relevant mutant or RNAi knockdown in pwls50 [Imp-1 ::GFP + Cb-unc-119(+)].
[0163] RNAi experiments in C.elegans. Bacteria from the Ahringer RNAi library expressing dsRNA against the relevant gene was fed to worms, and measurements were carried out in one-day old adults of the F1 progeny (Kamath and Ahringer, 2003). RNA knockdown was confirmed by probing mRNA levels of the candidate gene, assayed by RT-PCR. Briefly, total RNA was isolated using the Trizol-chloroform method; 2.5 pg of total RNA was converted to cDNA using oligo-dT primers. 5 pL of the RT reaction was used to set up a PCR using gene- specific primers. Actin mRNA was used as a control. PCR products were separated on a 1.5% agarose-TAE gel.
[0164] Cell culture methods and maintenance. BHK-21 cells, Human dermal fibroblasts (HDF), human fibroblast cells harboring mutations in ATP13A2 and homozygous for
1550C>T (L6025), J774A.1 and T47D cells were cultured in DMEM-F12 with 10% FBS, 100 U/ml penicillin and 100 pg/mL streptomycin and maintained at 37°C under 5 % C02.
Fibroblasts AG01518, GM08429, GM00112, GM16195, GM13205, GM03252, GM03393, GM11097, GM18414, GM23162 and GM17910 were purchased from Coriell Institute (Camden, NJ) and cultured with the suggested protocols from Coriell Institute.
[0165] Competition experiments. HDF cells were washed with 1 * PBS buffer and then incubated with 10 pM of maleylated BSA or BSA for 15 min. Next, cells were pulsed with 1 mM ChloropHore and 10 mM of maleylated BSA or BSA in cell culture media for 1 h. Cells were washed with 1 * PBS for three times and then imaged. Whole cell intensities of 20 cells per dish in the A647 channel was quantified.
[0166] Co-localization experiments in cells. Cells were transfected with LAMP1-RFP by CellLight® Reagents* BacMam 2.0 according to the manufacturer’s protocol. Briefly, CellLight® Reagents were added to the cells with the final number of particles per cell ca. 20 and incubated for 10 h. Transfected cells were then pulsed with 0.25 mg/ml_ of 10kDa FITC- Dextran (FITC-Dex) for 1 h, chased for 16 h and was followed by imaging. Crosstalk and bleed-through were measured and found to be negligible between the RFP channel and FITC-Dex channel. These experiments revealed that the pulse chase time point selected for FITC-Dex labelling of lysosomes showed ca. 80% colocalization with LAMP-1 RFP.
Lysosomes pre-labeled with TMR-Dex w utilized to examine the trafficking time scales for ChloropHore. Pre-labeled cells were pulsed with 1 mM of ChloropHore and chased for indicated time and imaged. Cross talk and bleed-through were recorded and disciver to be negligible between TMR channel and A647 channel. Pearson's correlation coefficient (PCC) measures the pixel-by-pixel covariance of two images while it range from 0-1 and 1 indicates complete colocalization . PCC are examined by the tool in ImageJ/Fiji 2.0.0-rc-54/1.51h. Upon pixel shift, PCC values decrease significantly suggesting non-random colocalization. For pH and Ca2+ sensors, pre-labeled cells were pulsed with 500nM of CalipHluorA647Ly and chased for indicated time and imaged. Cross talk and bleed-through were measured and found to be negligible between the TMR channel and Alexa 647 channel. Pearson's correlation coefficient (PCC) measures the pixel-by-pixel covariance in the signal levels of two images. Tools for quantifying PCC are provided in Fuji software. Pearson's correlation coefficient (PCC) measures the pixel-by-pixel covariance in the signal levels of two images. Tools for quantifying PCC are provided in Fuji software.
[0167] In cellulo clamping. pH and chloride clamping were carried out with ChloropHore using a previously published protocol. HDF cells were pulsed for 1 h and chased for 2 h with 2 mM of Chlorophore. After labeling, cells were fixed by 200 mL 4% paraformaldehyde for 5 min at 25 °C. The fixed cells were washed with 1 * PBS three times and incubated in the chloride clamping buffer of indicated pH and chloride concentration which also contained 50 mM nigericin, 50 mM monensin, and 25 mM tributyltin chloride (TBT-CI) for 1 h at 25 °C.
Clamping buffers with various concentration of chloride ions were prepared by adding chloride positive buffer (150 m M KCI, 5 mM NaCI, 1 mM CaCI2, 1 mM MgCI2, 10 mM HEPES, MES, sodium acetate) to a chloride negative buffer (150 mM KN03, 20 mM NaN03, 1 mM Ca(N03)2, 1 mM Mg(N03)2, 10 mM HEPES, MES, sodium acetate) at the same indicated pH in different ratios. The cells were then imaged in clamping buffer. pH and calcium clamping were carried out using CalipHluor"1^. Fibroblast cells were pulsed for 1 hour and chased for 2 hours with 500nM CalipHluor mLy. Cells are then fixed with 200 ml_ 4% paraformaldehyde (PFA) for 15 minutes at room temperature, washed three times and retained in 1 * PBS. To obtain the intracellular pH and calcium calibration profile, endosomal calcium concentrations were equalized by incubating the previously fixed cells in the appropriate calcium clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI2 (1 mM)] by varying amount of free [Ca2+] from 1 mM to 10 mM and adjusted to different pH values. The buffer also contained nigericin (50 pM), monensin (50 pM) and ionomycin (20 pM) and the cells were incubated for 2 hours at room temperature. For real-time pH and calcium measurements, fibroblast cells are pulsed with 500nM of CalipHluor mLy for 1 hour, chased for 9 hours (8 hours for L0625 cells) and then washed with 1 * PBS and imaged in Hank’s Balanced Salt Solution (HBSS). Imaging was carried out on IX83 research inverted microscope (Olympus Corporation of the Americas, Center Valley, PA) using a 100*, 1.42 NA, DIC oil immersion objective (PLAPON, Olympus Corporation of the Americas, Center Valley, PA) and Evolve Delta 512 EMCCD camera (Photometries, USA).
[0168] Single lysosome clamping. After the first round of cell clamping and imaging, the clamping buffer was replaced with the second which contained with 50 pM nigericin, 50 pM monensin, and 25 pM TBT-CI, either a pH or [CF] difference. The cells were incubated in the second clamping buffer for 1 h at room temperature post which the same cells were imaged.
[0169] DNA stability assay. DNA stability assay was performed as described previously. Cells are pulsed with 1 mg/ml_ TMR-Dex for 1 h and chased for 16 h. The TMR-Dex labeled cells are pulsed with 2 pM of ChloropHore for 1 h, chased for indicated time points for imaging.
[0170] Lysosomal pH and chloride measurements. Fibroblast cells are pulsed with 2 pM of lmLY, ChloropHore or ChloropHoreLY for 1 h, chased for 9 h (30 min pulse and 1 h chase for J774.A1, BHK-21 and T47D cells) and then washed with 1 * PBS and subjected for imaging.
[0171] 2-IM upon addition of lysosomal proton pump and ion channel blocker. Upon lysosome labeling, final concentration of 500 nM bafilomycinAI and 300 pM of 5-nitro-2-(3- phenylpropyl-amino) benzoic acid (NPPB) were added to cells and incubated for 45 min. Cells was then washed with PBS and imaged in Hank's Balanced Salt Solution (HBSS) containing the respective blocker compounds.
[0172] Preparation of pharmacologically induced cell culture model for Niemann Pick type A/B (NP-A/B) and type C (NP-C). Niemann Pick type A/B and C models were prepared according to the previous reported method. Cells were incubated with 65 mM of the acid sphingomyelinase (ASM) inhibitor amitriptyline (AH) and 20 mM of the NPC1 inhibitor U18666A for 24 h to create a NP-A/B and NP-C models respectively. The cells are pulsed with 2 pM of ChloropHoreLY and either 65 pM AH or 20 pM U18666A for 1 h, chased with either 65 pM AH or 20 pM U18666A for 9 h and then washed with 1 * PBS and imaged.
[0173] Lysosomal population rescue using ASM and b -CD. Recombinant human acid sphingomyelinase was incubated with NP-A/B patient cells according to the previous method. Primary human fibroblasts derived from NP-A/B patients were maintained in DMEM medium with 1% FBS for 24 h. The medium was then changed to DMEM with 1 % FBS containing 0.2% BSA and 5 pg of rhASM. After 24 h, the cells are pulsed with 2 pM of ChloropHoreLY for 1 h, chased for 9 h and then washed with 1 * PBS and imaged. Primary human fibroblasts from NP-C patient were incubated with 50 pM of o-Hydroxypropyl-b- cyclodextrin (b-CD) for 24 h. The cells are pulsed with 2 pM of ChloropHoreLY and 50 pM of b-CD for 1 h, chased with 50 pM of b-CD for 9 h and then washed with 1 * PBS and imaged.
[0174] Image acquisition. Image acquisition was carried out on wide field IX83 inverted microscope (Olympus Corporation of the Americas, Center Valley, PA) using a 60*, 1.42 NA, phase contrast oil immersion objective (PLAPON, Olympus Corporation of the
Americas, Center Valley, PA) and Evolve Delta 512 EMCCD camera (Photometries). Filter wheel, shutter and CCD camera were controlled using Metamorph Premier Ver 7.8.12.0 (Molecular Devices, LLC), suitable for the fluorophores used. Images on the same day were acquired under the same acquisition settings. Alexa 488 channel images (D) were obtained using 480/20 band pass excitation filter, 520/40 band pass emission filter and 89016- ET- FITC/Cy3/Cy5 dichroic filter. Alexa 647 channel images (A) were obtained using 640/30 band pass excitation filter, 705/72 band pass emission filter and 89016-ET-FITC/Cy3/Cy5 dichroic filter. FRET channel images were obtained using the 480/20 band pass excitation filter, 705/72 band pass emission filter and 89016-ET-FITC/Cy3/Cy5 dichroic filter. Rhod-5F channel images (O) were obtained using 545/25 band pass excitation filter, 595/50 band pass emission filter and a 89016-ET-FITC/Cy3/Cy5 dichroic filter. Confocal images were acquired on a Leica TCS SP5 II STED laser scanning confocal microscope (Leica
Microsystems, Inc., Buffalo Grove, IL) equipped with 63*, 1.4 NA, oil immersion objective. Alexa 488 was excited using an Argon ion laser for 488 nm excitation, Alexa 647 using He- Ne laser for 633 excitation and Rhod-5F using DPSS laser for 561 nm excitation with a set of dichroics, excitation, and emission filters suitable for each fluorophore.
[0175] Image analysis for pH and Cl sensors. Images were analyzed with ImageJ/Fiji 2.0.0-rc-54/1.51 h. pH and chloride measurements are performed as described previously.
To generate a density plot to represent lysosomal populations, the individual D/A (Y-axis) versus the corresponding R/G (X-axis) values was first plotted for each lysosome that was obtained for a given fibroblast sample comprising ~60 cells and -600 lysosomes. Density plots were generated using 2D Kernel Density plot on Origin 2018 SR1 b9.5.1.195
(OriginLab, USA). This converted the individual scatter points into a population matrix as a probability density function. The density plot is then pseudo colored to represent high density in red (values close to 1) and low density in blue (values closer to 0). Those points with <10% probability on the plot were discarded. For normalization between days, the point of highest density (value = 1) of normal HDF cells was designated as the center of the plot with x,y coordinates (1 ,1). To normalize between experiments done on different days, the lysosomal pH and Cl- of a sample of normal HDF cells was always measured as well, the density plot of which served as the reference. All density plots acquired on the same day with the same settings were thus normalized to the HDF cell density plots to enable comparison of data across different days.
[0176] Data was normalized as follows: The maximum and minimum values of D/A and R/G of the reference HDF cell density plot were considered as reference points to X and Y align the lysosome profile of a given sample. These maxima and minima were considered as 25% and 75% of the density plot respectively. The maxima and minima of D/A and R/G of newly obtained HDF cell data set was adjusted to this accordingly such that they overlaid. The center of the density profile (the point displaying highest density) was then adjusted to x = 1 , y = 1 coordinates. The density plot from other samples in the same set of experiments was aligned using the identical parameters used to align the HDF cell dataset with the reference HDF cell data set. The density plot of HDF cells obtained from independent experiments performed on different days were normalized and are shown in Figure 23 to demonstrate the reproducibility of this method.
[0177] Image analysis for pH and Ca2+ sensors. Image analysis for quantification of pH and calcium in single endosomes was done using custom MATLAB code. For each cell the most focused plane was manually selected in the Alexa 647 channel. This image and corresponding images from the same z-position in other channels were input into the program. Images from the different channels were then aligned using Enhanced Cross Correlation Optimization. To determine the location of the endosome first a low threshold was used to select the entire cell. Only the area within the cell was subsequently considered for endosome selection. Regions of interest corresponding to individual endosomes were selected in the Alexa 647 channel by adaptive thresholding using Sauvola's method. The initial selection was further refined by watershed segmentation and size filtering. After segmentation regions of interest were inspected in each image and selection errors were corrected manually. Using the cell boundary annular region 10 pixels wide around the cell was selected and used to calculate a background intensity in each image. Then, the mean fluorescence intensity was measured in each endosome in donor (D), acceptor (A), Rhod-5F (O) and Alexa 647 (R) channels and the background intensity corresponding to that cell and channel was subtracted. The two ratios of intensities (D/A and O/R) were then computed for each endosome. Mean D/A of each distribution was plotted as a function of pH and obtained the in vivo pH calibration curve. Mean O/R of each distribution was plotted as a function of free [Ca2+] to generate the in vivo Ca2+ calibration curve. Pseudo color pH and Ca2+ images were obtained by measuring the D/A and O/R ratio per pixel, respectively.
