WO2021198961A1 - Use of exosome-based delivery of nf-kb inhibitors - Google Patents

Use of exosome-based delivery of nf-kb inhibitors Download PDF

Info

Publication number
WO2021198961A1
WO2021198961A1 PCT/IB2021/052708 IB2021052708W WO2021198961A1 WO 2021198961 A1 WO2021198961 A1 WO 2021198961A1 IB 2021052708 W IB2021052708 W IB 2021052708W WO 2021198961 A1 WO2021198961 A1 WO 2021198961A1
Authority
WO
WIPO (PCT)
Prior art keywords
exo
exosome
photo
specific binding
inhibitor
Prior art date
Application number
PCT/IB2021/052708
Other languages
French (fr)
Inventor
Chulhee Choi
Kyungsun CHOI
Jae-Kwang YOO
Original Assignee
Ilias Biologics Inc.
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 Ilias Biologics Inc. filed Critical Ilias Biologics Inc.
Priority to KR1020217014385A priority Critical patent/KR20210124180A/en
Priority to AU2021247253A priority patent/AU2021247253A1/en
Priority to JP2022559527A priority patent/JP2023519700A/en
Priority to CA3173845A priority patent/CA3173845A1/en
Priority to CN202180024099.7A priority patent/CN115335079A/en
Priority to EP21781109.0A priority patent/EP4127136A4/en
Priority to KR1020227034736A priority patent/KR20220149781A/en
Priority to US17/227,359 priority patent/US20210299215A1/en
Publication of WO2021198961A1 publication Critical patent/WO2021198961A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5063Compounds of unknown constitution, e.g. material from plants or animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0686Kidney cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • the present disclosure is related to methods for treating acute kidney injury using exosome containing NF-kB inhibitors.
  • the present disclosure is related to methods for treating diseases induced by sepsis using exosome containing NF-KB inhibitor.
  • AKI not only contributes to short-term adverse outcomes, but the survivors may suffer from chronic kidney (CKD) and even end-stage renal disease (ESRD).
  • CKD chronic kidney
  • ESRD end-stage renal disease
  • NF-KB signaling blockades such as the renin-angiotensin-aldosterone system (RAAS) blockade or tumor necrosis factor-a (TNF- a) blocker
  • RAAS renin-angiotensin-aldosterone system
  • TNF- a tumor necrosis factor-a
  • TLRs Toll-like receptors
  • NF-KB tumor necrosis factor (TNF)-a and interleukin (IL)-l
  • TNF tumor necrosis factor
  • IL interleukin
  • NF-KB is involved in the apoptotic response, primarily through enhancing the transcription of antiapoptotic genes such as BC1-XL, A1 and A20.
  • Inflammation associated with NF-KB activation can thus be exacerbated by two interacting mechanisms, increased expression of pro-inflammatory mediators and prolongation of the life spans of cell populations such as neutrophils, which are activated to produce pro- inflammatory molecules and participate directly in acute inflammatory processes.
  • Sepsis is a systemic inflammatory syndrome caused by activation of the innate immune system due to acute microbial infection and continues to be the main cause of mortality in intensive care units.
  • diseases caused by sepsis that have vastly different symptoms, some of which are fatal.
  • the common treatment for Sepsis i.e., antibiotics
  • therapies there are no treatments available for clinical usage for diseases caused by sepsis. Thus, the development of effective alternative therapies is urgently needed.
  • NF-KB Although more than 700 inhibitors have been reported for NF-KB, no inhibitors have been approved as therapeutic agents to date. Moreover, various steroidal and nonsteroidal anti-inflammatory agents are known to block NF-KB, but their effects are not specific for inhibiting NF-KB.
  • a liposome is a synthetic vesicle with a phospholipid membrane that self-assembles into various sizes and shapes in an aqueous environment.
  • a PNP is a solid colloidal particle with the size between 10-1000 nm, and is made up of biodegradable polymers, in which the cargo proteins can be entrapped, encapsulated or attached to a nanoparticle matrix.
  • liposomes tend to fuse or aggregate with each other resulting in premature release of the liposome cargo over time.
  • PNPs may have better stability than liposomes, but their biocompatibility and long-term potential safety are still a concern.
  • encapsulating proteins to liposomes and PNPs requires generation of synthetic or recombinant proteins and ex vivo encapsulation processes, whereas EXPLOR technology requires generation and maintenance of stable cells that can produce cargo-loaded exosomes via natural exosome biogenesis.
  • exosomes have many desirable features of an ideal protein delivery system, such as biocompatibility, minimal or no inherent toxicity, long half-life in the circulation, and intrinsic ability to target tissues.
  • Exosomes have received significant attention as novel bio-carriers for gene/drug delivery.
  • Exosomes are extracellular vesicles (EVs) that play an important role in cell- to-cell communication, by transferring bioactive materials to recipient cells or affecting signaling pathways of target cells.
  • EVs extracellular vesicles
  • Exosomes are easier to store and exhibit greater stability than other bioactive agents.
  • Exosomes have a high capacity to overcome biological barriers, and can carry surface molecules targeting specific cell types, thus causing fewer off-target effects.
  • the present disclosure provides methods for treating acute kidney injury (AKI) in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-KB inhibitor to the subject.
  • AKI acute kidney injury
  • the present disclosure also provides methods for treating a disease caused by sepsis in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-KB inhibitor to the subject.
  • FIG. 1 Characterization of engineered exosomes produced from HEK293T [0011]
  • A Schematic diagram of DNA constructs used for producing super-repressor IKB- loaded exosomes (Exo-srlKB) (upper), and biogenesis of cargo protein-carrying exosomes using optically reversible protein- protein interactions, so called EXPLOR technology (lower).
  • B Representative TEM images of Exo-Naive and Exo-srlKB. Scale bar: 100 nm.
  • C Concentrations and size distributions of Exo-Naive (upper) and Exo-srlKB (lower) were determined by a Zetaview instrument.
  • FIG. 1 Renal protective effects of Exo-srlKB after kidney IRI
  • A Experimental scheme of kidney IRI surgery and exosome delivery. Each mouse group was intraperitoneally injected with 3 x 10 9 pn of Exo-naive or Exo-srlxB three times in a 1-h interval (total of 9 x 10 9 pn). Mice were sacrificed either 24 or 48 h after IRI surgery and serum and tissues were collected for further evaluation.
  • B-D Serum levels of BUN, creatinine, and NGAL among different groups depending on treatment type (Exo-naive vs. Exo-srlxB), drug delivery timing (Pre-treatment vs.
  • FIG. 4 The expression of pro-inflammatory cytokines/chemokines and adhesion molecules is negatively affected by EXO-SITKB treatment both locally and systemically.
  • (A) Whole kidney lysates from each experimental group were used for qRT-PCR, to measure expression levels of pro-inflammatory cytokines, including I1-1b, 11-6, Tnf-a, Ccl2,
  • FIG. 7 Exo-srlKB treatment modulates kidney immune cell populations following IR-induced AKI.
  • A Mice were sacrificed 24 h after surgery. KMNCs were enriched using enzymatic digestion, mechanical disruption, and Percoll density gradients. Total kidney cell numbers determined using those methods showed no statistical differences among the experimental groups.
  • B Graphical representation of flow cytometric analysis showed a higher percentage of kidney CD45 + cells after IRI among isolated kidney cells using enzymatic digestion, mechanical disruption, and Percoll density gradients. Exo-srlKB treatment alleviated the post-ischemic surge of renal immune cells.
  • Exo-srhcB Western blot analysis of NF-KB p65 expression using renal nuclear extracts from each experimental mouse group. Nuclear extracts were biochemically separated from cytoplasmic fractions, and NF-KB p65 and Lamin B1 expression was analyzed via western blotting.
  • Figure 9 (1). Generation and characterization of engineered exosomes [0019]
  • A Schematic of DNA constructs used for the production of super-repressor IKB- loaded exosome (Exo-srlKB) (upper). Schematic showing fusion proteins and their proposed activities (lower).
  • B Morphological characterization of Exo-Naive and Exo-srhcB through transmission electron microscopy (TEM)
  • C HEK293T cells that stably express mCherry or srhcB, and exosomes from these HEK293T cells, were lysed and immunoblotted against the indicated proteins.
  • TEM transmission electron microscopy
  • PBS phosphate-buffered saline
  • LPS Lipopolysaccharide
  • TNF tumor necrosis factor
  • IL-6 interleukin-6
  • IL-Ib interleukin-Ib
  • MIR macrophage inflammatory protein-Ib
  • C Representative images of cortical tubular cells in kidney sections from sham, CLP with PBS, CLP with Exo-Naive, and CLP with Exo-srhcB mice.
  • HEK293-NF-KB-luciferase cells (2x10 4 cells) were cultured either with Exo-Naive or with Exo-srlKB 2x10 5 particles. After 24 h, the cells were treated with 0.5ng/ml TNF-a for an additional 18 h. Luciferase activities were measured and normalized.
  • B Exo-srlKB dose- dependently repressed NF-KB activation in NF-KB-luciferase cells.
  • C THP-1 cells (5x10 5 cells) were stimulated with 1 pg/ml LPS and then treated with Exo-srhcBs 5x10 6 particles.
  • NTA nanoparticle tracking analysis
  • Figures 17-20 Protective effect of Exo- srli Bs on lung injury in LPS-induced sepsis [0027]
  • Figure 17 depicts H&E staining showing (1) Alveolar; (2) Capillary; (3) Neutrophil;
  • Figure 19 depicts the level of TNF-a in the plasma of Exo-srhcB-treated septic mice.