[0178] In vivo measurements of pH and [Ca2+]. In vivo pH calibration experiments of CalipHluorLy were carried out using protocols previously established (Modi et ai, 2009; Surana et ai, 2011). Briefly, CalipHluorLy (500 nM) was microinjected in pseudocoelom of young adult worms on the opposite side of the vulva. After microinjections, worms were incubated at 22 °C for 2 hours for maximum labelling of coelomocyte lysosomes. Then, worms were immersed in clamping buffer [CaCI2 (50 mM to 10 mM), HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI2 (1 mM)] of desired pH solutions containing the ionophores nigericin (50 pM), monensin (50 pM) and ionomycin (20 pM). Worm cuticle was perforated to facilitate the entry of buffer in to the body. After 75 min of incubations in clamping buffer, coelomocytes were imaged using wide-field microscopy. Three independent measurements, each with 10 worms, were made for each pH value.
[0179] Ca2+ clamping measurements were carried out using CalipHluorLy. Worms were injected with CalipHluorLy (500 nM) and incubated at 22 °C for 2 hours. After 2 hours, worms were immersed in Ca2+ clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI2 (1 mM)] by varying amount of free [Ca2+] from 1 pM to 10 mM and adjusted to different pH values (5.3-6.5). Three independent measurements, each with 10 worms, were made for Ca2+ value.
[0180] Early endosome and late endosome pH and free [Ca2+] measurements were carried out using CalipHluor, and lysosomal pH and free [Ca2+] measurements were carried out using CalipHluorLy. For real time pH and [Ca2+] measurements, 10 hermaphrodites were injected with 500 nM of CalipHluor and CalipHluorLy for EE, LE and Ly respectively and incubated for the indicated time points (5 min EE, 17 min LE, and 60 min Ly). Worms were anaesthetized using 40 mM of sodium azide in M9 solution and imaged on wide field microscopy. Image analysis was carried out using custom MATLAB code as described in image analysis. [0181] Calculating pH corrected [Ca2+] in EE, LE and Ly. The D/A and O/R ratios in Ly,
LE and EE were measured using CalipHluorLy and CalipHluor as mentioned above at single endosome resolution. Over 100 endosomes were analyzed in each measurement in worms to generate a Gaussian spread of D/A. Around 5%, endosomes which fell outside the range of Mean ± 2 S.D (S.D = standard deviation) which was set as a threshold for the
measurements in EE, LE and Ly. To get pH corrected [Ca2+] values, the pH value in each individual endosome was measured with single endosome resolution from their D/A ratios. pH values in endosomes were calculated using equation (2) which was derived from the in vivo pH calibration curve,
Figure imgf000052_0001
K K2 and pH1 2 represent parameters derived from a Boltzmann fit of the in vivo pH calibration curve, and Y represents the D/A ratio in a given endosome.
[0182] Next, the Kd of CalipHluorLy and fold change response in O/R ratios of CalipHluorLy from low [Ca2+] O/R to high [Ca2+] were obtained as functions of pH. The in vitro and in vivo Kd were measured at different pH points ranging from 4.5 to 7.2 by fitting Ca2+ calibration curves by fitting to the Hill equation (1). From in vitro and in vivo [Ca2+] calibration curves, the Kd of CalipHluorLy was plotted as a function of pH using following equation (3),
, pH , ,
Kd = 1.03 + 5.14 X 1012 X e c + 3.108 X 106 X e c TF (3)
[0183] By using equation (3), the Kd of CalipHluorLy can be deduced at any given pH in EE, LE and Ly. 0/Rmax (i.e. , O/R ratio at high [Ca2+]), was obtained by clamping the worms at 10 mM of free [Ca2+] at different pH points. In vitro and in vivo [Ca2+] calibration curves showed that CalipHluorLy retained its fold-change response of O/R from 1 mM to 10 mM at different pH points. O/Rmin (i.e., O/R ratio at low [Ca2+]) values were calculated (4) from fold change response as function of pH and normalized to 0/Rmax·
1
0/Rmin
4.24+0.12 xexp( 0.5 xpH) (4)
[0184] As mentioned above, the pH in EE, LE and Ly was measured from D/A by using equation (2) at single endosome resolution. pH and O/R, were used to calculate Kd and O/Rmin from equation (3) and (4). Finally, Kd, 0/Rmin, O/R and 0/Rmax were substituted in the following equation to get pH corrected free [Ca2+] values in endosome by endosome level.
Free [
Figure imgf000052_0002
[0185] Three independent measurements, each with 10 worms, were made for pH and
[Ca2+] values in EE, LE and Ly. [0186] Image analysis - pH corrected [Ca2+] images. High resolution images were acquired using confocal microscopy as mentioned in methods section. Images were acquired in four channels (Alexa 488, FRET, Rhod-5F and Alexa 647 channels) to quantify pH and [Ca2+] at single endosome resolution. To compensate for the pH component in Ca2+ measurements, the Kd of CalipHluorLy at single endo-lysosomal compartments was calculated based on the Kd calibration plot discussed above. The pH of endo-lysosomes was quantified by measuring the donor/acceptor values calibrated across physiological pH (4.0- 6.5). Donor (D) and acceptor (A) images were background subtracted by drawing an ROI outside the worms. Donor (D) image was duplicated and a threshold was set to create a binary mask. Background subtracted donor and acceptor images were then multiplied with the binary mask to get processed donor and acceptor images. This processed donor (D) image was divided by the processed acceptor (A) image to get a pseudocolor D/A image, using Image calculator module of ImageJ. The pH value was calculated by using the equation (2) formulated from in vivo and in vitro pH calibration plot.
[0187] The pseudo colored pH image was processed to get a Kd image as shown in Fig.3.
Kd of CalipHluorLy is a function of pH and this relation is formulated by the Kd calibration plot in vivo and in vitro using equation (3). For image processing of pH image to Kd image, background was set to a non-zero value. The Kd image represents the affinity of CalipHluorLy for calcium and thus compensating the calcium image (O/R) with Kd would precisely represent the calcium levels at single endo-lysosomes. The pH dependent Kd compensation is performed according to equation (5), where 0/Rmax and 0/Rmin are calculated by incubating CalipHluor coated beads at 10mM and 1 mM respectively. Image calculation were done using image calculator module in ImageJ. This image is multiplied with binary image to bring the background value to zero. The pH corrected Kd image were obtained for various mutants for accurate comparison of calcium levels in lysosomes.
[0188] Survival assay. +/mT1 II; cup-5(ok1698)/mT1 [dpy-10(e128)] III nematode strain was used for this assay. Homozygous lethal deletion of cup-5 gene is balanced by dpy-10- marked translocation. Heterozygotes are superficially wildtype [cup5+/-], Dpys (mT1 homozygotes) are sterile, and cup-5(ok1698) homozygotes are lethal. cup5+/- L4 worms were placed on plates containing RNAi bacterial strains for L4440 empty vector (positive control), mrp-4, catp-6, catp-5 and clh-6. These worms were allowed to grow for 24 hours and lay eggs after which the adult worms were removed from the plates. The eggs were allowed to hatch and grow to adult for 3 days. The worm plates were then imaged under Olympus SZX-Zb12 Research Stereomicroscope (Olympus Corporation of the Americas, Center Valley, PA) with a Zeiss Axiocam color CCD camera (Carl Zeiss Microscopy, Thornwood, NY). The images were analyzed using ImageJ software to count the number of adult worms per plate. Three independent plates were used for each RNAi background.
[0189] Lysosomal size recovery assay. arls37 [myo-3p::ssGFP + dpy-20(+)] I. cup- 5(ar465) is transgenic nematode strain which secretes GFP from the body muscle cells and this is endocytosed by coelomocytes which show enlarged GFP labelled vesicles as a result of defective degradation caused by cup 5 mutation. Similar to the previous assay, arls37 ; cup-5(ar465) L4 worms were placed on plates containing RNAi bacterial strains for empty vector (control), catp-6, catp-5 and mrp-4 (positive control). The worms lay eggs for 24 hours after which they are removed from the plates. The eggs thus hatch and grow to adulthood after which they were imaged to check for lysosomal size differences. Worms were imaged on a Leica TCS SP5 II STED laser scanning confocal microscope (Leica Microsystems, Inc., Buffalo Grove, IL) equipped with 63*, 1.4 NA, oil immersion objective upon excitation with Argon laser in the Alexa 488 channel. Lysosomal areas were measured using ImageJ. Out 100 lysosomes in arls37 worms, 7 lysosomes had an area in range of 7.0-9.5 pm2. Enlarged lysosomes are defined as those lysosomes whose diameter is > 33% of the diameter of the largest lysosome observed in normal N2 worms. The lysosomal area in arls37 ; cup-5(ar465) worms were measured in various RNAi bacteria containing plates. Lysosomal size recovery data was plotted as percentage of area occupied by large lysosomes to the total lysosomal area (n = 15 cells, > 100 lysosomes).
[0190] Bead calibration of CalipHluoiJ1lLy. Bead calibration was performed using
CalipHluormLy coated 0.6pm Monodisperse Silica Microspheres (Cospheric, USA). Briefly, silica microspheres were incubated in a solution of 5pM CalipHluorwLy in 20mM Sodium Acetate buffer (pH= 5.1) and 500mM NaCI for 1 h. This binding solution was spun down and the beads were reconstituted in clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (500 mM) and MgCI2 (1 mM)]. The amount of [Ca2+] was varied from about OmM to 10 mM and adjusted the pH to either pH 4.6 or pH 5.1. The beads were incubated in clamping buffer for 30 mins after which there were imaged on a slide on the IX83 inverted microscope in the G, O and R channels to obtain G/R (pH) and O/R (Ca2+) images.
[0191] Competition experiments in cells. HDF cells were washed with 1 * PBS buffer pH 7.4 prior to labeling. Cells were incubated with 10 pM of maleylated BSA (mBSA) or BSA for 15 minutes and pulsed with media containing 500 nM CalipHluormLy and 10 pM of mBSA or BSA for 1 hour to allow internalization by receptor mediated endocytosis, washed 3 times with 1 * PBS and then imaged under a wide-field microscope. Whole cell intensities in the Alexa 647 channel was quantified for >30 cells per dish. The mean intensity from three different experiments were normalized with respect to the autofluorescence and presented as the fraction internalized.
Example 1 : Preparation of Ca2+ fluorophore
Figure imgf000055_0002
1
Rhod-5F ester
Figure imgf000055_0001
[0192] BAPTA-5F aldehyde (1) was synthesized according to previous reported procedure (Grynkiewicz et al. (1985) J. Biol. Chem. 260:3440-3450; Collot et al. (2015) eUfe
4:e05808). POCI3 (1.12g, 7.3 mmol) was added to DMF (5 ml_) at 0 °C and allowed to stir for 10 minutes. After 10 minutes, BAPTA-5F (1.6 g, 2.9 mmol) in DMF (3ml_) was added to above solution and heated to 65 °C. After completion of the reaction, reaction mixture was poured in water and pH adjusted to 6.0 by adding aqueous. NaOH (1 M) solution. Product was extracted with ethylacetate (3x50 ml_) and solvent was evaporated. Crude product was purified by column chromatography on silica gel using hexane/EtOAc (70/30 to 60/40) as an eluent to obtain BAPTA-5F aldehyde (1) in 65% yield. 1 H-NMR (500 MHz, CDCI3) <5ppm 9.80 (s, 1 H), 7.30-7.45 (m, 2H), 6.83 (t, 1 H, J = 7 Hz), 6.76 (d, 1 H, J = 8 Hz), 6.59 (t, 2H, J = 8 Hz), 4.40 (t, 2H, J = 7.0 Hz), 4.35 (t, 2H, J = 7.5 Hz), 4.33 (s, 4H), 4.22 (s, 4H), 3.59 (s, 6H), 3.57 (s, 6H). 13C-NMR (125 MHz, CDCI3) <5ppm 190.5, 171.8, 171.2, 159.4, 157.5, 151.4,
151.3, 149.6, 145.1 , 135.6, 135.5, 130.0, 126.9, 120.3, 120.2, 116.6, 110.8, 107.3, 107.1 ,
101.3, 101.1 , 67.1 , 67.0, 53.5, 53.4, 51.9, 51.7. HRMS (ESI) m/z. [M]+ calcd for
C27H31 FN2011 + 578.1912, found: 578.1927.
[0193] To a solution of BAPTA-5F aldehyde (1) (50 mg, 0.086 mmol) in propionic acid (4 ml_), 3-(dimethylamino) phenol (26 mg, 0.19 mmol) and p-Toluenesulfonic acid (p-TSA) (1.5 mg, 0.009 mmol) were added and allowed to stir at room temperature for 12 hours. After 12 hours, Chloranil (21 mg, 0.086 mmol) in dichloromethane (3 ml_) was added to above reaction mixture and allowed to stir at room temperature overnight. After completion of the reaction, the crude product was extracted with dichloromethane (3x30 ml_). The crude product was then purified by column chromatography on silica gel using
dichloromethane/methanol (95/5 to 90/10 %) as an eluent to obtain Rhod-5F ester as a dark red solid in 35 % yield. LCMS (ESI) m/z [M]+ calculated for C43H48FN4O1 815.3298, found: 815.5. Rhod-5F ester (5 mg, 0.006 mmol) was dissolved in methanol and water mixture (1 :0.5 ml_), to which KOH (3.5 mg, 0.063mmol) was added and allowed to stir for 8 hours at room temperature. After completion of the reaction, solution pH was adjusted to 6.0 and crude Rhod-5F was extracted with dichloromethane (3x5 ml_). Product was purified by HPLC (50:50 acetonitrile/water, 0.1 % TFA) to obtained Rhod-5F. LCMS (ESI) m/ [M]+ calculated for C39H40FN4O1 759.2672, found: 759.4.
Example 2: Preparation of crosslinking-ready Ca2+ fluorophore
Figure imgf000056_0001
[0194] To a solution of 1-bromo-3-chloropropane (1 g, 6.4 mmol) in DMSO (8 ml_), sodium azide (0.5 g, 7.7 mmol) was added and allowed to stir at room temperature for 12 hours. After completion of the reaction, the mixture was diluted with water and the product was extracted with hexane to obtain 1-azido-3-chloropropane. Sodium iodide (1.5 g, 10 mmol) was then added to a solution of 1-azido-3-chloropropane (1 g, 8.4 mmol) in acetone (25 ml_) and allowed to stir at room temperature for 8 hours. After completion of the reaction, the solvent was evaporated under vacuum. The crude product was diluted with a saturated solution of Na2S203 to quench the unreacted iodine followed by extraction of the compound with ethyl acetate (3x50 ml_). This was dried over Na2S04 and the product 1-azido-3- iodopropane was used for further reactions without purification.
[0195] To a solution of 3-aminophenol (1 g, 9.2 mmol) in acetone (30 ml_), potassium carbonate (2.5 g, 18.4 mmol) was added and allowed to stir at room temperature for 20 min. After 20 minutes, iodomethane (1.3 g, 9.2 mmol) was added and the mixture was further stirred for 8 hours at room temperature. After completion of reaction, the solvent was evaporated and the crude product was extracted with dichloromethane (3x30 ml_). This was followed by purification of the crude product by column chromatography on silica gel using hexane/ ethyl acetate (80/20 %) as an eluent to obtained 3-(methylamino) phenol in 45 % yield.
[0196] To a solution of 3-(methylamino) phenol (1 g, 8.1 mmol) in DMF (8 ml_), N, N- diisopropylethylamine (1.26 g, 9.7 mmol) was added and stirred for 20 minutes at room temperature. After 20 minutes, 1-azido-3-iodopropane (1.7 g, 8.1 mmol) was added to above reaction mixture and heated at 65 °C for 8 hours. After completion of the reaction, the solvent was evaporated and the crude product was extracted with diethylether (3x40 ml_). Then, the crude product was purified by column chromatography on silica gel using hexane/ethyl acetate (90/10 %) as an eluent to obtained 3-((3-azidopropyl)(methyl)amino)phenol liquid in 72 % yield. 1H-NMR (500 MHz, CDCI3) <5ppm 7.09-7.13 (m, 1 H), 6.3 (d, 1 H, J = 7.5 Hz), 6.21 (dd, 2H, J = 2 Hz, 8.5 Hz), 3.42 (t, 2H, J = 6.5 Hz), 3.38 (t, 2H, J = 7 Hz), 2.94 (s, 3H), 1.87 (t, 2H, J = 6.5 Hz). 13C-NMR (125 MHz, CDCI3) <5ppm 156.7, 150.7, 130.2, 105.1 , 103.5, 99.3, 49.8, 49.2, 38.6, 26.3. HRMS (ESI) m/z [M]+ calculated for C10H14N4O+ 206.1168, found:206.1177.
[0197] To a solution of BAPTA-5F aldehyde (1) (50 mg, 0.086 mmol) in propionic acid (4 ml_), 3-((3-azidopropyl)(methyl)amino)phenol (40 mg, 0.19 mmol) and p-Toluenesulfonic acid (p-TSA) (1.5 mg, 0.009 mmol) were added and allowed to stir at room temperature for 12 hours. After 12 hours, Chloranil (21 mg, 0.086 mmol) in dichloromethane (3 ml_) was added to above reaction mixture and allowed to stir at room temperature overnight. After completion of the reaction, the solvent was evaporated and the crude product was extracted with dichloromethane (3x20 ml_). The crude product was then purified by column
chromatography on silica gel using dichloromethane/methanol (95/5 to 90/10 %) as an eluent to obtain Rhod-5F-OMe as a dark red solid in 30 % yield. 1H-NMR (500 MHz, DMSO- d6) <5ppm 7.55 (d, 2H, J - 8 Hz), 7.15-7.16 (m, 3H), 7.00-7.04 (m, 3H), 6.88 (dd, 2H, J = 3 Hz,
9 Hz), 6.75 (dd, 1 H, J = 6 Hz, 9 Hz), 6.65 (td, 1 H, J = 3 Hz, 6 Hz), 4.20-4.30 (m, 8H), 4.02 (s, 4H), 3.71 (t, 4H, J = 7 Hz), 3.53 (s, 6H), 3.47 (s, 10H), 3.25 (s, 6H), 1.88 (q, 4H, J = 7 Hz). 13C-NMR (125 MHz, DMSO-d6) <5ppm 171.2, 171.1 , 158.3, 157.3, 156.4, 156.1 , 140.7, 135.2, 135.1 , 131.9, 123.6, 123.1 , 119.1 , 116.8, 1 14.9, 114.4, 106.4, 106.2, 101.2, 101.0, 96.4,
67.3, 67.2, 54.9, 53.2, 53.0, 51.5, 51.2, 49.7, 48.6, 48.1 , 26.0, 22.1. HRMS (ESI) m/z [M]+ calculated for C47H54FN1o011 + 953.3952, found: 953.3967.
[0198] Rhod-5F-OMe (5 mg, 0.005 mmol) was dissolved in methanol and water mixture (1 :0.5 ml_) and KOH (3.5 mg, 0.063mmol) was added and allowed to stir for 8 hours at room temperature. After completion of the reaction, pH was adjusted to 6.0 and crude Rhod-5F-N3 was extracted with dichloromethane (3x5 ml_). Product was purified by HPLC (1 : 1 acetonitrile: water, 0.1 % TFA). LCMS (ESI) m/z. [M]+ calculated for C4 H FN10OI I+ 897.33, found: 897.5.
Example 3: Preparation of Ca2+ fluorophore conjugate
[0199] Rhod-5F-N3 (25 mM) was added to 5 pM of dibenzocyclooctyne (DBCO) labelled single-stranded nucleic acid molecule in 100 pL of sodium phosphate (10 mM) buffer containing KCI (100 mM) at pH 7.0 and allowed to stir overnight at room temperature. After completion of the reaction, 10 pL of 3 M sodium acetate (pH 5.5) and 250 pL of ethanol were added to reaction mixture and kept overnight at -20 °C for DNA precipitation. Then, the reaction mixture was centrifuged at 14000 rpm at 4 °C for 20 minutes to remove the unreacted Rhod-5F-N3 and the precipitate was re-suspended in ethanol and centrifuged.
This procedure was repeated 3 times for complete removal of unreacted Rhod-5F-N3. Rhod- 5F conjugation was confirmed by gel electrophoresis by running a native polyacrylamide gel containing 15% (19: 1 acrylamide: bis-acrylaimde) in 1 c TBE buffer (Tris HCI (100 mM), boric acid (89 mM), EDTA (2 mM), pH 8.3).
Example 4: Preparation of nucleic acid complexes of the disclosure
[0200] Sequences used to form CalipHluor, CalipHluorLy and CalipHluor171^ are provided in Table 1. D1 and D2 were used to form CalipHluorLy·, OG-D1 and D2 were used to form CalipHluor mLy. Bromo cytosines in D1 are bold and underlined. 01-A488, 02-A647 and 03 strands were used to form CalipHluor.
[0201] Rhod-5F was first conjugated to D2 strand or 03-DBCO strand as provided in Example 3. To prepare a CalipHluorLy and CalipHluo mLy sample, 5 pM of D1 or OG-D1 and 5 pM of Rhod-5F conjugated D2 strands were mixed in equimolar ratios in 10 mM sodium phosphate buffer (pH 7.2) containing 100 mM of KCI. The solution was heated to 90 °C for 15 minutes, cooled to room temperature at 5 °C per 15 minutes and kept at 4 °C for overnight (Modi et al. (2009) Nat. Nanotechnol. 4:325-330). For CalipHluor, 5 pM of 01- A488, 5 pM of 02-A647 and 5 pM of Rhod-5F conjugated 03 strands were mixed in equimolar ratios in 10mM sodium phosphate buffer at pH 5.5 containing 100 mM of KCI. Solution was heated to 90 °C for 15 minutes, then cooled to room temperature at 3 °C per 15 minutes and kept at 4 C for overnight.
Table 1.
Strand Sequence information (SEQ ID NO.)
5'- Alexa 488 - CCC CTA ACC CCT AAC CCC TAA CCC CAT ATA TAT CCT
D1
AGA ACG ACA GAC AAA CAG TGA GTC - 3' (SEQ ID NO:01)
5' - DBCO - GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /iAIexa 647N/
D2
AT ATT TTG TTA TGT GTT ATG TGT TAT-3' (SEQ ID NO:02)
5' - Alexa - 488 - CCCCAACCCCAAT ACATTTT ACGCCTGGTGCC - 3' (SEQ
01-A488
ID NO:03)
5' - CCGACCGCAGGATCCT AT AAAACCCCAACCCC - Alexa 647 - 3' (SEQ
02-A647
ID NO:04)
5'-TTA TAG GAT CCT GCG GTC GG /iDBCON/ GGC ACC AGG CGT AAA
03-DBCQ
ATG TA -3' (SEQ ID NO:05)
5'-Oregon Green - AT AAC ACA TAA CAC ATAACAAAA TAT ATA TCC TAG
OG-D1
AAC GAC AGA CAA ACA GTG AGT C - 3' (SEQ ID NO:06)
[0202] The formation of CalipHluorLy and CalipHluor were validated by electrophoretic mobility assay, using Native and Denaturing polyacrylamide gel electrophoresis (PAGE). Copper free click reaction of Rhod-5F-N3 to DBCO labeled strand (D2 strand for
CalipHluorLy and 03-DBCO strand for CalipHluor) was validated by 15% denaturing PAGE run in 1 * TBE, at 120 V for 3h. The slower mobility of Rhod-5F conjugated strand, due to addition of 1 KDa (Rhod-5F) to 10 KDa (DBCO-strand). Rhod-5F conjugation was further confirmed by recording the gel in TMR channel, where the lower mobility band shows strong fluorescence. Rhod-5F labeled strand was purified and hybridized with normalizing and pH sensing module as described in methods section. 12% Native PAGE was run to characterize the formation of complete sensor. 12% Acrylamide:bisacrylamide resolves duplex DNA from ssDNA. Slower mobility of CalipHluorLy and CalipHluor owing to higher molecular weight validates the formation, at very high yield (> 99%). This further confirmed with slower mobility band, shows fluorescence at Alexa 488, TMR (Rhod-5F) and Alexa647 channel.
Example 5: In vitro characterization of nucleic acid complexes of the disclosure
[0203] CalipHluorLy is a 57-base pair DNA duplex comprising two strands D1 and D2 and bears three distinct domains (Figure 1A, Table 1). The first domain in CalipHluorLy is a Ca2+- reporter domain that uses a novel small molecule that functions as a Ca2+ indicator denoted Rhod-5F. Rhod-5F consists of a BAPTA core, a rhodamine fluorophore (Aex = 560 nm; Aem = 580 nm) and an azide linker. In the absence of Ca2+, the rhodamine fluorophore in Rhod-5F is quenched by photoinduced electron transfer (PeT) from the BAPTA core. Upon Ca2+ chelation quenching is relieved resulting in high fluorescence. Note that protonation of the amines in BAPTA also relieves PeT. Thus, the percentage change in signal as well as the dissociation constant (Kd) in Rhod-5F will be affected as a function of pH. The Kd of Rhod- 5F for Ca2+ binding is pH dependent and shown in Figure 6.
[0204] Rhod-5F is attached to the D2 strand bearing a dibenzocyclooctyne (DBCO) group using click chemistry. Conjugation to D2 did not change the Kd of Rhod-5F in CalipHluorLy (Figure 1 B). In CalipHluorLy Rhod-5F (O, orange diamond) shows a Kd of 1.1 mM at pH 7.2 which increases as acidity increases (Figure 1 B). CalipHluorLy was characterized by gel electrophoresis (Figure 7A-7B).
[0205] For ratiometric quantification of Ca2+ Alexa 647 was incorporated as a reference dye (Aex = 630 nm; Aem = 665 nm) on CalipHluorLy positioned so that it does not FRET with Rhod- 5F. Alexa 647 was chosen for its negligible spectral overlap with Rhod-5F and insensitivity to pH, Ca2+ and other ions (sphere, Figure 1A). The fixed stoichiometry of Alexa 647 efficiently corrects for Rhod-5F intensity changes due to inhomogeneous probe distribution in cells, thus making the ratio of Rhod-5F (O) and Alexa 647 (R) intensities in CalipHluor probes proportional to pH and Ca2+. The second domain (gray line) constitutes a DNA based pH-reporter domain as previously described, called the l-switch (Figure 1A). This /- switch has been used to map pH in diverse endocytic organelles in living cells.
[0206] To map pH in early and late endosomes CalipHluor, a variant suited to the lower acidities in these organelles, is used. CalipHluor is comprises three strands 01 , 02, and 03 (Figure 7C; Table 1). CalipHluor were characterized by gel electrophoresis (Figure 7D-7E). The third‘integration’ domain comprises a 30-mer duplex that integrates the pH and the Ca2+ reporter domains into a single DNA assembly. One end is fused to the l-switch and the other is fused to the Ca2+ sensor. This domain also helps in targeting, because its anionic nature aids recognition and trafficking by scavenger receptors in a DNA sequence independent manner (Surana et al. (2011) Nat. Cornmun. 2:340).
[0207] The response characteristics of CalipHluor and CalipHluorLy were investigated as a function of pH as well as Ca2+ and their pH and Ca2+ sensitive regimes were determined (Figure 1C-1 D and Figure 7F-7G). A 3D surface plot of D/A as a function of pH and different values of free [Ca2+] is shown in Figure 1 C. These revealed that the pH reporting capabilities of CalipHluor and CalipHluorLy are between pH 5.0 - 7.0 and pH 4.0 - 6.5 with fold changes in D/A ratios of 4.0 and 5.5, respectively (Figure 1C). The fold changes in D/A ratios were invariant over a range of free Ca2+ concentrations from 20 nM-10 mM showing that pH sensing by these probes is unaffected by Ca2+ levels (Figure 1 C).
[0208] In parallel, the intensities of Rhod-5F (O) and Alexa647 (R) in CalipHluorLy obtained from direct excitation yielded O/R values. An analogous 3D surface plot of O/R values as a function of [Ca2+] and pH showed a sigmoidal increase as a function of Ca2+ with a about 9 fold change in O/R at pH 7.2 (Figure 1 D). At lysosomal pH in C. elegans, i.e., pH 5.5, CalipHluorLy showed a Kd of 7.2 mM. As expected, the percentage signal change upon chelating Ca2+ also decreases as acidity increases (Figure 1 D).
Example 6: In vivo performance of nucleic acid complexes of the disclosure
[0209] The in vivo reporter characteristics of CalipHluorLy were investigated as a function of lumenal pH and [Ca2+] When DNA-based reporters are injected into the pseudocoelom in C. elegans they are specifically uptaken by coelomocytes through the scavenger receptors mediated endocytosis and thereby label organelles on the endolysosomal pathway. After labeling endocytic organelles with CalipHluorLy thus, lumenal pH and [Ca2+] of coelomocytes were clamped. This was achieved by incubating worms in clamping buffers of fixed pH and [Ca2+] containing nigericin, monensin, ionomycin and EGTA at high [K+] which clamped the endosomal ionic milieu to that of the surrounding buffer. Post-clamping, the worms were then imaged in four channels; (i) the donor channel (D or Alexa 488) (ii) the FRET acceptor channel (A), which corresponds to the intensity image of A647 fluorescence upon exciting A488, (iii) the orange channel (O or Rhod-5F), and (iv) the red channel (R) which
corresponds to the intensity image of A647 fluorescence upon directly exciting Alexa 647. Figure 2A (i-iv) shows representative images of a CalipHluorLy labeled coelomocyte imaged in the four channels.
[0210] In a given clamping buffer of specified pH and Ca2+ concentration, the ratio of the donor channel (D) image to the acceptor channel (A) image yields a D/A image which corresponds to the clamping buffer pH (Figure 2A, v). Similarly, the O/R image corresponds to the Ca2+ concentration at that pH (Figure 2A, vi). Representative D/A and O/R images of coelomocytes clamped at the indicated pH and [Ca2+] are shown in Figure 2B and Figure 8. The distribution of D/A and O/R values of lysosomes clamped at different indicated pH and [Ca2+] values are shown in Figures 2C-2D. To compare the in vivo and in vitro sensing performances of both ion-sensing modules across a wide range of pH and Ca2+ two parameters were plotted for each module in CalipHluorLy. For the pH sensing module these were the fold change in D/A (FCD/A), as well as the transition pH (pH1 2) (Figures 2E-2F, Figures 9A-9D). For the Ca2+ sensing module, these were the fold change in O/R (FC0/R) as well as the Kd for Ca2+ (Figures 2G-2H). The values of FCD/A, FC0/R, pH1 2 and Kd in vivo and in vitro were consistent revealing that the in vitro performance characteristics of CalipHluorLy was quantitatively recapitulated in vivo (Figures 2E-2F and Figures 2I-2K).
[0211] The calibrated D/A and O/R ratios were calculated by measuring the intensity values at single lysosome resolution in all four channels, as described in methods. Plotting D/A against pH values, at different Ca2+ concentration shows the insensitivity of pH sensing module towards Ca2+ levels. Figures 9A-9D show the comparison of D/A vs pH plots at different Ca2+ levels between in vitro and in vivo. Non-significant change in fold change and pH1 2, shows the robustness of the nucleic acid complexes of the disclosure in biological systems. In vivo Ca2+ clamping of coelomocytes was performed in Ca2+ clamping buffer [HEPES (10 mM), MES (10 mM), sodium acetate (10 mM), EGTA (10 mM), KCI (140 mM), NaCI (5 mM) and MgCI2 (1 mM)] by varying amount of free [Ca2+] from 1 mM to 10 mM and adjusting pH (5.5-6.5) in presence of nigericin (50 mM), monensin (50 pM) and ionomycin (20 pM). Because of difficulty clamping coelomocytes below 1 pM of free [Ca2+], clamping points <1 pM were extrapolated from the in vivo calibration to get the Kd of CalipHluorLy at pH 6.5 and 5.7. As shown in Figures 9E-9H, in vivo [Ca2+] calibration profiles of CalipHluorLy correspond well with in vitro calibration profiles from pH 5.5 to 6.5 and from 1 pM to 10 mM of free [Ca2+] Further, in vivo Kd values of CalipHluorLy as function of pH were consistent with in vitro Kd values (Figure 2J). These results confirm that pH and [Ca2+] sensing properties of CalipHluorLy were preserved in coelomocytes.