  • Figure 20 depicts H&E staining showing (1) central vein; and (2) infiltration of immune cells.
  • the present disclosure provides for EXPLOR technology (Yim et ah, Nature Communications 7, 12277 (2016), which was used to load srIi ⁇ B into exosomes and systemically deliver Exo-srl KB, in order to assess their effect on the course of ischemic AKI.
  • Control exosomes (Exo-Naive) were generated from intact HEK293T cells. The results confirmed that optogenetically engineered exosomes could be utilized as therapeutics, and that Exo-srlKB could alleviate IR-induced kidney damage through immunologic response modulation and apoptosis.
  • the term “about” means modifying, for example, lengths of nucleotide sequences, degrees of errors, dimensions, the quantity of an ingredient in a composition, concentrations, volumes, process temperature, process time, yields, flow rates, pressures, and like values, and ranges thereof, refers to variation in the numerical quantity that may occur, for example, through typical measuring and handling procedures used for making compounds, compositions, concentrates or use formulations; through inadvertent error in these procedures; through differences in the manufacture, source, or purity of starting materials or ingredients used to carry out the methods; and like considerations.
  • the term “about” also encompasses amounts that differ due to aging of, for example, a composition, formulation, or cell culture with a particular initial concentration or mixture, and amounts that differ due to mixing or processing a composition or formulation with a particular initial concentration or mixture. Whether modified by the term “about” the claims appended hereto include equivalents to these quantities.
  • the term “about” further may refer to a range of values that are similar to the stated reference value. In certain embodiments, the term “about” refers to a range of values that fall within 50, 25, 10, 9, 8,7, 6, 5,4, 3, 2, 1 percent or less of the stated reference value.
  • the term "subject” refers to a human or any non-human animal (e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate).
  • the immune response according to NF-KB signal transduction may be involved in AKI, and Exo-srl KB may exert an effect of improving kidney damage in the kidney damage caused by sepsis.
  • SrlxB was delivered to mice before and after IRI surgery using the EXPLOR technique, and the results were compared with the control group.
  • the Exo-srl kB-treated mice exhibited lower levels of serum BUN and creatinine compared to the control group, thereby exerting prophylactic and therapeutic effects from IR-induced AKI.
  • Exo-srlxB was found to significantly reduce the nuclear translocation and nuclear binding activity of NF-KB compared to the control group.
  • the NF-KB pathway may play an important role in the immune response process.
  • IL-Ib As a result of comparing the gene expression of Pan-inflammatory cytokines/chemokines in the Exo-srh ⁇ B treatment group and the control group, IL-Ib, IL-6, Significant reductions in a number of inflammatory mediators were identified, including CCL2, CCL5, CXCL2 and TNF-a.
  • flow cytometric analysis was conducted to compare whether NF-KB inhibitor treatment affects the renal immune cell population, and the frequency of multiple immune cell populations including neutrophils, monocytes/macrophages and T cells was observed to be significantly reduced.
  • IRI may induce an inflammatory cascade and oxidative stress, causing a cytokine storm, leading to apoptosis and structural damage to adjacent tissues.
  • the NF-KB signaling pathway may regulate cell survival and inflammation in a rapid response to IRI stimulation, specifically, high levels from damaged cells during the early stages of hypoxia/reperfusion.
  • endogenous factors such as mobility group box 1 (HMGB1), heat shock proteins (HSPs) and pathogen-/damage associated molecular patterns (P AMPs/DAMPs) are released.
  • HSPs heat shock proteins
  • P AMPs/DAMPs pathogen-/damage associated molecular patterns
  • TER Toll-like Receptor
  • PRP Pattern Recognition Receptor
  • TER and IL-1R share the same intracellular domain and phosphorylate the inhibitory protein IKB residue and activate IKB kinase (IKK), which ultimately leads to degradation of IKB by the proteasome.
  • IKK IKB kinase
  • This process allows a heterodimer (e.g., p50/p65) to move from the cytoplasm to the nucleus, which binds to DNA and promotes transcription of inflammatory mediators including TNF-a, IL-1, IL-6 and IL-8, which further accelerate NF-KB signaling.
  • This pro-inflammatory cascade may regulate the surrounding microenvironment by inducing apoptosis and promoting leukocyte migration/activation in renal duct cells exposed to IRI.
  • the ischemic AKI model of the present disclosure can also induce the activation of NF-KB signaling, which induces the expression of a number of inflammatory cytokines/chemokines genes, significantly affecting the renal immune cell population compared to the false surgical group.
  • NF-KB signaling which induces the expression of a number of inflammatory cytokines/chemokines genes, significantly affecting the renal immune cell population compared to the false surgical group.
  • Recent studies include TLR antagonists, cytokine antagonists, IKK complex antagonists, proteasome inhibitors, decoy oligo deoxynucleotides (ODNs) specific to specific NF-kB complexes, and ODNs that inhibit Rel protein translation in various organs. It shows the effect of blocking NF-KB in IRI by inhibiting NF-KB signaling using a variety of pharmacological inhibitors, including.
  • IkBa proteins having mutations at serine residues 32 and 36 are not phosphorylated and thus exist as multimers with NF-KB in the cytoplasm and are not degraded, which holds NF-KB nuclear translocation.
  • This non-degradable IkBa so-called super repressor IkBa (srlxB)
  • srlxB super repressor IkBa
  • Exo-srh ⁇ B regulates the process of AKI by down-regulating the production of pro-inflammatory mediators that can limit leukocyte migration/activation, and by inducing early apoptosis of inflammatory cells, leading to early termination of the inflammatory phase.
  • Exo-srlxB only affects the frequency of the immune cell population in the kidney and does not affect the frequency of the immune cell population in the spleen.
  • ExosrlicB blocks the outflow of splenic immune cells to the damaged kidney.
  • Exo-srlxB is shown to alleviate IR-induced AKI in mice by inhibiting the activity of NF-KB signal.
  • in vivo delivery of EXO-SHKB can improve the overall inflammatory state of the kidney by reducing the expression of pro- inflammatory genes and reducing the renal immune cell population including neutrophils, monocytes/macrophages and T cells.
  • the present disclosure relates to an exosome on which srlxB is mounted, and more particularly, to an exosome on which srlxB represented by the amino acid sequence of SEQ ID NO: 1 is mounted.
  • srlxB may be expressed as a cargo protein.
  • the AKI described herein is Ischemia-reperfusion (IR)-induced AKI, lipopolysaccharide (LPS)-induced AKI or sepsis-induced AKI but not limited thereto.
  • Ischemia-reperfusion (IR) injury (IRI) is a major cause of acute kidney injury (AKI).
  • Ischemia- reperfusion (IR)-induced acute kidney injury (AKI) is a relatively common but severe medical condition that can happen when a subject suffers from systemic hypoperfusion that is followed by restoration of blood flow and re- oxygenation. The most common clinical examples include patients undergoing renal transplant or cardiac surgery and patients with sepsis, trauma, or myocardial infarction.
  • Renal IR injury is associated with a high morbidity/mortality and is known to induce dysfunction in other distant organs, including the heart, lung, and liver.
  • IRI is associated with a high morbidity/mortality and is known to induce dysfunction in other distant organs, including the heart, lung, and liver.
  • ROS reactive oxygen species
  • Immunologic responses may significantly affect renal IRI and repair.
  • Nuclear factor (NF)-KB signaling which controls cytokine production and cell survival, is significantly involved in IR- induced AKI; its inhibition can ameliorate ischemic AKI.
  • Exo-srlxB the exosomal super-repressor inhibitor of NF-KB
  • Exo-srlxB decreased NF-KB activity in post-ischemic kidneys, leading to reduced apoptosis levels.
  • Post-ischemic kidneys showed decreased gene expression of pro- inflammatory cytokines and adhesion molecules with Exo-srlxB treatment as compared with the control.
  • Exo-srlxB treatment also significantly affected post-ischemic renal immune cell populations, lowering neutrophil, monocyte/macrophage, and T cell frequencies than those in the control.
  • the modulation of NF-KB signaling through exosomal delivery can be used as a novel therapeutic method for AKI, including IR-induced AKI.
  • the exosomes are engineered to contain a mutant form of the biological inhibitor of NF-KB, called super-repressor (SR) IKB (srlxB , SEQ ID NO:l) (N. Yim et al, Nature Communications 7, 12277 (2016).
  • SR super-repressor
  • EXPLOR optogenetically engineered exosome system
  • Exo-srlKB alleviates inflammatory responses in monocytic THP-1 cells and human umbilical vein endothelial cells ( HUVECs ) in response to lipopolysaccharide (LPS) or tumor necrosis factor ( TNF)-a .
  • LPS lipopolysaccharide
  • TNF tumor necrosis factor
  • the present disclosure provides a method for treating acute kidney injury (AKI) in a subject in need thereof, comprising administering an effective amount of a composition comprising an exosome comprising an NF-kB inhibitor to the subject.
  • the NF-KB inhibitor is selected from the group consisting of a NF-KB inhibitory protein, its fragment, and a mixture thereof.
  • compositions are physiologically acceptable water-based solutions, or suspensions, of exosomes.
  • the NF-KB inhibitor is selected from an NF-KB inhibitory drug, an NF-KB inhibitory protein or its fragment, or a mixture thereof.
  • the NF-KB inhibitor is selected from the group comprising super-repressor IKB, IkB-a, IkB-b, IkB-e, BCF-3, a mutant thereof, and a mixture thereof.
  • the exosomes of the present disclosure contain a mutant form of the biological inhibitor of NF-KB, called super repressor (SR) IKB (SAKB, SEQ ID NO:l).
  • SR super repressor
  • srhcB has Ser32 and Ser36 of IKB (SEQ ID NO: 1]
  • SEQ ID NO: 1 Homo sapiens, super-repressor-IkB (srhcB)
  • the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO: 1 or SEQ ID NO:2. In some embodiments, the NF- KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:l. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 86% sequence identity to SEQ ID NO:l. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 87% sequence identity to SEQ ID NO:l. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 88% sequence identity to SEQ ID NO: 1.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 89% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 91% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 92% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 93% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 94% sequence identity to SEQ ID NO: 1.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 95% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 96% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 97% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 98% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 99% sequence identity to SEQ ID NO: 1. In embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 1.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 86% sequence identity to SEQ ID NO:2. In some embodiments, the NF-kB inhibitor comprises an amino acid sequence having at least 87% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 88% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 89% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO:2.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 91% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 92% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 93% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 94% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 95% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 96% sequence identity to SEQ ID NO:2.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 97% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 98% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 99% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 2. [0053] In a certain embodiment of the present disclosure, the exosome further comprises a photo- specific binding protein.
  • the photo-specific binding protein comprises a first photo-specific binding protein and/or a second photo-specific binding protein, which interacts reversibly with each other upon irradiation.
  • the first photo- specific binding protein is conjugated to an exosome specific marker to form a first fusion protein (fusion protein I).
  • the second photo- specific binding protein is conjugated to the NF-KB inhibitory protein to form a second fusion protein (fusion protein II).
  • the fusion protein I and the fusion protein II are linked reversibly through the first photo-specific binding protein and the second photo-specific binding protein.
  • the first photo-specific binding protein is conjugated to the exosome specific marker to be located in the direction toward inside of the exosome.
  • the first photo-specific binding protein and the second photo-specific binding protein are selected from the group comprising CIB, CIBN, PhyB, PIF, FKF1, GIGANTEA, CRY or PHR, but not limited thereto.
  • the first photo-specific binding protein is CIB or CIBN and the second photo-specific binding protein is CRY or PHR; or the first photo-specific binding protein is CRY or PHR and the second photo-specific binding protein is CIB or CIBN.
  • the first photo-specific binding protein is PhyB and the second photo-specific bind protein is PIF or the first photo-specific binding protein is PIF and the second photo-specific binding protein is PhyB.
  • the first photo-specific binding protein is GIGANTEA and the second photo-specific binding protein is FKFl or the first photo-specific binding protein is FKFl and the second photo-specific binding protein is GIGANTEA.
  • the exosome specific marker is selected from the group comprising CD9, CD63, CD 81 or CD 82, but not limited thereto.
  • the exosome specific marker is CD9.
  • the exosome specific marker is CD63.
  • the exosome specific marker is CD81.
  • the exosome specific marker is CD82.
  • the exosome has a diameter of between about 50 nm and about 200 nm. In some embodiments, the exosome has a diameter of between about 50 nm and about 75 nm. In some embodiments, the exosome has a diameter of between about 75 nm and about 100 nm. In some embodiments, the exosome has a diameter of between about 100 nm and about
  • the exosome has a diameter of between about 125 nm and about
  • the exosome has a diameter of between about 150 nm and about
  • the exosome has a diameter of between about 175 nm and about
  • the exosome has a diameter of between about 50 nm and about 150 nm. In some embodiments, the exosome has a diameter of about 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150 or more and/or of about 75, 80, 85,
  • the present disclosure also provides sustained release of the therapeutically effective drugs, while conventional sustained effects required repeated dosing of the therapeutics.
  • the present disclosure also further provides pharmaceutical compositions containing extracellular vesicles (exosomes) carrying NF-KB inhibitors and a pharmaceutically acceptable carrier or carriers.
  • IRI IKB kinase
  • heterodimers translocate from the cytosol to the nucleus, where they promote the transcription of inflammatory mediators, including TNF-a, IL-1, IL-6, and IL-8, which in turn further promote NF-KB signaling.
  • This proinflammatory cascade modulates surrounding microenvironments, by inducing apoptosis in renal tubular cells subjected to IRI, and promoting leukocyte migration/activation.
  • the instant experimental ischemic AKI model could reproduce the activation of NF-KB signaling following renal IRI, leading to the increased expression of multiple inflammatory cytokines/chemokines and tissue apoptosis, which significantly affect renal immune cell populations.
  • NF-KB blockade on IRI, using selective pharmacologic inhibitors of NF-KB signaling, including TLR antagonists, cytokine antagonists (33), IKK complex antagonists, proteasome inhibitors, and decoy oligodeoxynucleotides specific to a particular NF-KB complex, in various organs.
  • srh ⁇ B is a non-degradable IkBa protein with mutations at serine residues 32 and 36. This protein prevents NF-KB nuclear translocation and subsequent NF-KB signaling.
  • srh ⁇ B has proven protective effects in the lung IRI model, as it decreases neutrophil infiltration and pulmonary edema, and is also known to have anti-tumor effects, because it decreases chemoresistance.
  • the effects of srh ⁇ B treatment in the course of kidney injury has not been fully evaluated.
  • Choi et al. recently showed the therapeutic benefits of srh ⁇ B treatment in the septic AKI model.
  • the below examples also demonstrate the protective effects of Exo-srlKB treatment in the renal IRI model, expanding the therapeutic potential of srh ⁇ B delivery through exosomes in various types of AKI.
  • Inflammatory process modulation has been a major therapeutic consideration in the field of ischemic AKI.
  • Splenectomy as well as the use of the anti-TNF-a agent, infliximab, showed protective effects in the experimental ischemic AKI model, by lowering inflammatory cytokine expression and macrophages/monocyte accumulation.
  • Inhibiting the migration of leukocytes and macrophages also preserved renal function and alleviated cell death the provided examples have shown that Exo-srlKB treatment downregulates the expression of pro- inflammatory cytokines/chemokines and adhesion molecules, reduces apoptosis, and alleviates multi-lineage immune cell accumulation in IR-injured kidneys.
  • Exosomes can have several benefits over the use of other types of vectors as conveyors.
  • EVs are naturally protected from degradation during circulation and are non- immunogenic when used autologously.
  • Exosomes can also overcome natural barriers such as the blood-brain barrier, through their intrinsic cell targeting properties. Additionally, exosomes use intrinsic mechanisms of recipient cells in the course of uptake, intracellular trafficking and the final delivery of cargoes to target cells.
  • EXPLOR technology made it possible to significantly increase intracellular levels of cargo proteins through controllable, reversible detachment from exosomes.
  • the below examples showed that efficient delivery of Exo-srlxB using EXPLOR technology could bring significant improvement in the renal outcome after experimental IRI.
  • Systemic Exo-srlxB treatment may only affect immune cell population frequencies in the kidney but not those in the spleen. This can be attributed to Exo-srl KB either selectively acting on renal resident immune cells alleviating proliferation or promoting apoptosis, or the systemic delivery of Exo-srlxB blocking the efflux of splenic immune cells into injured kidneys. Kidney resident immune cells and immune cells from lymphoid organs are known to have distinct origins, phenotypes, and functional characteristics. Further studies using chimeric mice or in vivo imaging would be able to differentiate resident renal immune cells from circulating immune cells, and provide more information about immune cell populations affected by systemic Exo-srlitB treatment.
  • exosome as a carrier to deliver a cargo of active therapeutic agents is a safe and efficient method for delivering srlxB into the IR-induced AKI model.
  • Exo-srlitB treatment alleviates IR-induced AKI in mice by down-regulating NF-KB signaling and ameliorating inflammation and apoptosis in the ischemic injured kidney.
  • the direct intracellular delivery of immunosuppressive proteins into target cells using exosomes is a promising therapeutic tool in IR-induced AKI and the therapeutic potential of Exo-srlxB in human kidney IRI should be explored further.
  • Sepsis which is accompanied by a large number of organ failures and excessive inflammatory reactions, has complex pathophysiological features, and it is not easy to develop a therapeutic agent that can effectively treat all symptoms. Sepsis is initiated through the strong activation of the innate immune system and is mediated by the activation of the pattern recognition receptor (PRR) by pathogens, leading to activation of the complement system, coagulation system and vascular endothelium and thus the difficulty to normal homeostasis.
  • PRR pattern recognition receptor
  • the effect of Exo-srlxB was capable of resolving the inflammatory response and reducing organ damage through inhibition of NF-KB activity in a sepsis mouse model.
  • srhcB is a non-degradable form of IkBa that prevents NF-KB from entering the nucleus and acting as a transcription factor.
  • a recent technology capable of mounting a functional protein on an exosome was utilized.
  • Exo-srlxB By delivering Exo-srlxB to the whole body in a sepsis mouse model (e.g., systemic delivery), the examples herein confirmed that exosomes are delivered to neutrophils and macrophages, which are key players of the sepsis inflammatory response.
  • Exo-srlxB treatment significantly reduced pro- inflammatory markers such as TNF-a, IF-Ib and IF-6, but did not decrease the anti-inflammatory cytokine IF- 10 Even when exosomes were injected 1 hour after EPS injection, a significant therapeutic effect of Exo-srlxB was observed.