[0212] In receptor mediated endocytosis, endocytosed cargo traffics through the early endosomes (EE) and late endosomes (LE) to reach lysosomes (Ly) for degradation and recycling. To find out estimated time points of CaiipHiuor^ 7 to reach EE, LE and Ly, time dependent colocalization experiments were performed in worms expressing GFP tagged endosomal markers GFP::RAB-5 (EE), GFP::RAB-7 (LE) and LMP-1 ::GFP (Ly). These results indicate that CaiipHiuor^ 7 is present in EE, LE, and Ly at 5 minutes, 17 minutes, and 60 minutes, respectively. These time points were used to measure the pH and [Ca2+] in EE, LE and Ly in wild-type worms.
Example 7: Measuring [Ca2+] in organelles of the endo-lysosomal pathway
[0213] Endosomal maturation, critical to both organelle function and cargo trafficking, is accompanied by progressive acidification of the organelle lumen (Figure 3A). Unlike pH, little is known about lumenal Ca2+ changes as a function of endosomal maturation. The applicability of the probe was demonstrated across a range of acidic organelles by mapping lumenal Ca2+ as a function of endosomal maturation. The time points at which CalipHluorLy localized in the early endosome, the late endosome and the lysosome were determined in coelomocytes as described previously (Surana et al. (2011) Nat. Commun. 2:340) (Figures 3B-3C). Post injection, CalipHluorLy was found to localize in early endosomes (EE), late endosomes (LE) and lysosomes (LY) at 5, 17 and 60 minutes, respectively (Figures 3B-3C, Figure 10). [0214] The pH and apparent Ca2+ were measured at each stage in wild type N2 nematodes with single endosome addressability using the probes. A Kd correction factor was then incorporated for each endosome according to its measured pH, and then computed the true value of Ca2+ in every endosome. Figure 3D shows a representative set of coelomocytes for which this method of analysis was performed. Early and late endosomes were labeled with CalipHluor whereas lysosomes were labeled with the CalipHluorLy variant and then generated the D/A and O/R maps of coelomocytes (Figure 3D, i and iv). The D/A map was directly converted into a pH map using the calibrated D/A values obtained from the in vivo pH clamping experiments (Figure 3D, ii, Table 2). The in vivo and in vitro Ca2+ response characteristics at every pH (Figure 2H), provides the Kd for Ca2+ at every pH value for both CalipHluor and CalipHluorLy.
Table 2. Free [Ca2+] in clamping buffer at pH 5.5 calculated using Maxchelator software.
Added calcium Amount calcium Concentration of Free [Ca2*] (mM) in 50 mΐ
(mM) added (mI_) calcium added
0 0 0 0
1 1 50 mM 3.89 E-2
2 2 50 mM 7.80 E-2
10 1 0,5 mM 3.89 E-1
20 2 0.5 mM 7.80 E-1
50 1 2.5 mM 1.9
100 2 2.5 mM 3.9
200 1 10 mM 7.9
500 1 25 mM 20.4
1 E3 1 50 mM 43.1
2E3 2 50 mM 96.3
5E3 1 250 M 360.3
10E3 2 250 mM 1.88 E3
20E3 2 500
Figure imgf000063_0001
[0215] Using the pH map in Figure 3D, ii, a "Kd map" was constructed which corresponds to the Kd for Ca2+ at each pixel in the pH map (Figure 3D, iii). Multiplying the value of Kd at each pixel in the Kd map with the equation (O/R - 0/Rmin)/(0/Rmax-0/R) the true Ca2+ map was obtained (Figure 3D, v). In this equation, O/R corresponds to the observed O/R value at a given pixel in the O/R map, 0/Rmin and 0/Rmax correspond to O/R values at 1 mM and 10 mM Ca2+ at the corresponding pH value at that particular pixel. Thus, the pH and Ca2+ maps of early endosomes, late endosomes and lysosomes were obtained in N2 worms (Figure 3E) and the corresponding distributions of D/A and pH-corrected O/R are shown in Figures 3F- 3G. The mean values of pH and Ca2+ in each endosomal stage is shown in Figures 3H-3I.
[0216] Table 3 provides mean pH and free [Ca2+] in EE, LE and Ly of wild type (N2) worms, lysosomes of catp-6, cup-5 7- and catp-6 RNAi in cup-5 7- worms using CalipHluorLy. pH decreases progressively with endosomal maturation, with lumenal acidity showing a about 3- fold decrease at each endocytic stage. In contrast, Ca2+ in the early endosome and the late endosome were comparable and fairly low i.e. , 0.3 mM. Interestingly, from the late endosome to the lysosome, lumenal Ca2+ increases sharply by about 35 fold, indicating a stage-specific enrichment of Ca2+ and consistent with the lysosome being an acidic Ca2+ store (Table 3). The 100-fold difference between lysosomal and cytosolic Ca2+ is consistent with the stringent regulation of lysosomal Ca2+ channels to release lumenal Ca2+ and control lysosome function.
Table 3
Worm pH Free
Figure imgf000064_0001
EE of N2 6.48 + 0.07 0.3 ± 0.1 LE of U2 5.95 ± 0.02 0.3 ± 0.1 Ly of N2 5.30 + 0.02 11 + 0.8
Ly of catp-6 5.47 ± 0.03 1.6 ± 0.4 Ly of cup-5 +/- 5.15 + 0.01 40 ± 1.5
Ly of CATP-6 RNAs in cup-5 +/- 5.50 ± 0.10 16 ± 4.9
Early endosome (EE), Late endosome (LE) and Lysosomes (Ly)
For all experiments n = 15 cells, 50 endosomes; data represent the mean ± S.E.M.
Experiments were repeated thrice independently with similar results.
Example 8: Catp-6 is identified as a potential lysosomal Ca2+ importer
[0217] This surge in lumenal Ca2+ specifically in the lysosome stage, implicates the existence of factors that aid lysosomal import of Ca2+. However, players that mediate lysosomal Ca2+ accumulation are still unknown in higher eukaryotes. Inspiration was taken from the well-known Ca2+ importer i.e., SERCA, a P-Type ATPase which is present on the endoplasmic reticulum (ER). Other Ca2+ importers like plasma membrane Ca2+ ATPase (PMCA) and the secretory pathway Ca2+ ATPase (SPCA1) are also P-type-ATPases.
Potential P-type ATPases in the human lysosomal proteome were manually identified. It was found that the P5-ATPase ATP13A2 was described to transport cations like Mn2+, Zn2+,
Mg2+, and Cd2+ but not Ca2+ based on toxicity assays. As Ca2+ homeostasis is critical to all major signaling pathways, compensatory mechanisms in cells can counter excess Ca2+ and thereby omit the identification of Ca2+ transport by ATP13A2.
[0218] C. elegans has two homologs of ATP13A2 i.e., catp-5 and catp-6 (Figure 4A). To test whether catp-6 mediated lysosomal Ca2+ accumulation, it was investigated whether its knockdown would rescue a phenotype arising due to high lumenal Ca2+ (Figure 4B).
TRPML1 is a well-known lysosomal Ca2+ release channel whose knockdown would be expected to elevate lysosomal Ca2+. Mutations in TRPML1 result in lysosomal dysfunction that leads to the lysosomal storage disease Mucopolysaccharidoses Type IV (MPS IV). In C. elegans, loss of cup-5, the C. elegans homolog of TRPML1 , results in lysosomal storage and embryonic lethality. Therefore it was also tested whether catp-6 knockdown a cup-5+/- genetic background could reverse cup-5 -I- lethality. In this strain, the homozygous lethal deletion of cup-5 is balanced by dpy-10 marked translocation. A survival assay was performed by knocking down specific genes in cup-5+/- worms and scoring for lethality (Figure 4C and Figure 11).
[0219] Multidrug resistance protein-4 (MRP-4) is a versatile efflux transporter for drugs, toxins, peptides and lipids and is known to rescue cup-5-/- lethality. It is hypothesized that in the absence of cup-5, mrp-4 mis-localizes in endocytic compartments causing toxicity that is then alleviated upon its knockdown. RNAi knockdown of either catp-6 or catp-5 rescued cup- 5-/- lethality favorably compared to mrp-4 knockdown (Figure 11). Knocking down clh-6, another lysosome-resident channel that regulates lumenal chloride, showed no such rescue.
Example 9: Catp-6 facilitates lysosomal Ca2+ accumulation
[0220] Given that the rescue of lethality might occur without restoring lysosomal function, it was tested whether any of the candidate genes reversed lysosomal phenotypes. Cup-5 knockdowns show abnormally large lysosomes due to lysosomal storage. Therefore the hypomorph ar645 was used with a G401 E mutation in cup-5 leading to dysfunction that is insufficient for lethality, yet leads to engorged lysosomes. In the arls37,cup-5(ar465) strain, soluble GFP that is secreted from the muscle cells into the pseudocoelom is internalized by the coelomocytes and trafficked for degradation to dysfunctional lysosomes. Thus, in these worms, the lysosomes in coelomocytes are abnormally enlarged and labeled with GFP (Figure 4D).
[0221] RNAi knockdowns of catp-6 in these nematodes rescued lysosomal morphology (Figures 4D-4E). Knocking down either catp-5 or mrp-4 showed only a marginal recovery of phenotype. Given that mrp-4 is not a lysosome resident protein and its inability rescue the lysosomal phenotype suggests that mechanistically, its rescue of cup-5 -/- lethality is likely to be extra lysosomal, consistent with previous hypotheses. [0222] Next, it was checked whether cafp-6-mediated rescue of a physical phenotype i.e., lysosome morphology, also led to a restoration of a chemical phenotype, i.e., its lumenal Ca2+. Lysosomal Ca2+ measurements using CalipHluorLy in cup-5 +/- nematodes and in catp- 6 knockdowns. Wild type nematodes showed lysosomal Ca2+ levels of 1 1 ± 0.8 mM (Figures 4F-4H). In cup-5 +/- nematodes lysosomal Ca2+ was elevated to 40 ± 1.5 mM, consistent with cup-5 being a Ca2+ release channel (Figures 4F-4H). Interestingly, catp-6 knockdown restored lysosomal Ca2+ to wild-type levels. Thus catp-6 function directly opposes that of cup-5 as it rescues cup-5 deficient phenotypes at three levels - the whole organism in terms of lethality, at the sub-cellular level in terms of lysosome phenotype, at sub-organelle level in terms of its lumenal chemical composition. Cumulatively, these indicate that catp-6 facilitates lysosomal Ca2+ import. Accordingly, catp-6 deletion led to lysosomal Ca2+ dropping to 1.6 ± 0.4 pM, consistent with it facilitating Ca2+ import.
Example 10: ATP13A2 facilitates lysosomal Ca2+ accumulation
[0223] Mutations in ATP13A2, the human homolog of catp-6, belong to the PARK9
Parkinson's disease (PD) susceptibility locus. These mutations lead to the Kufor-Rakeb syndrome, a severe, early onset, autosomal recessive form of PD with dementia. PD is strongly connected to Ca2+ dysregulation as excessive cytosolic Ca2+ causes excitotoxicity of dopaminergic neurons. Overexpressing ATP13A2 suppresses toxicity and reduces cytosolic Ca2+. Further, loss of ATP13A2 function leads to neuronal ceroid lipofuscinosis, a lysosomal storage disorder, implicating the lysosome as its potential site of action.
[0224] To confirm whether ATP13A2 also facilitated lysosomal Ca2+ import, lysosomal Ca2+ in human fibroblasts was mapped. A variant called CalipHluorwLy suited to measure the high acidity of mammalian lysosomes was created (Figure 12A). CalipHluormLy showed similar pH and Ca2+ response characteristics in vitro, on beads and in cellulo and its Ca2+ sensing characteristics are unaffected by the new pH sensing module (Figures 12B-12E)
[0225] CalipHluormLy was localized in lysosomes of primary human dermal fibroblasts (HDF cells) obtained from punch-skin biopsies. CalipHluormLy labels lysosomes in HDF cells by scavenger receptor mediated endocytosis (Figures 5A-5B; Figure13). Briefly, a 1 hour pulse of 500 nM CalipHluormLy followed by a 9 hour chase efficiently labels lysosomes in this cell type (Figures 5A-5B).
[0226] Lysosomal Ca2+ was measured in fibroblasts from normal individuals and L6025 primary fibroblasts isolated from male patients with Kufor Rakeb syndrome, that are homozygous for a OT mutation in 1550 of ATP13A2. This mutation results in ATP13A2 being unable to exit the ER and the lysosomes are devoid of ATP13A2. After confirming its lysosomal localization in L6025 cells, using CalipHluorwLy lysosomal pH and Ca2+ were measured (Figures 5B-5D). Lysosomes in KRS patients showed 14-fold lower Ca2+ and about 2-fold lower [H+] than normal (Figures 5C-5E) confirming that ATP13A2 mediates lysosomal Ca2+ accumulation.
Example 11 : Design and in vitro characterization of ChloropHore
[0227] ChloropHore is a 61-base pair DNA duplex comprising three strands C1 , C2 and P (Table 4) and bears three distinct domains (Figure 14A). Two of these are fluorescent, ratiometric reporter domains that are previously reported, namely a CF reporter domain, Clensor and a pH-reporter domain called the l-switch. Each reporter domain is fused to either end of an“integration domain”, which comprises a 27-mer duplex, that serves to integrate the pH and the CF reporter domains into a single DNA assembly. This 27-mer duplex also helps in targeting, because its anionic nature aids recognition and trafficking by scavenger receptors in a DNA sequence independent manner. To match the pH sensing regime of ChloropHore to the low pH regimes encountered in mammalian lysosomes ChloropHoreLy, a variant that used modified 5'-bromocytosines in the C-rich region, was also made (Table 4). Sequences used for ChloropHoreLY assembly are oligo P, oligo C1-Br and oligo C2.
[0228] Specifically, formation of ChloropHore was validated by electrophoretic mobility shift assay utilizing native polyacrylamide gel electrophoresis (PAGE) (Figure 18). The hybridization of C2 and P1 was revealed by the lower mobility of sample (C2 + P) compared with sample C2 in ethidium bromide (EB) and Alexa 647 channel. Meanwhile, the lowest electrophoretic mobility of sample (C1 + C2 + P1) in EB, Alexa 546 and Alexa 647 channel indicates the formation of ChloropHore which constructed by C1 , C2 and P1 in excellent yield.
[0229] To validate the conformational change of ChloropHore upon acidification, circular dichroism (CD) spectrometer was employed to validate the structural change in vitro.
ChloropHore shows a positive peak at 272 nm and a negative peak at 248 nm in pH 7.5 characteristic of duplex DNA. However, at pH 4.0, a positive peak at 285 nm and a negative peak at 248 nm was observed. The difference spectra of ChloropHore at pH 4.0 and pH 7.5 showed a positive peak at 292 nm and a negative peak at 263 nm which consistent with the CD signature of an i-motif. This was also how i-motif formation was proved in the parent /- switch.
[0230] In Figure 14, the sensing characteristics of the H+ sensing module were
demonstrated to be unaffected by integration to the CF sensing module and vice versa thus enabling potential detection of these two ions in parallel. The response of ChloropHore to various ions such as Mg(N03)2, Ca(N03)2, NaN03, Na2HP04, MgS04, NaHC03 and CH3COONa and 30% glycerol was also investigated (Figure 20). This reveals that the sensitivity of ChloropHore to diverse biologically abundant ions including Na+, Ca2+, Mg2+, N03- and P04 3- is negligible. BAC is also quenched by halides such as bromide and iodide (Brand I-). However, the combined concentrations of all these ions including CIST and SCN- in biological cells are < 0.1 % the value of chloride. At this low bioavailability, their contribution to the changes in BAC fluorescence in biological systems is negligible hence BAC acts as a chloride sensor in biological systems.
[0231] The formation and specificity of ChloropHore and ChloropHoreLy were confirmed by a gel shift assay, circular dichroism spectroscopy and UV melting studies (Figures 18-20).
Table 4
Figure imgf000068_0001
[0232] The fluorescence response characteristics of ChloropHore and ChloropHoreLy were investigated as function of pH and [Cl-] in order to determine their pH and [Cl-] sensitive regimes. The gradual increase of D/A ratio of ChloropHore and ChloropHoreLy revealed their pH reporting capabilities between pH 5.5 and 6.5 (Figure 14B) and pH 4.5 and 6.5 respectively (Figure 21 B), the latter being well suited to measure the pH of highly acidic human lysosomes. Both ChloropHore and ChloropHoreLy show a sigmoidal increase in D/A as a function of pH in the sensitive regime, and fold changes in D/A ratios of 5.5 and 3 respectively. Notably, the fold change in D/A ratio remained invariant for both ChloropHore and ChloropHoreLy over [Cl-] ranging from 5 mM to 120 mM (Figure 14C and Figure 21C). Thus, the pH sensing characteristics of ChloropHore and ChloropHoreLy are insensitive to changes in physiological [Cl ]. This is illustrated by a 3D surface plot of D/A as a function of pH performed at different fixed values of [Cl-] is shown in Figure 14D (Figure 21 D).
[0233] In parallel, the R/G ratio shows a linear dependence with increasing [Cl-], showing about 2.5 fold change upon increasing [CF] from 5 mM to 120 mM (Figures 14B-14C and Figures 21 B, 21 C). Again, for both ChloropHore and ChloropHoreLy the Stern Volmer constant (Ksv) and the fold change in R/G stayed constant as a function of pH from pH 4.5- 7.0 (Figure 14C and Figure 21C). This is illustrated by a 3D surface plot of R/G as a function of CF performed at different fixed values of pH as shown in Figure 14E (Figure 21 E). Thus, the [CF] sensing characteristics of ChloropHore and ChloropHoreLy are insensitive to changes in physiological pH. This indicates that in ChloropHore and ChloropHoreLy, the sensing characteristics of the H+ sensing module is unaffected by integration to the CF sensing module and vice versa thus enabling potential detection of these two ions in parallel.
Example 12: Two Ion measurement with single endosome addressability
[0234] To simultaneously measure lysosomal pH and [CF] in live cells, ChloropHore to the lysosomes of human dermal fibroblasts (HDF) was targeted. Human dermal fibroblasts (HDF) express scavenger receptors (SR) that uptake anionic ligands. Therefore DNA nanodevices can be trafficked to organelles on the endolysosomal pathway in diverse living systems (Figure 15A). Upon incubating ChloropHore with HDF cells for 1 h (referred to as 1 h "pulse"), uptake into punctate endosomes was excellent (Figures 15B, 15D, 15F). The uptake was effectively competed out by 10 equivalents of maleylated BSA revealing that in HDF cells, ChloropHore is internalized via the SR pathway (Figures 15B-15C).
[0235] The time required for lysosomal localization of ChloropHore was then estimated by its time-dependent colocalization with a lysosomal marker. Pulsing HDF cells with 10 KDa fluorescent dextrans (0.25 mg/ml_) for 1 h followed by a 16 h chase effectively marked lysosomes, as revealed by colocalization with LAMP1-RFP (Figures 15D-15E). Next, ChloropHore was pulsed in HDF cells where lysosomes were pre-labeled with TMR-Dextran as above, washed and imaged the cells at various chase times. ChloropHore and TMR- Dextran showed maximal co-localization at 9 h (Figures 15F-15G, Figure 22). Stability measurements revealed that ChloropHore and ChloropHoreLy had a half-life of 20 hours and were stable for at least 10 h in HDF lysosomes (Figure 22).
[0236] Next, the in-cell pH and [CF] sensing characteristics of ChloropHore were
investigated. Lumenal pH and [CF] in C/7/orop/-/ore-labeled HDF cells was clamped by incubation in clamping buffers of fixed pH and [CF] containing nigericin, monensin and tributyltin chloride at high [K+] Figure 16A shows representative fluorescence images of a cell clamped at the indicated pH and [CF] imaged in the D, A, R and G channels along with the corresponding D/A and R/G maps. Histograms of D/A and R/G values of 150 endosomes clamped at different pH and [Cl-] are shown in Figure 23. ChloropHore showed a ~5.5 fold- change in D/A value from pH 4.0 to pH 7.0 across all tested values of [Cl-], with the in vitro 3D surface plot being quantitatively recapitulated in cells (Figure 16B, Figure 24). Similarly, ChloropHore response in terms of R/G as a function of [CF] was performed at different fixed values of physiological pH (Figure 16C). Both the Ksv and fold-change in R/G from 5 mM to 120 mM [CF] was constant from pH 4.0-7.0, with the in vitro 3D surface plot of R/G being quantitatively recapitulated in cells (Figure 16C, Figure 24). This revealed that ChloropHore and ChloropHoreLy can simultaneously report pH and [CF] with performance characteristics in cells that closely match their in vitro pH and [CF] sensing properties.
[0237] Next, both ions in endosomes were simultaneously mapped while retaining this information with single endosome addressability in live cells. Therefore, the D/A value - reflecting lumenal pH - in a given endosome is plotted against the R/G value in the same endosome - reflecting lumenal [CF] - for 150 endosomes, which is represented as a scatter plot with each data point corresponding to a single endosome (Figure 16D). For clearer visualization, this data is represented as a density plot color coded according to their frequencies of occurrence (Figure 16E). This is a method of simultaneous quantitative imaging of two ions in a single endosome, while retaining concentration information with single endosome addressability as two-ion measurement (2-IM) and the corresponding density plot as a 2-IM profile. Figures 16F-16H shows the 2-IM profile of 150 endosomes clamped at the same [CF] (100 mM) but different pH, while Figures 16I-16K shows the 2-IM profile of 150 endosomes clamped at the same pH (pH 6.5), but different [CF]
[0238] To illustrate the capability of 2-IM to address single endosomes, ChloropHoreLy- labeled HDF were subjected cells to different clamped states of pH and [CF] in series due to its optimal pH responsivity in lysosomes (Figures 25). ChloropHoreLy labeled endosomes were clamped at the indicated pH and CF and imaged in D, A, R and G channels (Figures 161, 16N). Figure 16L(i-ii) shows the D/A and corresponding R/G maps of cells clamped at pH 4.5 and 5 mM CF. The D/A and corresponding R/G maps of these same cells
subsequently clamped at the same value of [CF], but at pH 6.5 are shown in Figure 16L(iii— iv) (Figures 26). Figure 16M shows a plot of individual endosomes in each clamped state, with black lines connecting the same endosome in either clamped state. It is clear that when [CF] was constant and pH changed, all the endosomes show increased D/A and negligible variation of R/G, moving parallel to the Y-axis. Similarly, Figure 16N(i— iv) shows the D/A and R/G maps of cells clamped at 100 mM CF and pH 6.5 that have subsequently been clamped at the same value of pH, but at 5 mM [CF] Again, when pH was constant and [CF] was changed, individual endosomes moved from right to left, parallel to the X-axis (Figure 160). Thus, in addition to population measurements, 2-IM provides information with single endosome resolution.
Example 13: Two Ion measurement chemically resolves lysosome populations
[0239] The 2-IM profile of lysosomes in fibroblasts of healthy individuals reveals populations with two distinct chemotypes. ChloropHoreLy labeled lysosomes in HDF cells were imaged to obtain D/A and R/G maps (Figure 17A(i)-(ii)). The corresponding 2-IM profile revealed a major population of -68% lysosomes that contained relatively low chloride, with R/G < 1.3 (Figure 17B(i)). However, there is a minor population (-23%) with R/G > 1.5 and D/A < 1.25 with higher lumenal CF and proton content (Figure 17B(i)). This was consistent over experimental replicates on samples derived from the same individual as well as across multiple normal individuals (Figure 17B(i)-(vi)). 2-IM in lysosomes of diverse cell types such as BHK-21 cells, murine macrophages and T-47D cells also showed lysosomes chemotypes with these characteristics (Figures 27-29). The high CF, high acidity population was lost upon pharmacologically inhibiting either V-ATPase or chloride channels with bafilomycin A1 or 5-nitro-2-(3-phenylpropylamino) benzoic acid, that selectively block lysosomal proton or CF accumulation respectively (Figures 17C-17D).
[0240] In order to understand how these lysosomal populations are affected upon pathological lysosomal storage, fibroblasts of patients with lysosomal storage disorders were subjected to 2-IM profiling. Lysosomal storage disorders arise due to genetic defects in proteins that affect the lysosomal degradation of specific biomolecules. Further,
dysfunctional lysosomes in a range of lysosomal storage disorders show reduced lumenal CF and/or H+ as a result of flawed lysosomal integrity. 2-IM was applied to study three related lysosomal storage disorders, i.e. , the Niemann Pick disease variants, due to their similarity of presentation, the fact that mutations lie in only one of three identified genes, all three gene products are lysosome-resident, and importantly, therapeutics are available for these diseases.
[0241] Niemann Pick A (NP-A) and Niemann Pick B (NP-B) diseases arise due to defects in the enzyme acid sphingomyelinase (ASM), for which enzyme replacement therapy is available. Niemann Pick C (NP-C) arises due to defective cholesterol transport due to mutations in any one of two key proteins, NPC1 or NPC2, and for which clinical trials using cyclodextrin derivatives are under way. Three patient samples corresponding to NP-A and NP-C disease and two for NP-B were studied based on sample availability, common mutations and characterization by enzyme activity (Table 5). Table 5
Figure imgf000072_0001
DA = day, Mo = month, Yr = year, Fw = fetal week
[0242] After verifying that ChloropHoreLy could label the lysosomes in every patient sample (Figure 30, g-l) were each subjected to 2-IM (Figure 17E). Typical D/A and R/G maps of ChloropHoreLy labeled lysosomes in fibroblasts derived from NP-A and NP-C patients are shown in Figure 17A(iii)-(vi). The 2-IM profiles of fibroblasts derived from skin biopsies of all the patient samples showed that the high chloride, high acidity lysosome population was absent (Figure 17E(i)-(viii)). Particularly, the 2-IM profile of NP-C patient samples showed a high degree of monodispersity compared to healthy samples (Figure 17E(vi)-(viii)).
[0243] Although lysosomal pH correlates with spatial position in certain cell types, no such dependence is available yet for lysosomal Cl-. Peripheral lysosomes show lower acidity in C2C12 murine myoblasts, human adipose microvascular endothelial cells and HeLa cells, but not primary human dendritic cells or CHO cells. Interestingly, 2-IM profiles in primary HDF cells showed that peripheral lysosomes had higher pH than perinuclear lysosomes (Figure 17A(i), arrowhead). Further, in NP-A and NP-C cells the spatial heterogeneity in lysosomal pH is lost, with lysosomes becoming uniformly hypoacidic (Figure 17A(iii)&(v)). In contrast to pH, no such spatial correlation could be observed for lumenal CF in HDF cells from normal individuals (Figure 17A(ii)). However, in NP-A and NP-C patient samples, the lumenal CFof the perinuclear lysosomes was more affected and lower than those of peripheral lysosomes (Figure 17A(iv)&(vi)). Taken together, this suggests that the milieu of the perinuclear lysosomes is more compromised than the rest, and that the high chloride high acidity population could correspond to these lysosomes. The correlation between lumenal ions and lysosome size is provided in Figure 17F (i)-(iv). These studies revealed that the high chloride lysosomes were smaller in size, and their numbers were depleted in patient cells. No such clear correlation emerged for lysosomal pH.
Example 14. ChloropHoreLy enables evaluation of therapeutic efficacy
[0244] In order to understand how the high chloride high acidity population was affected upon inducing pathological lysosomal storage, cell culture models of NP-A/B and NP-C diseases were created. In order to mimic the sphingomyelin and cholesterol accumulation characterizing these disorders, HDF cells from a healthy individual were treated with amitriptyline hydrochloride (AH) and U18666A that inhibit ASM and NPC1 respectively. 2-IM was performed after verifying that ChloropHoreLy effectively labels lysosomes in these cells (Figure 30, c-f). The 2-IM profiles showed a monodisperse lysosome population in both cell culture models, and a depletion of the high chloride, high acidity lysosome population (Figure 17G(i)&(ii)).