  • the treatment with Exo-srlxB may also be applied to the early stages of sepsis before the transition to an immune suppression state.
  • Pro- inflammatory cytokines may play an important role in acute kidney injury (AKI) induced by sepsis.
  • AKI acute kidney injury
  • CEP surgery or LPS injection in the examples herein, as a result of histopathological examination, it was confirmed that the glomerular structure was destroyed, renal tubular epithelial cells were degenerated, and severe intracellular edema and congestion occurred in the renal tubules in the sepsis-induced group.
  • Exo-srlxB administration reduced the severity of lesions, kidney damage, and invasion of inflammatory cells.
  • Tissue infiltration of neutrophils may be a major process in sepsis.
  • TNF-a and IL-Ib are major mediators for the expression of chemokines such as MCP-1 and IL- 8, which are important chemoattractants for neutrophils and monocytes/macrophages during the inflammatory response. These chemokines initiate local invasion of monocytes/macrophages after activation.
  • Exo-srlKB was shown to attenuate the release of FPS- induced MCP-1 and IL-8, thereby inhibiting monocyte/macrophage invasion.
  • the spleen shows higher levels of neutrophils compared to other organs such as kidney and liver.
  • Pro-inflammatory cytokines may occur in the spleen and affect other organs. Most of the genes encoding pro-inflammatory cytokines may be under the control of NF-KB, one of the important transcription factors in the septic inflammatory response caused by LPS. Expression of the NF-KB response reporter protein according to the presence or absence of Exo-srhcB treatment was investigated in the examples herein, and the translocation of p65 was confirmed. Exo-srlKB inhibited NF-KB signal activation by blocking nuclear translocation of the NF-KB subunit p65.
  • exosomes may be used as a mechanism for therapeutic delivery of immunosuppressive proteins in a sepsis mouse model.
  • Exosomes may be excellent carriers capable of delivering srhcB into cells, providing a new option for the treatment of sepsis.
  • a method of directly delivering an immunosuppressive protein into a target cell in vivo has not been established before, but Exo-srhcB according to the present disclosure acts as an inhibitor of NF-KB, thereby inhibiting the expression of inflammation-related genes and directly inflammatory response. The examples herein shows it can be suppressed, thereby alleviating the pro-inflammatory cytokine storm of sepsis and the resulting organ damage.
  • NF-KB inhibitors with diverse chemical properties and mechanisms of action protect animals from septic lethality. Although more than 700 inhibitors of NF-KB have been reported, no NF-KB blocker has been approved for human use to date.
  • steroids and non- steroid anti-inflammatory drugs have been found to block NF-KB, but their effects are highly pleiotropic and lack specificity.
  • Novel therapeutic strategies aimed at specific inhibition of key elements in the NF-KB activation pathway have been under development in recent years, and expectations are high regarding their potential efficacy as sepsis treatments and treatments for one or more diseases caused by sepsis.
  • Genetic constructs expressing an engineered IkBa protein without sites for phosphorylation, super-repressor IKB (srlicB), have also been used.
  • IkBa mutation at specific phosphorylation sites (Ser32 and Ser36 replaced to Ala) results in a dominant active form of IkBa with a prolonged half-life. These srlxBs lead to a stable cytoplasmic pool of IkBa, thereby preventing nuclear NF-KB activation.
  • exosomes were utilized as a therapeutic conveyor to deliver srlxB to a therapeutic target.
  • Exosomes have been recognized as potent therapeutic vehicles for transferring various proteins and regulatory genes to target cells. They function as non-immunogenic nanovesicles, and can protect their cargo from serum proteases and immune responses. Loading of soluble proteins into exosomes has been enabled by a technology called EXPLOR, which exploits the natural exosome biogenesis process and reversible protein-protein interactions controlled by optogenetics.
  • HEK293T cell line was generated that stably expresses two recombinant proteins, CIBN-EGFP-CD9 and srlxB- mCherry-CRY2 ( Figure 1 A). Consistent with previous studies, EXPLOR technology was used to produce srlxB-loaded exosomes. The srlicB loading exosomes were designated as EXO-SHKB, and exosomes generated from intact HEK293T cells as Exo-Naive. By applying EXO-SAKB to sepsis models for therapeutic purposes, sepsis-induced organ injury was ameliorated and inhibited the secretion of pro-inflammatory cytokines, thereby improving the overall survival of sepsis patients.
  • the present disclosures provides for methods for treating a disease caused by sepsis in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-KB inhibitor to the subject.
  • the disease is selected from the group consisting of pneumonia, cytokine storm syndrome, respiratory distress syndrome, and organ failure but not limited thereto.
  • the disease is pneumonia.
  • the disease is cytokine storm syndrome.
  • the disease is respiratory distress syndrome.
  • the disease is organ failure.
  • the NF-KB inhibitor is selected from the group consisting of a NF-KB inhibitory protein, its fragment, and a mixture thereof.
  • the NF-KB inhibitor is selected from the group consisting of super-repressor (SR)-IKB (srlicB), IkB-a, IkB-b, IkB-e, BCL-3, a mutant thereof, and a mixture thereof.
  • the NF-KB inhibitor is super repressor (SR)-IKB (srlicB).
  • the composition of is administered via oral, transdermal, intraperitoneal, intravenous, intramuscular, subcutaneous, or mixed routes.
  • the composition is administered via oral routes.
  • the composition is administered via transdermal routes.
  • the composition is administered via intraperitoneal routes.
  • the composition is administered via intravenous routes.
  • the composition is administered via intramuscular routes.
  • the composition is administered via subcutaneous routes.
  • the composition is administered via mixed routes.
  • the method further comprises administering an anti-inflammatory agent to the subject.
  • the exosome comprising a NF-KB inhibitor further comprises a photo- specific binding protein.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:l or SEQ ID NO:2.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO: 1.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 86% sequence identity to SEQ ID NO: 1.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 87% sequence identity to SEQ ID NO: 1.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 88% sequence identity to SEQ ID NO: 1.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 89% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 91% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 92% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 93% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 94% sequence identity to SEQ ID NO: 1.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 95% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 96% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 97% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 98% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 99% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 1.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 86% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 87% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 88% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 89% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO:2.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 91% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 92% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 93% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 94% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 95% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 96% sequence identity to SEQ ID NO:2.
  • the NF-KB inhibitor comprises an amino acid sequence having at least 97% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 98% sequence identity to SEQ ID NO:2. In some embodiments, the NF-kB inhibitor comprises an amino acid sequence having at least 99% sequence identity to SEQ ID NO:2. In embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 2.
  • the photo-specific binding protein is a first photo-specific binding protein and/or a second photo-specific binding protein.
  • the first photo specific binding protein is conjugated to an exosome specific marker to form fusion protein I; and the second photo-specific binding protein is conjugated to the NF-KB inhibitor to form fusion protein II.
  • the fusion protein I and the fusion protein II are linked reversibly through the first photo-specific binding protein and the second photo-specific binding protein.
  • the first photo-specific binding protein is conjugated to an exosome specific marker to be located in the direction toward inside of the exosome.
  • the first photo-specific binding protein and the second photo specific binding protein are selected from the group consisting of CIB, CIBN, PhyB, PIF, FKF1, GIGANTEA, CRY and PHR.
  • the first photo-specific binding protein is CIB and the second photo-specific binding protein is CRY.
  • the first photo specific binding protein is CIBN and the second photo-specific binding protein is CRY.
  • the first photo-specific binding protein is CIB and the second photo-specific binding protein is PHR.
  • the first photo-specific binding protein is CIBN and the second photo-specific binding protein is PHR.
  • the first photo-specific binding protein is CRY or PHR and the second photo-specific binding protein is CIB or CIBN. In some embodiments, the first photo specific binding protein is CRY and the second photo-specific binding protein is CIB. In some embodiments, the first photo-specific binding protein is CRY and the second photo-specific binding protein is CIBN. In some embodiments, the first photo-specific binding protein is PHR and the second photo-specific binding protein is CIB. In some embodiments, the first photo specific binding protein is PHR and the second photo-specific binding protein is CIBN.
  • the first photo-specific binding protein is PhyB and the second photo- specific binding protein is PIF. In embodiments, the first photo-specific binding protein is PIF and the second photo-specific binding protein is PhyB. [0089] In some embodiments, the first photo-specific binding protein is GIGANTEA and the second photo-specific binding protein is FKF1. In embodiments, the first photo-specific binding protein is FKF1 and the second photo- specific binding protein is GIGANTEA.
  • the exosome specific marker is selected from the group consisting of CD9, CD63, CD81 and CD82. In some embodiments, the exosome specific marker is CD9. In some embodiments, the exosome specific marker is CD63. In some embodiments, the exosome specific marker is CD81. In some embodiments, the exosome specific marker is CD82. [0091] In some embodiments, the exosome has a diameter of between about 50 nm and about 200 nm. In some embodiments, the exosome has a diameter of between about 50 nm and about 75 nm. In some embodiments, the exosome has a diameter of between about 75 nm and about 100 nm. In some embodiments, the exosome has a diameter of between about 100 nm and about
  • the exosome has a diameter of between about 125 nm and about
  • the exosome has a diameter of between about 150 nm and about
  • the exosome has a diameter of between about 175 nm and about
  • the exosome has a diameter of between about 50 nm and about 150 nm. In some embodiments, the exosome has a diameter of about 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150 or more and/or of about 75, 80, 85,
  • the composition is a pharmaceutical composition further comprising a physiologically acceptable carrier or carriers.