[0245] Next, it was investigated whether this high chloride, high acidity lysosome population could be recovered upon complementing patient cells with the corresponding therapeutic. Recombinant human acid sphingomyelinase (rhASM) is used to treat NP-A/B patients by enzyme replacement therapy. Fibroblasts from NP-A and NP-B patient samples were incubated with rhASM as outlined in literature. These cells are expected to internalize rhASM from the extracellular milieu, and traffic it to lysosomes, where it degrades sphingomyelin and mitigates storage. The 2-IM profile of patient cells treated with rhASM showed the reemergence of the high chloride high acidity lysosome population (Figure 17H(i)&(ii)).
[0246] NP-C does not arise from the deficiency of a degradative enzyme, but rather due to a transport defect, for which there is still no FDA approved therapeutic. However, o-2- hydroxypropyl^-cyclodextrin (bqϋ) treatment for 24 h has been shown to improve cholesterol transport from the lysosome to the endoplasmic reticulum thereby delivering cholesterol to the ER for esterification and reducing storage in the lysosome. Interestingly, treating NP-C patient samples mutated either in NPC1 (Figure 17H(iii)&(iv)) or NPC2 (Figure 17H(v)) with bΰϋ showed a loss of monodispersity in the 2-IM profile and the appearance of lysosomes with higher lumenal CF and H+ content.
[0247] Some embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Of course, variations on these described embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventor expects skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.
[0248] Various exemplary embodiments of the disclosure include, but are not limited to the enumerated embodiments listed below, which can be combined in any number and in any combination that is not technically or logically inconsistent.
[0249] Embodiment 1 provides a method for simultaneously determining 1) pH, and 2) Ca2+ concentration or Cl- concentration in a sample comprising:
providing a nucleic acid complex comprising
a first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore or a Cl- fluorophore crosslinked to the first strand; and
a second single-stranded nucleic acid molecule that is partially or fully complementary to the first single-stranded molecule,
wherein the nucleic acid complex further comprises a first label conjugated to the first single-stranded nucleic acid molecule or the second single- stranded nucleic acid molecule and the first label is capable of producing a signal, wherein the intensity of the signal is dependent on change in pH; measuring the intensity of the signal; and
determining the pH, and Ca2+ or Cl- concentration from the measured signal.
[0250] Embodiment 2 provides the method of embodiment 1 , wherein the sample is a biological sample selected from a cell, cell extract, cell lysate, tissue, tissue extract, bodily fluid, serum, blood, and blood product.
[0251] Embodiment 3 provides the method of embodiment 1 , wherein the sample is a live cell.
[0252] Embodiment 4 provides the method of any of embodiments 1-3, wherein determining is in early endosome, late endosome, plasma membrane, lysosome, autophagolysosome, recycling endosome, cis Golgi network, trans Golgi network, endoplasmic reticulum, peroxisomes, or secretory vesicles.
[0253] Embodiment 5 provides the method of any of embodiments 1-4, wherein the nucleic acid complex comprises the Cl- fluorophore crosslinked to the first strand.
[0254] Embodiment 6 provides the method of embodiment 5, wherein the Cl- fluorophore comprises 10,10'-bis[3-carboxypropyl]-9,9'-biacridinium dinitrate.
[0255] Embodiment 7 provides the method of any of embodiments 1-4, wherein the nucleic acid complex comprises the Ca2+ fluorophore crosslinked to the first strand. [0256] Embodiment 8 provides the method of embodiment 7, wherein the Ca2+ fluorophore is a single wavelength indicator.
[0257] Embodiment 9 provides the method of any of embodiment 7, wherein the Ca2+ fluorophore comprises Rhod-5F, XRhod-5F, Rhod-FF, XRhod-FF, Oregon Green 488 BAPTA-6F, Fluo 5F, Fluo 4FF, Oregon Green BAPTA-5N, Fluo-5 N, or Mag-Fluo-4 indicator.
[0258] Embodiment 10 provides the method of any of embodiments 7-9, wherein the Ca2+ fluorophore comprises Rhod-5F, XRhod-5F, or Rhod-FF indicator.
[0259] Embodiment 11 provides the method of any of embodiments 7-9, wherein the Ca2+ fluorophore comprises Rhod-5F indicator.
[0260] Embodiment 12 provides the method of any of embodiments 7-11 , wherein the Ca2+ fluorophore is crosslinked to the first strand through a triazole, thioether, or alkenyl sulfide group.
[0261] Embodiment 13 provides the method embodiment 12, wherein the Ca2+ fluorophore further comprises a linker moiety configured to form the triazole, thioether, or alkenyl sulfide group through a reaction of an azide or thiol moiety on the Ca2+ fluorophore and a alkyne or alkene moiety on the first strand.
[0262] Embodiment 14 provides the method of any of embodiments 7-13, wherein the crosslinked Ca2+ fluorophore is:
Figure imgf000075_0001
[0263] Embodiment 15 provides the method of any of embodiments 7-14, wherein the first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore is of formula:
Figure imgf000076_0001
[0264] Embodiment 16 provides the method of any of embodiments 7-11 , wherein the Ca2+ fluorophore is crosslinked to the first strand through a linker moiety stable under
physiological conditions.
[0265] Embodiment 17 provides the nucleic acid complex of any of embodiments 1-16, wherein the first label comprises Oregon green, FITC, or pHrhodo label.
[0266] Embodiment 18 provides the method of any of embodiments 1-16, wherein the intensity of the signal dependent on change in pH varies as a function of the conformation of the nucleic acid complex.
[0267] Embodiment 19 provides the method of embodiment 18, wherein the second single- stranded nucleic acid molecule comprising the sequence CnXCnYCnZCn, (SEQ ID NO: 13), wherein C is cytosine; X, Y, and Z are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
[0268] Embodiment 20 provides the method of embodiment 19, wherein each of X, Y, and Z is independently AA or TAA.
[0269] Embodiment 21 provides the method of embodiment 18, wherein the second single- stranded nucleic acid molecule comprising the sequence CnXCn, wherein C is cytosine; X and Y are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
[0270] Embodiment 22 provides the method of embodiment 21 , wherein each of X and Y is independently AA or TAA.
[0271] Embodiment 23 provides the method of any of embodiments 19-22, wherein each n is independently 3, 4, or 7. [0272] Embodiment 24 provides the method of any of embodiments 19-23, wherein the modification is selected from one or more of a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group.
[0273] Embodiment 25 provides the method of any of embodiments 19-23, wherein the modification is a methyl or bromo group.
[0274] Embodiment 26 provides the method of any of embodiments 19-23, wherein the modification is at the 5' position of the cytosine.
[0275] Embodiment 27 provides the method of any of embodiments 1-26, wherein the nucleic acid further comprises a second label conjugated to the first single-stranded nucleic acid or the second single-stranded nucleic acid.
[0276] Embodiment 28 provides the method of embodiment 27, wherein the intensity of the signal varies as a function of at least one of the distance between the first label and second label and the relative orientation of the first label and second label.
[0277] Embodiment 29 provides the method of embodiment 27, wherein the first label and second label comprise a donor and acceptor pair.
[0278] Embodiment 30 provides the method of any of embodiments 1-29, wherein the signal is measured using a FRET technique.
[0279] Embodiment 31 provides the method of any of embodiments 1-30, wherein the signal is measured at 2 different wavelengths.
[0280] Embodiment 32 provides the method of any of embodiments 27-30, wherein the signal is measured at 4 different wavelengths.
[0281] Embodiment 33 provides the method of any of embodiments 27-32, wherein the Ca2+ concentration is determined from the measured signal from the Ca2+ fluorophore with the measured signal from the second label.
[0282] Embodiment 34 provides the method of any of embodiments 27-32, wherein the CF concentration is determined from the measured signal from the CF fluorophore with the measured signal from the first label or the second label.
[0283] Embodiment 35 provides the method of any of embodiments 1-34, wherein the first single-stranded nucleic acid molecule and second single-stranded nucleic acid form an i- motif under acidic conditions.
[0284] Embodiment 36 provides the method of any of embodiments 1-34, wherein the second single-stranded nucleic acid is capable of forming an intramolecular complex comprising two parallel-stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
[0285] Embodiment 37 provides the method of any of embodiments 1-36, wherein the nucleic acid complex further comprises a third single-stranded nucleic acid molecule that is partially complementary to the first single-stranded molecule.
[0286] Embodiment 38 provides the method of embodiment 37, wherein the second single- stranded nucleic acid molecule comprising the sequence CnXCnYCnZCn, (SEQ ID NO: 13), wherein C is cytosine; X, Y, and Z are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
[0287] Embodiment 39 provides the method of embodiment 38, wherein each of X, Y, and Z is independently AA or TAA.
[0288] Embodiment 40 provides the method of embodiment 37, wherein the third single- stranded nucleic acid molecule comprising the sequence CnXCn, wherein C is cytosine; X and Y are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
[0289] Embodiment 41 provides the method of embodiment 37, wherein each of X and Y are independently AA.
[0290] Embodiment 42 provides the method of embodiment 37 or 04, wherein each n is independently 3, 4, or 7.
[0291] Embodiment 43 provides the method of any of embodiments 37-42, wherein the modification is selected from one or more of a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group.
[0292] Embodiment 44 provides the method of any of embodiments 37-42, wherein the modification is a methyl or bromo group.
[0293] Embodiment 45 provides the method of any of embodiments 37-44, wherein the modification is at the 5' position of the cytosine.
[0294] Embodiment 46 provides the method of any of embodiments 37-45, wherein the first label is on the second single-stranded nucleic acid.
[0295] Embodiment 47 provides the method of any of embodiments 37-46, wherein the nucleic acid complex further comprises the second label conjugated to the third single- stranded nucleic acid. [0296] Embodiment 48 provides the method of any of embodiments 37-47, wherein the second single-stranded nucleic acid and third single-stranded nucleic acid form an i-motif under acidic conditions.
[0297] Embodiment 49 provides the method of any of embodiments 37-47, wherein the second single-stranded nucleic acid or the third single-stranded nucleic acid is capable of forming an intramolecular complex comprising two parallel-stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
[0298] Embodiment 50 provides the method of any of embodiments 27-49, wherein the first label and the second label are independently selected from the group consisting of an Atto dye, an Alexa Flour® dye, a Cy® dye, and a BODIPY dye.
[0299] Embodiment 51 provides the method of any of embodiments 27-50, wherein the first label and the second label comprise a donor fluorophore and an acceptor quencher.
[0300] Embodiment 52 provides the method of embodiment 51 , wherein the donor fluorophore and an acceptor quencher pair are: FITC and TRITC, or Cy3 and Cy5, or Alexa- 488 and Alexa-647.
[0301] Embodiment 53 provides the method of any of embodiments 1-52, wherein the nucleic acid complex further comprises a targeting moiety.
[0302] Embodiment 54 provides the method of embodiment 53, wherein the targeting moiety is a nucleic acid sequence.
[0303] Embodiment 55 provides the method of embodiment 53, wherein the targeting moiety has a cognate artificial protein receptor.
[0304] Embodiment 56 provides the method of any of embodiments 53-55, wherein the targeting moiety is encoded on the same nucleic acid strand as the first single-stranded nucleic acid molecule, the second single-stranded nucleic acid molecule, the third single- stranded nucleic acid molecule, or any combination thereof.
[0305] Embodiment 57 provides the method of any of embodiments 53-55, wherein the targeting moiety is selected from an aptamer, a duplex domain targeted to an artificial protein receptor, a nucleic acid sequence that binds an anionic-ligand binding receptor, and an endocytic ligand.
[0306] Embodiment 58 provides the method of any of embodiments 53-55, wherein the targeting moiety comprises a peptide directly or indirectly conjugated to the nucleic acid molecule. [0307] Embodiment 59 provides the method of any of embodiments 53-55, wherein the targeting moiety comprises one or more of a fusogenic peptide, a membrane-permeabilizing peptide, a sub-cellular localization sequence, or a cell-receptor ligand.
[0308] Embodiment 60 provides the method of embodiment 59, wherein the sub-cellular localization sequence targets the nucleic acid complex to a region of a cell where spatial localization of a targeted protein is present.
[0309] Embodiment 61 provides the method of embodiment 60, wherein the sub-cellular localization sequence targets the nucleic acid complex to a region of the cell selected from the group consisting of: the cytosol, the endoplasmic reticulum, the mitochondrial matrix, the chloroplast lumen, the medial trans-Golgi cistemae, the lumen of lysosome, the lumen of an endosome, the peroxisome, the nucleus, and a specific spatial location on the plasma membrane.
[0310] Embodiment 62 provides the method of any of embodiments 1-61 , wherein the first and/or second single-stranded nucleic acid molecule is less than 200 nucleotides; or less than 100 nucleotides; or less than 50 nucleotides.
[0311] Embodiment 63 provides the method of any of embodiments 1-62, wherein the determined Ca2+ concentration is in a range of 10 nM to 10 mM.
[0312] Embodiment 64 provides the method of any of embodiments 1-62, wherein the determined Ca2+ concentration is in a range of 10 nM to 1 mM.
[0313] Embodiment 65 provides the method of any of embodiments 1-62, wherein the determined Ca2+ concentration is in a range of 1 pM to 10 mM.
[0314] Embodiment 66 provides the method of any of embodiments 1-62, wherein the determined Cl- concentration is in a range of 10 nM to 10 mM.
[0315] Embodiment 67 provides the method of any of embodiments 1-62, wherein the determined Cl- concentration is in a range of 10 nM to 1 pM.
[0316] Embodiment 68 provides the method of any of embodiments 1-62, wherein the determined Cl- concentration is in a range of 1 pM to 10 mM.
[0317] Embodiment 69 provides the method of any of embodiments 1-68, wherein the determined pH is less than pH 5.5.
[0318] Embodiment 70 provides the method of any of embodiments 1-68, wherein the determined pH is more than pH 7.0.
[0319] Embodiment 71 provides a nucleic acid complex comprising: a first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore or a CF fluorophore crosslinked to the first strand; and
a second single-stranded nucleic acid molecule that is partially or fully
complementary to the first single-stranded molecule,
wherein the nucleic acid complex further comprises a first label conjugated to the first single-stranded nucleic acid molecule or the second single-stranded nucleic acid molecule and the first label is capable of producing a signal.
[0320] Embodiment 72 provides the method of embodiment 71 , wherein the nucleic acid complex comprises the Cl- fluorophore crosslinked to the first strand.
[0321] Embodiment 73 provides the method of embodiment 72, wherein the Cl- fluorophore comprises 10,10'-bis[3-carboxypropyl]-9,9'-biacridinium dinitrate.
[0322] Embodiment 74 provides the method of embodiment 71 , wherein the nucleic acid complex comprises the Ca2+ fluorophore crosslinked to the first strand.
[0323] Embodiment 75 provides the nucleic acid complex of embodiment 74, wherein the Ca2+ fluorophore is a single wavelength indicator.
[0324] Embodiment 76 provides the nucleic acid complex of embodiment 74, wherein the Ca2+ fluorophore comprises Rhod-5F, XRhod-5F, Rhod-FF, XRhod-FF, Oregon Green 488 BAPTA-6F, Fluo 5F, Fluo 4FF, Oregon Green BAPTA-5N, Fluo-5 N, or Mag-Fluo-4 indicator.
[0325] Embodiment 77 provides the nucleic acid complex of embodiment 74, wherein the Ca2+ fluorophore comprises Rhod-5F, XRhod-5F, or Rhod-FF indicator.
[0326] Embodiment 78 provides the nucleic acid complex of embodiment 74, wherein the Ca2+ fluorophore comprises Rhod-5F indicator.
[0327] Embodiment 79 provides the nucleic acid complex of any of embodiments 74-78, wherein the Ca2+ fluorophore further comprises a linker moiety configured to form the triazole, thioether, or alkenyl sulfide group through a reaction of an azide or thiol moiety on the Ca2+ fluorophore and a alkyne or alkene moiety on the first strand.
[0328] Embodiment 80 provides the nucleic acid complex of any of embodiments 74-79, wherein the crosslinked Ca2+ fluorophore is:
Figure imgf000082_0001
[0329] Embodiment 81 provides the nucleic acid complex of any of embodiments 74-80, wherein the first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore is of formula:
Figure imgf000082_0002
, wherein R is a linker.
[0330] Embodiment 82 provides the nucleic acid complex of any of embodiments 71-81 , wherein the first label comprises Oregon green, FITC, or pHrhodo label.
[0331] Embodiment 83 provides the nucleic acid complex of any of embodiments 71-81 , wherein the second single-stranded nucleic acid molecule comprising the sequence CnXCn, wherein C is cytosine; X and Y are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
[0332] Embodiment 84 provides the nucleic acid complex of embodiment 83, wherein each of X and Y are independently AA or TAA.
[0333] Embodiment 85 provides the nucleic acid complex of any of embodiments 71-81 , wherein the second single-stranded nucleic acid molecule comprising the sequence CnXCnYCnZCn, (SEQ ID NO: 13), wherein C is cytosine; X, Y, and Z are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
[0334] Embodiment 86 provides the nucleic acid complex of embodiment 85, wherein each of X, Y, and Z are independently AA or TAA.
[0335] Embodiment 87 provides the nucleic acid complex of any of embodiments 83-86, wherein each n is independently 3, 4, or 7.
[0336] Embodiment 88 provides the nucleic acid complex of any of embodiments 83-87, wherein the modification is selected from one or more of a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group.
[0337] Embodiment 89 provides the nucleic acid complex of any of embodiments 83-87, wherein the modification is a methyl or bromo group.
[0338] Embodiment 90 provides the nucleic acid complex of any of embodiments 83-89, wherein the modification is at the 5' position of the cytosine.
[0339] Embodiment 91 provides the nucleic acid complex of any of embodiments 83-90, further comprising a second label capable of producing a signal conjugated to the first single-stranded nucleic acid or the second single-stranded nucleic acid.
[0340] Embodiment 92 provides the nucleic acid complex of embodiment 91 , wherein the first label and second label comprise a donor and acceptor pair.
[0341] Embodiment 93 provides the nucleic acid complex of any of embodiments 83-92, wherein the first single-stranded nucleic acid molecule and second single-stranded nucleic acid are configured to form an i-motif under acidic conditions.
[0342] Embodiment 94 provides the nucleic acid complex of any of embodiments 83-92, wherein the second single-stranded nucleic acid is configured to form an intramolecular complex comprising two parallel-stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
[0343] Embodiment 95 provides the nucleic acid complex of any of embodiments 83-94 further comprising a third single-stranded nucleic acid molecule that is partially
complementary to the first single-stranded molecule.
[0344] Embodiment 96 provides the nucleic acid complex of embodiment 95, wherein the third single-stranded nucleic acid molecule comprises the sequence CnXCnYCnZCn, (SEQ ID NO: 13), wherein C is cytosine; X, Y, and Z are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
[0345] Embodiment 97 provides the nucleic acid complex of embodiment 96, wherein each of X, Y, and Z are independently AA or TAA.
[0346] Embodiment 98 provides the nucleic acid complex of embodiment 95, wherein the third single-stranded nucleic acid molecule comprises the sequence CnXCn, wherein C is cytosine; X and Y are each one or more of adenine, thymine, guanine, or combinations thereof; and n is greater than or equal to 2; and wherein at least 2 cytosine residues are modified.
[0347] Embodiment 99 provides the nucleic acid complex of embodiment 98, wherein each of X and Y are independently AA.
[0348] Embodiment 100 provides the nucleic acid complex of embodiment 96-99, wherein each n is independently 3, 4, or 7.
[0349] Embodiment 101 provides the nucleic acid complex of any of embodiments 96-100, wherein the modification is selected from one or more of a methyl, fluoro, bromo, hydroxymethyl, formyl, or acetyl group.
[0350] Embodiment 102 provides the nucleic acid complex of any of embodiments 96-100, wherein the modification is a methyl or bromo group.
[0351] Embodiment 103 provides the nucleic acid complex of any of embodiments 96-102, wherein the modification is at the 5' position of the cytosine.
[0352] Embodiment 104 provides the nucleic acid complex of any of embodiments 96-103, wherein the first label is on the second single-stranded nucleic acid.
[0353] Embodiment 105 provides the nucleic acid complex of any of embodiments 96-104, wherein the nucleic acid complex further comprises the second label conjugated to the third single-stranded nucleic acid.
[0354] Embodiment 106 provides the nucleic acid complex of any of embodiments 96-105, wherein the second single-stranded nucleic acid and third single-stranded nucleic acid are configured to form an i-motif under acidic conditions.
[0355] Embodiment 107 provides the nucleic acid complex of any of embodiments 96-105, wherein the second single-stranded nucleic acid or the third single-stranded nucleic acid is configured to form an intramolecular complex comprising two parallel-stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions. [0356] Embodiment 108 provides the nucleic acid complex of any of embodiments 71-107, wherein the first label and the second label are independently selected from the group consisting of an Atto dye, an Alexa Flour® dye, a Cy® dye, and a BODIPY dye.
[0357] Embodiment 109 provides the nucleic acid complex of any of embodiments 91-108, wherein the first label and the second label comprise a donor fluorophore and an acceptor quencher.
[0358] Embodiment 110 provides the nucleic acid complex of embodiment 109, wherein the donor fluorophore and an acceptor quencher pair are: FITC and TRITC, or Cy3 and Cy5, or Alexa-488 and Alexa-647.
[0359] Embodiment 111 provides the nucleic acid complex of any of embodiments 71-107, wherein the nucleic acid complex further comprises a targeting moiety.
[0360] Embodiment 112 provides the nucleic acid complex of embodiment 111 , wherein the targeting moiety is a nucleic acid sequence.
[0361] Embodiment 113 provides the nucleic acid complex of embodiment 111 , wherein the targeting moiety has a cognate artificial protein receptor.
[0362] Embodiment 114 provides the nucleic acid complex of any of embodiments 111-113, wherein the targeting moiety is encoded on the same nucleic acid strand as the first single- stranded nucleic acid molecule, the second single-stranded nucleic acid molecule, the third single-stranded nucleic acid molecule, or any combination thereof.
[0363] Embodiment 115 provides the nucleic acid complex of any of embodiments 111-113, wherein the targeting moiety is selected from an aptamer, a duplex domain targeted to an artificial protein receptor, a nucleic acid sequence that binds an anionic-ligand binding receptor, and an endocytic ligand.
[0364] Embodiment 116 provides the nucleic acid complex of embodiment 115, wherein the targeting moiety comprises a peptide directly or indirectly conjugated to the nucleic acid molecule.
[0365] Embodiment 117 provides the nucleic acid complex of embodiment 115, wherein the targeting moiety comprises one or more of a fusogenic peptide, a membrane-permeabilizing peptide, a sub-cellular localization sequence, or a cell-receptor ligand.
[0366] Embodiment 118 provides the nucleic acid complex of embodiment 117, wherein the sub-cellular localization sequence targets the nucleic acid complex to a region of a cell where spatial localization of a targeted protein is present. [0367] Embodiment 119 provides the nucleic acid complex of embodiment 1 18, wherein the sub-cellular localization sequence targets the nucleic acid complex to a region of the cell selected from the group consisting of: the cytosol, the endoplasmic reticulum, the mitochondrial matrix, the chloroplast lumen, the medial trans-Golgi cistemae, the lumen of lysosome, the lumen of an endosome, the peroxisome, the nucleus, and a specific spatial location on the plasma membrane.
[0368] Embodiment 120 provides the nucleic acid complex of embodiment 71 , wherein the first strain has the sequence 5’-the Ca2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and
the second strain has the sequence 5’-the first label-CCC CTA ACC CCT AAC CCC TAA CCC CAT ATA TAT CCT AGA ACG ACA GAC AAA CAG TGA GTC-3’(SEQ ID NO:08).
[0369] Embodiment 121 provides the nucleic acid complex of embodiment 71 , wherein the first strain has the sequence 5’-TTA TAG GAT CCT GCG GTC GG/the Ca2+ fluorophore/ GGC ACC AGG CGT AAA ATG TA-3’(SEQ ID NO:09);
the second strain has the sequence: 5’-the first label-CCC CAA CCC CAA TAC ATT TTA CGC CTG GTG CC-3’ (SEQ ID NO: 10); and
the third strain has the sequence: 5’-CCG ACC GCA GGA TCC TAT AAA ACC CCA ACC CC-the second label-3 (SEQ ID NO: 1 1).
[0370] Embodiment 122 provides the nucleic acid complex of embodiment 71 , wherein the first strain has the sequence 5’-the Ca2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and
the second strain has the sequence 5'- the first label - AT AAC ACA TAA CAC ATAACAAAA TAT ATA TCC TAG AAC GAC AGA CAA ACA GTG AGT C - 3' (SEQ ID NO: 12).
[0371] Embodiment 123 provides the nucleic acid complex of embodiment 71 , wherein the first strain has the sequence 5’-the Cl- fluorophore-ATC AAC ACT GCA-Lys-COOH
(SEQ ID NO:22);
the second strain has the sequence 5'-TAT TGT GTA TTG TGT ATT GTT TTA TAT AT /the first label/ A TAG GAT CTT GCT GTC TGG TGT GCA GTG TTG AT-3'(SEQ ID NO:23); and
the third strain has the sequence: 5’-CAC CAG ACA GCA AGA TCC TAT ATA TAT ACC CCA ATC CCC AAT CCC CAA TCC CC-the second label-3' (SEQ ID NO:24).
[0372] Embodiment 124 provides the method of embodiment 1 , wherein the first strain has the sequence 5’-the Ca2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and
the second strain has the sequence 5’-the first label-CCC CTA ACC CCT AAC CCC TAA CCC CAT ATA TAT CCT AGA ACG ACA GAC AAA CAG TGA GTC-3’(SEQ ID NO:08).
[0373] Embodiment 125 provides the method of embodiment 1 , wherein
the first strain has the sequence 5’-TTA TAG GAT CCT GCG GTC GG/the Ca2+ fluorophore/ GGC ACC AGG CGT AAA ATG TA-3’(SEQ ID NO:09);
the second strain has the sequence: 5’-the first label-CCC CAA CCC CAA TAC ATT TTA CGC CTG GTG CC-3’ (SEQ ID NO: 10); and
the third strain has the sequence: 5’-CCG ACC GCA GGA TCC TAT AAA ACC CCA ACC CC-the second label-3 (SEQ ID NO: 11).
[0374] Embodiment 126 provides the method of embodiment 1 , wherein
the first strain has the sequence 5’-the Ca2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT-3’ (SEQ ID NO:07); and
the second strain has the sequence 5'- the first label - AT AAC ACA TAA CAC ATAACAAAA TAT ATA TCC TAG AAC GAC AGA CAA ACA GTG AGT C - 3' (SEQ ID NO: 12).
[0375] Embodiment 127 provides the method of embodiment 1 , wherein
the first strain has the sequence 5’-the Cl- fluorophore-ATC AAC ACT GCA-Lys-COOH
(SEQ ID NO:22);
the second strain has the sequence 5'-TAT TGT GTA TTG TGT ATT GTT TTA TAT AT /the first label/ A TAG GAT CTT GCT GTC TGG TGT GCA GTG TTG AT-3'(SEQ ID NO:23); and
the third strain has the sequence: 5’-CAC CAG ACA GCA AGA TCC TAT ATA TAT ACC CCA ATC CCC AAT CCC CAA TCC CC-the second label-3' (SEQ ID NO:24).
[0376] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be incorporated within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated herein by reference for all purposes.