  • Exosomes received significant attention as novel bio-carriers for gene/drug delivery.
  • Exosomes are extracellular vesicles (EVs) that play an important role in cell-to-cell communication, by transferring bioactive materials to recipient cells or affecting signaling pathways of target cells.
  • EVs extracellular vesicles
  • Exosomes are easier to store and exhibit greater stability.
  • Exosomes have a high capacity to overcome biological barriers, and can carry surface molecules targeting specific cell types, thus causing fewer off-target effects.
  • EXPLOR Exosomes for protein loading via optically reversible protein-protein interactions
  • Yim etal significant advancements in the production efficiency and biological compatibility of exosomes were achieved.
  • This EXPLOR technology has been recently adopted in the experimental sepsis model by Choi et al. ; they delivered exosomes containing the super-repressor IKB (srlxB) to mice. This was a non-degradable form of inhibitor of kappa B (IKB) that prevented the nuclear translocation of NF-KB.
  • IKB inhibitor of kappa B
  • mice Male C57BL/6J mice were bred under specific pathogen-free conditions at the central animal facility of the Yonsei Medical Center. 6-7-week-old male mice were used for all experiments. Additionally, C56BL/6, C57BL/6N, BALB/c mouse were purchased from Orientbio (Seong-Nam, Republic of Korea). LysM GFP/+ mice intrinsically expressing GFP in neutrophils and macrophages were generously provided by Dr. M. Kim (University of Rochester, Rochester, NY, USA).
  • Exosome isolation The HEK293T cell line, which expresses two recombinant proteins, CIBN-EGFP-CD9 and srlKB-mCherry-CRY2, were used to produce srhcB-loaded exosomes.
  • Intact HEK293T cells were used for producing Exo-Naive. Stable cells were seeded into T175 flasks for 24 h at 37 °C. After removing the medium, cells were washed and resuspended with exosome-depleted medium. Cells were then exposed to continuous blue light illumination from a 460-nm light emitting diode in a CO2 incubator.
  • Exo-srhcB the stable cells were seeded into T175 flasks. After 1 day, the medium was carefully removed, the cells were rinsed with PBS, and exosome-depleted medium was added. Then, the cells were exposed to continuous blue light illumination from a 460-nm light emitting diode in a CO2 incubator.
  • the cell culture supernatant was harvested and centrifuged at 1,000 g for 15 min to remove cells and cell debris, and then filtered through a 0.22-um polyethersulfone (PES) filter to remove large particles.
  • PES polyethersulfone
  • the exosomes were isolated using molecular weight cutoff (MWCO)- based membrane filtration. The isolated exosomes were purified through SEC.
  • Nanoparticle tracking analysis The particle number and size distribution of EVs were measured by NTA using the Zetaview instrument (PMX120, Particle Metrix, Germany) with an embedded 488 nm laser. Samples were diluted (serum 1:100-1:10000) in particle-free PBS according to the manufacturer’s instructions. Samples were analyzed under constant flow conditions at 25 °C with a camera level of 78 and shutter of 80. Size distributions determined by NTA corresponded to hydrodynamic diameters of EVs in the suspension. Particle number was calculated from the rate of Brownian motion and size was determined using the two-dimensional Stokes-Einstein equation based on the velocity of particle movement.
  • Certain samples were diluted in 0.2-mih filtered PBS between 1 : 100 and 1 : 10,000.
  • EV concentrations were measured based on counts of 50-200 particles per frame. For each measurement, two cycles of scanning at 11 cell positions were performed with the following settings: focus, autofocus; camera sensitivity for all samples, 78.0; shutter, 70; and cell temperature, 25°C. The EV concentration was expressed in particle numbers per ml (pn/ml).
  • TNF-a, IL-8 and MCP-1 levels in supernatants collected from THP-1 and HUYEC cultures were measured using commercial ELISA kits (R&D Systems, Minneapolis, MN, USA), and NF-KB activity in HUVECs was measured using the Trans AM NF-KB p65 kit (Active Motif, Carlsbad, CA, USA).
  • TNF-a, CCL-4/macrophage inflammatory protein (MIP)- 1 b, IL-6, and IL-1 b levels in mouse plasma collected through cardiac puncture at the indicated time points after the high-grade CLP procedure were measured using the Mouse Magnetic Luminex eScreening Assay kit (R&D Systems). The assay was performed according to the manufacturer’s instructions.
  • NE-PER nuclear and cytoplasmic extraction Kidneys were collected, cut into small pieces, washed with PBS, and homogenized with beads in certain volumes of cytoplasmic extraction reagent (CER) I, obtained from the NE-PER Nuclear and Cytoplasmic Extraction Reagent kit (ThermoFisher, Waltham, MA), according to the kidney weight. After 15 s of vigorous vortexing and 10 min of incubation on ice, CER II was added to each kidney sample, followed by incubation on ice for 1 min and centrifugation at 16,000 x g for 5 min. The supernatant was collected as a cytoplasmic extract, and the insoluble fraction was suspended in nuclear extraction reagent. After centrifuging for 10 min at 16,000 x g, all supernatants were collected and used as nuclear extracts.
  • CER cytoplasmic extraction reagent
  • kidney injury score Histopathologic assessment of kidney injury score and immunohistochemistry: Five micrometer-thick sections of formalin-fixed and paraffin- embedded kidney tissue samples were processed via hematoxylin-eosin (HE) and IHC staining. For histologically evaluating kidney injury, each slide was stained with HE. A renal pathologist (JIY) scored the percentage of necrotic tubules out of total tubules in cortical areas in a blind manner. Kidney injury was scored according to the percentage of damaged tubules among total tubules: 0, normal; 1, ⁇ 25% damage; 2, 25-50% damage; 3, 50-75% damage, 4, 75-90% damage; and 5, > 90% damage.
  • HE hematoxylin-eosin
  • Histological criteria for injured tubules included the loss of tubular brush border, vacuolization, chromatin condensation, and denuded tubular basement membrane. Renal tubules in five randomly selected high-power fields, typically including 50-100 tubules, were evaluated and scored.
  • tissue sections were deparaffinized, rehydrated in ethyl alcohol, and washed in tap water. Antigens were retrieved in 10 mM sodium citrate buffer for 20 min, using a Black & Decker vegetable steamer. Slides were blocked with 10% donkey serum for 30 min at 22 °C, and washed using PBS. Primary antibodies for NF-KB (Clone 8242S, Cell signaling technology, Danvers, MA) and ICAM-1 (Clone AF796, R&D Systems) were diluted to appropriate concentrations using 2% casein in bovine serum albumin, added to the slides, and incubated overnight at 4 °C.
  • NF-KB Cell signaling technology, Danvers, MA
  • ICAM-1 Clone AF796, R&D Systems
  • Quantitative real-time polymerase chain reaction The established method for whole kidney RNA extraction was used. Briefly, whole kidney samples were homogenized with 700 m ⁇ of RNAiso reagent (Takara Bio Inc., Otsu, Shiga, Japan). Next, 160 m ⁇ of chloroform was added into homogenized samples of kidney cells. Next, the mixture was shaken vigorously for 30 s, kept at 22 °C for 3 min, and centrifuged at 16,000 x g for 15 min at 4 °C. The aqueous phase, located on the top of three phases, was transferred to a fresh tube carefully, to prevent contamination with other phases.
  • PCR conditions were as follows: the initial heating step was performed for 9 min at 95 °C, followed by 35 cycles of denaturation for 30 min at 94.5 °C, annealing for 30 s at 60 °C, and extension for 1 min at 72 °C, and final extension for 7 min at 72 °C. Sequence data for each primer are shown in Table 1.
  • cytokine assay in serum Thirteen inflammatory cytokines and chemokines, including IL-la, IL-Ib, IL-2, IL-4, IL-6, IL-10, IL-17A, CCL2, CCL3, CCL5, CXCL2, IFN-g, and TNF-a were analyzed using Milliplex Mouse Cytokine/Chemokine Magnetic bead Panel - Multiplex Assay kits, according to the manufacturer’s protocol (EMD Millipore, Billerica, MA), using 1 :2 diluted serum. Quantification was performed using a Fuminex 200 microplate reader (EMD Millipore).
  • Terminal deoxynucleotidyl transferase dUTP nick-end labeling assay Cell death was evaluated by the FITC-labelled TUNEL assay, using a commercially available kit (S7110, Merck Millipore, Billerica, MA). TUNEL-positive cells in formalin-fixed renal tissue were identified by examining at least five fields in each section under a 400 x magnification and with digital image analysis (MetaMorph version 4.6r5, Universal Imaging Corp.).
  • KMNCs were isolated according to an established protocol. Both kidneys were removed after exsanguination, decapsulated, finely minced, and incubated in collagenase type 1 (1 mg/ml) (Worthington Biochemical, Lakewood, NJ) for 30 min at 37 °C. After enzymatic digestion, a single-cell kidney suspension was achieved by mechanical disruption of the tissue through a 70 pm strainer (BD bioscience, San Jose, CA). KMNCs were collected following centrifugation at RT using isotonic Percoll density gradient (GE Healthcare, Chicago, IL) (1,500 x g for 30 min in brake off mode), as per the manufacturer’s instructions. Collected cells were washed and re-suspended in RPMI media containing 5% FBS and automatically counted using the Countess® II FL Automated Cell Counter (Life Technologies, Waltham, MA).
  • Splenocytes were collected through mechanical disruption of the spleen using a 70 pm strainer (BD bioscience) and incubated with RBC lysis solution (Qiagen, Hilden, Germany) for 3 min, to remove red blood cells.