Claims

What is claimed is:
1. A method for simultaneously determining 1) pH, and 2) Ca2+ concentration or Cl- concentration in a sample comprising:
providing a nucleic acid complex comprising
a first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore or a Cl- fluorophore crosslinked to the first strand; and
a second single-stranded nucleic acid molecule that is partially or fully complementary to the first single-stranded molecule,
wherein the nucleic acid complex further comprises a first label conjugated to the first single-stranded nucleic acid molecule or the second single- stranded nucleic acid molecule and the first label is capable of producing a signal, wherein the intensity of the signal is dependent on change in pH; measuring the intensity of the signal; and
determining the pH, and Ca2+ or Cl- concentration from the measured signal.
2. The method of claim 1 , wherein determining is in early endosome, late endosome, plasma membrane, lysosome, autophagolysosome, recycling endosome, cis Golgi network, trans Golgi network, endoplasmic reticulum, peroxisomes, or secretory vesicles.
3. The method of claim 1 or 2, wherein the nucleic acid complex comprises the Cl- fluorophore crosslinked to the first strand.
4 The method of claim 3, wherein the Cl- fluorophore comprises 10, 10'-bis[3- carboxypropyl]-9,9'-biacridinium dinitrate.
5. The method of claim 1 or 2, wherein the nucleic acid complex comprises the Ca2+ fluorophore crosslinked to the first strand.
6. The method of claim 5, wherein the Ca2+ fluorophore is a single wavelength indicator.
7. The method of claim 5, wherein the crosslinked Ca2+ fluorophore is:
Figure imgf000089_0001
wherein the first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore is of formula:
Figure imgf000089_0002
8. The method of any of claims 1-7, wherein the intensity of the signal dependent on change in pH varies as a function of the conformation of the nucleic acid complex.
9. The method of any of claims 1-8, wherein the nucleic acid further comprises a second label conjugated to the first single-stranded nucleic acid or the second single- stranded nucleic acid.
10. The method of claim 9, wherein the intensity of the signal varies as a function of at least one of the distance between the first label and second label and the relative orientation of the first label and second label.
11. The method of any of claims 1-10, wherein the first single-stranded nucleic acid molecule and second single-stranded nucleic acid form an i-motif under acidic conditions.
12. The method of any of claims 1-10, wherein the second single-stranded nucleic acid is capable of forming an intramolecular complex comprising two parallel-stranded C— HC+ base paired duplexes that are intercalated in an anti-parallel orientation at acidic conditions.
13. The method of any of claims 1-12, wherein the nucleic acid complex further comprises a third single-stranded nucleic acid molecule that is partially complementary to the first single-stranded molecule.
14. The method of any of claims 1-13, wherein the nucleic acid complex further comprises a targeting moiety.
15. The method of any of claims 1-14, wherein the first and/or second single-stranded nucleic acid molecule is less than 200 nucleotides; or less than 100 nucleotides; or less than 50 nucleotides.
16. The method of any of claims 1-15, wherein the determined Ca2+ concentration is in a range of 10 nM to 10 mM; or in a range of 10 nM to 1 mM; or in a range of 1 pM to 10 mM; or wherein the determined Cl- concentration is in a range of 10 nM to 10 mM; or in a range of 10 nM to 1 pM; or in a range of 1 pM to 10 mM.
17. The method of any of claims 1-16, wherein the determined pH is less than pH 5.5; or wherein the determined pH is more than pH 7.0.
18. A nucleic acid complex comprising:
a first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore or a Cl- fluorophore crosslinked to the first strand; and
a second single-stranded nucleic acid molecule that is partially or fully
complementary to the first single-stranded molecule,
wherein the nucleic acid complex further comprises a first label conjugated to the first single-stranded nucleic acid molecule or the second single-stranded nucleic acid molecule and the first label is capable of producing a signal.
19. The nucleic acid complex of claim 18, wherein the crosslinked Ca2+ fluorophore is:
Figure imgf000091_0001
wherein the first single-stranded nucleic acid molecule comprising a Ca2+ fluorophore is of formula:
Figure imgf000091_0002
wherein R is a linker.
20. The method of claim 1 or nucleic acid complex of claim 18, wherein
(a)
the first strain has the sequence 5’-the Ca2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT- 3’ (SEQ ID NO:07); and
the second strain has the sequence 5’-the first label-CCC CTA ACC CCT AAC CCC TAA CCC CAT ATA TAT CCT AGA ACG ACA GAC AAA CAG TGA GTC-3’(SEQ ID NO:08);
(b)
the first strain has the sequence 5’-TTA TAG GAT CCT GCG GTC GG/the Ca2+
fluorophore/ GGC ACC AGG CGT AAA ATG TA-3’(SEQ ID NO:09); the second strain has the sequence: 5’-the first label-CCC CAA CCC CAA TAC ATT TTA CGC CTG GTG CC-3’ (SEQ ID NO: 10); and
the third strain has the sequence: 5’-CCG ACC GCA GGA TCC TAT AAA ACC CCA ACC CC-the second label-3 (SEC ID NO:11); the first strain has the sequence 5’-the Ca2+ fluorophore-GAC TCA CTG TTT GTC TGT CGT TCT AGG ATA /the second label /AT ATT TTG TTA TGT GTT ATG TGT TAT- 3’ (SEC ID NO:07); and
the second strain has the sequence 5'- the first label - AT AAC ACA TAA CAC
ATAACAAAA TAT ATA TCC TAG AAC GAC AGA CAA ACA GTG AGT C - 3' (SEC ID NO:12); or the first strain has the sequence 5’-the Cl- fluorophore-ATC AAC ACT GCA-Lys-COOH (SEC ID NO:22);
the second strain has the sequence 5'-TAT TGT GTA TTG TGT ATT GTT TTA TAT AT /the first label/ A TAG GAT CTT GCT GTC TGG TGT GCA GTG TTG AT-3'(SEC ID NO:23); and
the third strain has the sequence: 5’-CAC CAG ACA GCA AGA TCC TAT ATA TAT ACC CCA ATC CCC AAT CCC CAA TCC CC-the second label-3' (SEC ID NO:24).
PCT/US2019/064053 2018-12-02 2019-12-02 Methods of determining ph and calcium or chloride concentration in samples WO2020117701A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2021531481A JP7307797B2 (en) 2018-12-02 2019-12-02 Method for Determining pH and Calcium or Chloride Ion Concentration in a Sample
US17/295,704 US20220011325A1 (en) 2018-12-02 2019-12-02 Methods of Determining pH and Calcium or Chloride Concentration in Samples
CN201980090973.XA CN113383075A (en) 2018-12-02 2019-12-02 Method for determining the pH and calcium concentration or chlorine concentration in a sample
EP19893431.7A EP3887519A4 (en) 2018-12-02 2019-12-02 Methods of determining ph and calcium or chloride concentration in samples

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862774314P 2018-12-02 2018-12-02
US62/774,314 2018-12-02

Publications (1)

Publication Number Publication Date
WO2020117701A1 true WO2020117701A1 (en) 2020-06-11

Family

ID=70973692

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/064053 WO2020117701A1 (en) 2018-12-02 2019-12-02 Methods of determining ph and calcium or chloride concentration in samples

Country Status (5)

Country Link
US (1) US20220011325A1 (en)
EP (1) EP3887519A4 (en)
JP (1) JP7307797B2 (en)
CN (1) CN113383075A (en)
WO (1) WO2020117701A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113252630A (en) * 2021-05-13 2021-08-13 西湖大学 Method and system for detecting concentration of predetermined ions in cells

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160376441A1 (en) * 2013-11-27 2016-12-29 Paris Sciences Et Lettres - Quartier Latin Fluorescent red emitting functionalizable calcium indicators
US20180245137A1 (en) * 2015-05-19 2018-08-30 The University Of Chicago Methods and compositions for determining ph

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9250252B2 (en) * 2009-05-29 2016-02-02 National Centre For Biological Sciences Intracellular pH sensor using nucleic acid assemblies
US10815379B2 (en) * 2015-03-27 2020-10-27 The University Of Tokyo pH sensitive fluorescent probe
CN108051492B (en) * 2017-12-21 2019-08-30 江南大学 A method of based on controllable self assembly forceps constructions Electrochemical Detection single-stranded target DNA

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160376441A1 (en) * 2013-11-27 2016-12-29 Paris Sciences Et Lettres - Quartier Latin Fluorescent red emitting functionalizable calcium indicators
US20180245137A1 (en) * 2015-05-19 2018-08-30 The University Of Chicago Methods and compositions for determining ph

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
SAHA ET AL.: "A pH-independent DNA nanodevice for quantifying chloride transport in organelles of living cells", NATURE NANOTECHNOLOGY, vol. 10, no. 7, July 2015 (2015-07-01), pages 645 - 652, XP055713940 *
See also references of EP3887519A4 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113252630A (en) * 2021-05-13 2021-08-13 西湖大学 Method and system for detecting concentration of predetermined ions in cells

Also Published As

Publication number Publication date
CN113383075A (en) 2021-09-10
JP2022510369A (en) 2022-01-26
EP3887519A1 (en) 2021-10-06
US20220011325A1 (en) 2022-01-13
EP3887519A4 (en) 2022-08-24
JP7307797B2 (en) 2023-07-12

Similar Documents

Publication Publication Date Title
US11898195B2 (en) Methods and compositions for determining pH
US20090274621A1 (en) Use of pyrene to carry non-peptide agents across the blood brain barrier
US8216850B2 (en) DNA-based molecular switches and uses thereof
US9250252B2 (en) Intracellular pH sensor using nucleic acid assemblies
US7807399B2 (en) Methods for detecting thiol-containing nucleotide diphosphates with thiol-reactive fluorescent reagents
US20120282169A1 (en) Detection and treatment of traumatic brain injury
US20220011325A1 (en) Methods of Determining pH and Calcium or Chloride Concentration in Samples
EP4090761B1 (en) A dna-based voltmeter for organelles
US20210096129A1 (en) Methods and Compositions for Spacial and Temporal Measurement of Catalytic Activity
US10443089B2 (en) Methods of multiplexing DNA sensors and localizing DNA sensor
CA2960193C (en) Methods for detecting amyloid beta oligomers
JP2007525967A (en) Modified molecular beacon
Class et al. Patent application title: DNA-based molecular switches and uses thereof Inventors: Yamuna Krishnan (Bangalore, IN) Yamuna Krishnan (Bangalore, IN) Saikat Chakraborty (Bangalore, IN)

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19893431

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021531481

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2019893431

Country of ref document: EP