  • FACS sorting of kidney CD4 + T cells For FACS sorting, KMNCs were pre-incubated with anti-CD 16/CD32 Fc block (Clone 93, Biolegend, San Diego, CA) for 10 min on ice and stained with monoclonal Ab anti-CD45 (APC, Clone 30-Fl 1, Biolegend), anti-Ly6G (APC- Fire750, Clone 1A8, Biolegend), anti-Ly6C (FITC, Clone HK1.4, Biolegend), anti-F4/80 (PerCP-Cy5.5, Clone BM8, Biolegend), and anti-CD3 (PE, Clone 17A2, Biolegend) for 30 min at 4 °C.
  • DAPI diamidino-2- phenylindole
  • LPS endotoxemia model To generate the LPS endotoxemia mouse model, LPS derived from Escherichia coli (Sigma-Aldrich, Milwaukee, WI, USA) was injected into male mice.
  • High-grade CLP sepsis model and treatment regiment Male C57BL/6 mice purchased from ORIENTBIO (Seongnam-si, Gyeonggi-do, Republic of Korea) were subjected to high- grade CLP at 9-10 weeks of age, using a previously described procedure with minor modification. All mice were housed, and experiments were performed, in a specific-pathogen- free area of Yonsei Biomedical Research Institute, Yonsei University College of Medicine, which was certified by the Association for Assessment and Accreditation of Laboratory Animal Care International. Mice were anesthetized through i.p. injection of a mixture of 80 mg/kg ketamine and 10 mg/kg xylazine prior to all procedures.
  • the peritoneum was opened in a sterile manner and the cecum was ligated using 4-0 black silk thread 1 cm from the distal end and punctured with a 23 -gauge needle. After puncturing, the needle was removed and a small amount of stool was extruded through both punctures to ensure patency. Then, the cecum was placed back into the abdominal cavity and the abdominal incision was closed with 6-0 nylon sutures, with stainless steel removable wound clips used on the skin. After this procedure, 1 ml of prewarmed saline per 20 g body weight was administered subcutaneously. Sham-treated mice underwent the same procedure except for the ligation and puncture of the cecum.
  • 1.0 x 10 9 particles of Exo-naive or Exo-srhcB were administered to mice via i.p. injection at 0, 6, 12 and 18 h.
  • an equivalent volume of phosphate buffer solution (PBS) was injected in the same manner.
  • the animals were assessed every 2 h during the initial 48 h after CLP, and then every 4 h for 5 days. Samples were collected to evaluate the outcome of the procedure within 18 h after CLP.
  • Intravital imaging A laser-scanning intravital confocal microscope (IVM-C; IVIM Technology, Daejeon, South Korea) was used to visualize the biodistribution and anti-septic effects of Exo-srlKB.
  • IVM-C intravital confocal microscope
  • mouse body temperature was maintained at 37°C using a homoeothermic controller.
  • Mice were anaesthetized with intramuscular injections of Zoletil (30 mg/kg) and xylazine (10 mg/kg).
  • a small incision of 10 mm was made in both the skin and peritoneum.
  • Exposed organs were kept hydrated through repeated application of saline during imaging. The biodistribution of exosomes was visualized through wide-area z-stack imaging before and after the injection of Exo-Naive or Exo-srhcB.
  • Luciferase assay HEK293 cells were stably transfected with a luciferase reporter construct regulated under the NF-KB response element (SL-0012; Signosis, Santa Clara, CA, USA) and grown according to the manufacturer’s instructions. Cells were treated with exosomes for 24 h at 37°C, and luciferase activity was measured (El 501; Promega, Madison, WI, USA) using a SpectraMax ID3 microplate reader (Molecular Devices, Sunnyvale, CA, USA).
  • Flow cytometry The levels of surface markers expressed on HUYECs were assessed using flow cytometry. The cells were separated and harvested through centrifugation, labeled with phycoerythrin (PE)-conjugated antibodies specific for human ICAM-1 (BD Biosciences) on ice for 30 min in the dark, and then washed extensively. All samples were analyzed with a BD Celesta flow cytometer (BD Biosciences). The data were analyzed using BD FACSDiva software. PE-conjugated antibodies specific to IgG l k (BD Biosciences) were used as the isotype control.
  • PE phycoerythrin
  • Example 1 Characterization and analysis of engineered exosomes [0123] The production, collection, and purification of exosomes have been thoroughly described in previous studies as shown in Figure 13. The size of particles mostly ranged from 30 to 120 nm, with a mean size of 101 nm.
  • Transmission electron microscopy (TEM) revealed intact cup-shaped membrane vesicles with sizes in accordance with nanoparticle tracking analysis (NT A) results ( Figure 1, B and C).
  • Immunoblotting analysis results for Exo-srlxB revealed the robust expression of target proteins, including srlxB, mCherry, CD9, and GFP with positive exosome markers, such as CD63, TSG101, Alix, and GAPDH.
  • Exo-srlKB lacked the expression of cell organelle markers, including prohibitin, calnexin, GM130, and nucleoporin p62. Exo-Naive did not express any markers except exosome markers ( Figure ID).
  • mice group that received Exo-srlxB showed a significantly lower level of serum blood urea nitrogen (BUN) and creatinine than the Exo-Naive-injected group after IRI surgery, both in pre-treatment and post-treatment models (24-h/48-h BUN and creatinine in both models, P ⁇ 0.001) ( Figure 2B and 2C).
  • Serum neutrophil gelatinase-associated lipocalin (NGAL) levels were significantly reduced in the Exo-srlKB-injected group, in the pre- and post treatment models as compared with those in the Exo-Naive group ( P ⁇ 0.001) ( Figure 2D).
  • Exo-srlKB treatment significantly downregulated the DNA binding activity of NF-KB after kidney IRI as compared with that in the Exo-Naive group, regardless of treatment timings (Pre-treatment: 24-/48-h after IRI, P ⁇ 0.05; Post-treatment: 24-/48-h after IRI, P ⁇ 0.01) ( Figure 3B). Further validation through NF-KB immunohistochemical (IHC) staining showed a significant reduction in NK-KB expression in the Exo-srlKB-treated group as compared with that of the control treatment group (Pre-treatment: 24- h after IRI, P ⁇ 0.05; 48-h, P ⁇ 0.001 and post-treatment: 24-/48-h after IRI, P ⁇ 0.05) ( Figure 3C).
  • systemic Exo-srlicB treatment can reduce renal NF-KB signaling in post-IR kidneys.
  • Example 4 EXO-SITKB ameliorates inflammation in post-ischemic kidneys [0126] To further investigate whether systemic exosomal srlxB treatment locally alleviates renal IRI-induced inflammation, gene expression levels of pivotal inflammatory mediators was determined, including //-7a, II- 1b, 11-2, 11-4, 11-6, II- 10, II- 17a, Ccl2, Ccl3, Ccl5, Cxcl2, Ifii-g, and Tnf-a through quantitative real-time polymerase chain reaction (qRT-PCR).
  • qRT-PCR quantitative real-time polymerase chain reaction
  • EXO-SITKB alleviates apoptosis in post-ischemic kidneys [0128] Based on the well-known role of NF-KB in modulating programmed cell death, how Exo-srlKB affected apoptosis in post-ischemic kidneys was investigated. Kidney IRI surgery induced significant apoptosis in renal cells, which showed an abrupt increase in cleaved caspase- 3 and cleaved Poly (ADP-ribose) polymerase (P RP) levels in western blot analysis.
  • ADP-ribose ADP-ribose
  • Example 6 Biodistribution of exosomes after renal ischemia- reperfusion injury
  • the biodistribution of Exo-srlxB in renal IRI model was investigated to better understand which cell types are orchestrating the protective effect of Exo-srlxB in post- ischemic kidneys.
  • Mice were intravenously injected with fluorochrome-labeled Ly6G and F4/80 antibodies 1 h before IRI surgery and intravenously injected with 9 x 10 9 pn of Exo-srlxB 1 hr after reperfusion. Intravital imaging was performed at 10 min, 5 h, and 24 h after reperfusion ( Figure 6A).
  • Example 7 Systemic delivery of Exo-srlnB affects the renal immune cell population after kidney IRI
  • Exo-srlKB-injected group had significantly lower frequency of neutrophil (0)45P ⁇ 60 + ) ( P ⁇ 0.01), pro-/anti-inflammatory mononuclear phagocytic cells (0)453 ⁇ 4t6u7 CD45 + F4/80 + ) (P ⁇ 0.01 IP ⁇ 0.05, respectively), and T cells (CD45 + CD3 + ) (P ⁇ 0.05) among total KMNCs than those in the Exo-Naive-injected group ( Figure 7C).
  • Results of immunofluorescence of IR-damaged kidneys also showed decreased neutrophil and mononuclear phagocytic cell frequencies in Exo-srlKB-injected kidneys ( Figure 7D).
  • Exo-srlxB treatment did not have significant effects on the total immune cell numbers or frequencies in the spleen after ischemic AKI. There were no differences in total cell numbers and frequencies of total immune cells (CD45 + ), neutrophils (CD45 + Fy6G + ), and pro-/anti-inflammatory mononuclear phagocytic cells (CD45 + Fy6C + / CD45 + F4/80 + ), except T cells (CD45 + CD3 + ) between the Exo-srlxB and Exo-Naive treatment groups. This confirms that systemic Exo-srlxB treatment before ischemic AKI has significant local effects on renal immune cell proliferation/trafficking, but not on splenic immune cells.
  • Example 9 More characterization and analysis of srli B-loaded exosomes
  • culture medium was collected from each cell type.
  • HUVECs were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA) and cultured in F-12K medium (ATCC) containing 10% fetal bovine serum (FBS; Atlas Biologicals, Fort Worth, CO, USA), heparin (Sigma-Aldrich), endothelial cell growth supplement (BD Biosciences) and 1% penicillin/streptomycin (Thermo Fisher). HUVECs between the third and sixth passages were used in all experiments.
  • ATCC American Type Culture Collection
  • FBS fetal bovine serum
  • heparin Sigma-Aldrich
  • endothelial cell growth supplement BD Biosciences
  • penicillin/streptomycin Thermo Fisher
  • Human monocytes were purchased from ATCC and cultured in RPMI 1640 medium (Welgene, Daegu, Korea) with 10% FBS, 1% penicillin/streptomycin and 0.05 mM 2-mercaptoethanol (Sigma-Aldrich).
  • Exosomes were isolated using a combination of tangential flow filtration (TFF) and size-exclusion chromatography (SEC) (Figure 13 A). To reduce the risk of loading the SEC column with impurities that would exceed its binding capacity, the samples were subjected to diafiltration and concentration through TFF. After TFF, the concentrated medium was loaded onto an SEC column for further purification. A second TFF was then performed to concentrate the exosomes. Each type of exosome isolated through sequential purification was analyzed via nanoparticle tracking analysis (NTA), which characterizes the size and concentration of exosomes ( Figure 13B).
  • NTA nanoparticle tracking analysis
  • Exo-srlKB -mediated effects were associated with modest protection from the LPS-induced temperature drop (Figure 14A).
  • a mouse mouse model of LPS-induced sepsis was developed using BALB/c mice.
  • Exo-srhcB treatment significantly improved the survival rate of LPS-induced sepsis in BALB/c mice ( Figure 10A, middle).
  • the role of Exo-srhcB in survival was examined next, using a model of CLP-induced sepsis, as a standardized murine model of intra-abdominal sepsis. The animals survived the 7-day monitoring period, indicating that Exo-srhcB provides lasting protection against sepsis mortality (Figure 10A, lower).
  • Exo-srhcB treatment alleviated the inflammation induced by sepsis
  • enzyme-linked immunosorbent assay (ELISA) was used to determine the concentrations of key inflammation factors in the serum.
  • Significant reductions significant reductions were observed in the expression levels of the pro- inflammatory cytokines TNFa, IL-Ib, and IL-6, and the chemokine carbon tetrachloride (CCL4), in the Exo-srhcB treatment group with LPS-induced sepsis ( Figure 10B, upper).
  • Serum levels of TNF-a and CCL4 in the CLP group were also observed to be significantly higher than those in sham-operation control mice, but were not elevated in mice treated with both CLP and Exo- srhcB (Figure 10B, lower). These results suggest that Exo-srhcB alleviates inflammation induced by LPS or CLP and protects septic mice against the acute inflammatory process.
  • Exo-srlKB treatment markedly decreased damage to tubular areas, preserving proximal tubules and demonstrating the therapeutic potential of Exo-srhcB for sepsis-induced renal injury. These results are consistent with those for LPS-induced AKI ( Figure 14B). The severity of histological kidney damage was further quantified, and found that Exo-srhcB treatment significantly reduced the injury score by 50% in both sepsis models ( Figure 10D). Exo-srlxB ameliorated degenerative changes in the renal tubules, although some damaged areas were present. These data suggest the critical importance of Exo-srhcB in improving renal physiological structure and function in septic mice.
  • Exosomes were delivered mainly to the neutrophils of the liver ( Figure 1 IB), and increased recruitment of neutrophils and macrophages was observed, as well as increased uptake of exosomes, in the spleens of LPS-induced mice ( Figure 11C). mCLING- labeled exosomes were also delivered to neutrophils in the kidneys of LPS-injected mice ( Figure 15A). These observations suggest that therapeutic exosomes were successfully delivered to target neutrophils and macrophages within 30 minutes in the septic mouse model.
  • Example 12 EXO-SITKB alleviates inflammation associated with sepsis [0141] The target specificity and efficiency of Exo-srhcB in vitro was identified. Exo-srlKB, but not Exo-Naive, significantly blocked TNF-a induced NF-KB activation in an HEK293 cell stably expressing an NF-KB luciferase reporter gene (Figure 12A). Increasing doses of Exo- srlxB decreased NF-KB reporter activity in a dose-dependent manner, indicating that srhcB released from exosomes prevents NF-KB transcriptional activity ( Figure 12B).
  • THP-l cells are widely employed to represent monocytes in cell culture models.
  • THP-1 cells were stimulated with LPS, treatment with Exo-srlKB led to decreased secretion of the inflammatory cytokines TNF-a and monocyte chemoattractant protein (MCP)-l, which are under NF-KB transcriptional control, compared with Exo-Naive ( Figure 12C).
  • MCP monocyte chemoattractant protein
  • Exo-srlKB was also compared with those of the commonly used NF-KB inhibitor JSH-23, which interferes with the binding of NF-KB to its target DNA.
  • the example data shows that Exo-srl B and JSH- 23 had comparable effects with respect to inhibiting EPS-induced NF-KB activation and cytokine production in THP-1 cells ( Figure 12C, lane 4 versus lane 6).
  • Sepsis is characterized by monocyte adherence to the endothelium. Finder normal conditions, endothelial cells are quiescent and do not interact with monocytes. However, in an inflammatory environment, activated endothelial cells express adhesion molecules, such as intercellular cell adhesion molecule (ICAM)-1, which bind to monocytes. The expression of adhesion molecules is widely considered to be activated during the development of sepsis.
  • IAM intercellular cell adhesion molecule
  • IF-8 and MCP-1 also triggered the migration and adhesion of monocytes to the vascular endothelium and led to extravasation of these cells into the surrounding tissues. Expression of IF-8 and MCP- 1 has been observed previously in various tissues during acute inflammation caused by bacterial and viral infections, and is associated with severe sepsis.
  • Example 13 Protective effect of Exo- srli Bs on lung injury in LPS-induced sepsis
  • Animals received a single intraperitoneal injection with a lethal dose of EPS (30 mg/kg body weight for 30 min, followed by a single Intravenous injection of Exo-srlkB (lxlO 10 ) for 10 hrs.
  • Lung tissues were analyzed by H&E staining as shown in Figure 17.
  • the group injected with LPS for 10 hrs showed the neutrophil infiltration and alveolar macrophage proliferation, intra-alveolar and external hyperemia, and bleeding proceeded. Alveolar endothelial cell damage as well as alveolar necrosis are observed.
  • Exosome administration alleviated alveolar wall expansion, neutrophil infiltration and alveolar macrophage proliferation even if congestion is slightly observed.
  • Embodiment 1 A method for treating acute kidney injury (AKJ) in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-kB inhibitor to the subject.
  • AKJ acute kidney injury
  • Embodiment 2 The method of any of the above embodiments, wherein the NF-KB inhibitor is selected from the group consisting of a NF-KB inhibitory protein, its fragment, and a mixture thereof.
  • Embodiment 3 The method of any of the above embodiments, wherein the NF-KB inhibitor is selected from the group consisting of super-repressor (SR)-IKB (srlicB), IkB-a, IkB-b, IkB-e, BCF-3, a mutant thereof, and a mixture thereof
  • Embodiment 4 The method of any of the above embodiments, wherein the NF-KB inhibitor is super-repressor (SR)-IKB (srlKB).
  • Embodiment 5 The method of any of the above embodiments, wherein the composition of is administered via oral, transdermal, intraperitoneal, intravenous, intramuscular, subcutaneous, or mixed routes.
  • Embodiment 6 The method of any of the above embodiments, wherein the subject is human.
  • Embodiment 7 The method of any of the above embodiments, wherein the method further comprises administering an anti-inflammatory agent to the subject.
  • Embodiment 8 The method of any of the above embodiments, wherein the exosome comprising a NF-KB inhibitor further comprises a photo-specific binding protein.
  • Embodiment 9 The method of any of the above embodiments, wherein the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:l or SEQ ID NO:2.
  • Embodiment 10 The method of any of the above embodiments, wherein the NF-KB inhibitor is represented by SEQ ID NO: 1.
  • Embodiment 11 The method of any of the above embodiments, wherein the NF-KB inhibitor is represented by SEQ ID NO: 2.
  • Embodiment 12 The method of any of the above embodiments, wherein the photo specific binding protein is a first photo-specific binding protein and/or a second photo-specific binding protein; the first photo-specific binding protein is conjugated to an exosome specific marker to form fusion protein I; and the second photo-specific binding protein is conjugated to the NF-KB inhibitor to form fusion protein II.
  • the photo specific binding protein is a first photo-specific binding protein and/or a second photo-specific binding protein
  • the first photo-specific binding protein is conjugated to an exosome specific marker to form fusion protein I
  • the second photo-specific binding protein is conjugated to the NF-KB inhibitor to form fusion protein II.
  • Embodiment 13 The method of embodiment 12, wherein the fusion protein I and the fusion protein II are linked reversibly through the first photo-specific binding protein and the second photo-specific binding protein.
  • Embodiment 14 The method of embodiment 12 or 13, wherein the first photo-specific binding protein is conjugated to an exosome specific marker to be located in the direction toward inside of the exosome.
  • Embodiment 15 The method of any of embodiments 12-14, wherein the first photo specific binding protein and the second photo-specific binding protein are selected from the group consisting of CIB, CIBN, PhyB, PIF, FKFl, GIGANTEA, CRY and PHR.
  • Embodiment 16 The method of embodiment 15, wherein the first photo-specific binding protein is CIB or CIBN and the second photo-specific binding protein is CRY or PHR.
  • Embodiment 17 The method of embodiment 15, wherein the first photo-specific binding protein is CRY or PHR and the second photo-specific binding protein is CIB or CIBN.
  • Embodiment 18 The method of embodiment 15, wherein the first photo-specific binding protein is PhyB and the second photo- specific binding protein is PIF.
  • Embodiment 19 The method of embodiment 15, wherein the first photo-specific binding protein is PIF and the second photo-specific binding protein is PhyB.
  • Embodiment 20 The method of embodiment 15, wherein the first photo-specific binding protein is GIGANTEA and the second photo-specific binding protein is FKFl.
  • Embodiment 21 The method of embodiment 15, wherein the first photo-specific binding protein is FKFl and the second photo-specific binding protein is GIGANTEA.
  • Embodiment 22 The method of embodiments 12-21, wherein the exosome specific marker is selected from the group consisting of CD9, CD63, CD81 and CD82.
  • Embodiment 23 The method of any of the above embodiments, wherein the exosome has a diameter of between about 50 nm and about 200 nm.
  • Embodiment 24 The method of any of the above embodiments, wherein the exosome has a diameter of between about 50 nm and about 150 nm.
  • Embodiment 25 The method of any of the above embodiments, wherein the composition is a pharmaceutical composition further comprising a physiologically acceptable carrier.
  • Embodiment 26 A composition comprising an exosome comprising an NF-KB inhibitor as described in Embodiments 1-16 for treating AKX.
  • Embodiment 27 Else of a composition comprising an exosome comprising an NF-KB inhibitor as described in Embodiments 1-16 for treating AKI.
  • Embodiment 1 A method for treating a disease caused by sepsis in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-KB inhibitor to the subject.
  • Embodiment 2 The method of any of the above embodiments, wherein the disease is selected from the group consisting of pneumonia, cytokine storm syndrome, respiratory distress syndrome, and organ failure.
  • the disease is pneumonia.
  • the disease is cytokine storm syndrome.
  • the disease is respiratory distress syndrome.
  • the disease is organ failure.
  • Embodiment 3 The method of any of the above embodiments, wherein the NF-KB inhibitor is selected from the group consisting of a NF-KB inhibitory protein, its fragment, and a mixture thereof.
  • Embodiment 4 The method of any of the above embodiments, wherein the NF-KB inhibitor is selected from the group consisting of super-repressor (SR)-IKB (srlxB), IkB-a, IkB-b, IkB-e, BCL-3, a mutant thereof, and a mixture thereof.
  • SR super-repressor
  • IkB-a IkB-a
  • IkB-b IkB-e
  • BCL-3 a mutant thereof
  • a mutant thereof a mixture thereof.
  • Embodiment 5 The method of any of the above embodiments, wherein the NF-KB inhibitor is super-repressor (SR)-IKB (srbcB).
  • SR super-repressor
  • srbcB super-repressor
  • Embodiment 6 The method of any of the above embodiments, wherein the composition of is administered via oral, transdermal, intraperitoneal, intravenous, intramuscular, subcutaneous, or mixed routes.
  • Embodiment 7 The method of any of the above embodiments, wherein the subject is human.
  • Embodiment 8 The method of any of the above embodiments, wherein the method further comprises administering an anti-inflammatory agent to the subject.
  • Embodiment 9 The method of any of the above embodiments, wherein the exosome comprising a NF-KB inhibitor further comprises a photo-specific binding protein.
  • Embodiment 10 The method of any of the above embodiments, wherein the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:l or SEQ ID NO:2.
  • Embodiment 11 The method of embodiment 10, wherein the NF-KB inhibitor is represented by SEQ ID NO: 1.
  • Embodiment 12 The method of embodiment 10, wherein the NF-KB inhibitor is represented by SEQ ID NO: 2.
  • Embodiment 13 The method of any of the above embodiments, wherein the photo specific binding protein is a first photo-specific binding protein and/or a second photo-specific binding protein; the first photo-specific binding protein is conjugated to an exosome specific marker to form fusion protein I; and the second photo-specific binding protein is conjugated to the NF-KB inhibitor to form fusion protein II.
  • the photo specific binding protein is a first photo-specific binding protein and/or a second photo-specific binding protein
  • the first photo-specific binding protein is conjugated to an exosome specific marker to form fusion protein I
  • the second photo-specific binding protein is conjugated to the NF-KB inhibitor to form fusion protein II.
  • Embodiment 14 The method of embodiment 13, wherein the fusion protein I and the fusion protein II are linked reversibly through the first photo-specific binding protein and the second photo-specific binding protein.
  • Embodiment 15 The method of embodiment 13 or 14, wherein the first photo-specific binding protein is conjugated to an exosome specific marker to be located in the direction toward inside of the exosome.
  • Embodiment 16 The method of any one of embodiments 13-15, wherein the first photo- specific binding protein and the second photo- specific binding protein are selected from the group consisting of CIB, CIBN, PhyB, PIF, FKF1, GIGANTEA, CRY and PHR.
  • Embodiment 17 The method of embodiment 16, wherein the first photo-specific binding protein is CIB or CIBN and the second photo-specific binding protein is CRY or PHR.
  • Embodiment 18 The method of any of the above embodiments, wherein the first photo-specific binding protein is CRY or PHR and the second photo- specific binding protein is CIB or CIBN.
  • Embodiment 19 The method of embodiment 16, wherein the first photo-specific binding protein is PhyB and the second photo- specific binding protein is PIF.
  • Embodiment 20 The method of embodiment 16, wherein the first photo-specific binding protein is PIF and the second photo-specific binding protein is PhyB.
  • Embodiment 21 The method of embodiment 16, wherein the first photo-specific binding protein is GIGANTEA and the second photo-specific binding protein is FKF1.
  • Embodiment 22 The method of embodiment 16, wherein the first photo-specific binding protein is FKF1 and the second photo-specific binding protein is GIGANTEA.
  • Embodiment 23 The method of any one of embodiments 13-22, wherein the exosome specific marker is selected from the group consisting of CD9, CD63, CD81 and CD82.
  • Embodiment 24 The method of any of the above embodiments, wherein the exosome has a diameter of between about 50 nm and about 200 nm.
  • Embodiment 25 The method of any of the above embodiments, wherein the exosome has a diameter of between about 50 nm and about 150 nm.
  • Embodiment 26 The method of any of the above embodiments, wherein the composition is a pharmaceutical composition further comprising a physiologically acceptable carrier.
  • Embodiment 27 A composition comprising an exosome comprising an NF-KB inhibitor as described in Embodiments 1-16 for treating a disease caused by sepsis..

Abstract

The present disclosure relates to methods for treating acute kidney injury using exosome containing NF-κB inhibitors. The present disclosure relates to methods for treating diseases induced by sepsis using exosome containing NF-κB inhibitor.

Description

USE OF EXOSOME-BASED DELIVERY OF NF-KB INHIBITORS
FIELD OF THE INVENTION
[0001] The present disclosure is related to methods for treating acute kidney injury using exosome containing NF-kB inhibitors. The present disclosure is related to methods for treating diseases induced by sepsis using exosome containing NF-KB inhibitor.
BACKGROUND OF THE INVENTION
[0002] AKI not only contributes to short-term adverse outcomes, but the survivors may suffer from chronic kidney (CKD) and even end-stage renal disease (ESRD). As one of the most important components of the pathogenesis, systematic inhibition of NF-KB affects the severity of AKI. In a disease model induced by folic acid, inhibition of NF-KB mitigates AKI-injury by reduction of RelA and NF-KB2 activation.
[0003] More than 800 synthetic and natural materials are known to be partly involved in modulating NF-KB activation. Several studies have shown that NF-KB signaling blockades, such as the renin-angiotensin-aldosterone system (RAAS) blockade or tumor necrosis factor-a (TNF- a) blocker, can alleviate kidney damage. However, their mechanisms of action are pleiotropic and lack specificity. Recent advancements in nano-technology made it feasible to produce specific gene sequences or nanoparticles to target NF-KB signaling. However, no NF-KB inhibitor has been commercially approved for human use yet.
[0004] Uncontrolled inflammation is a prominent feature of the septic response. Host- pathogen interactions mediated by Toll-like receptors (TLRs) stimulate the production of pro- inflammatory cytokines, chemokines, and immune-activating molecules. This pro-inflammatory response is followed by a compensatory immunosuppressive response that involves various quantitative and functional defects of immune cells. Pathogen-induced cellular modification is accompanied by marked changes in host gene expression, with nuclear factor kappa-B (NF-KB) transcription factors playing a pivotal role in modulating these changes. Several cytokines under the regulatory control of NF-KB, including tumor necrosis factor (TNF)-a and interleukin (IL)-l, can induce further activation of this transcriptional factor, leading to potentiation of the inflammatory response and septic shock. In addition, NF-KB is involved in the apoptotic response, primarily through enhancing the transcription of antiapoptotic genes such as BC1-XL, A1 and A20. Inflammation associated with NF-KB activation can thus be exacerbated by two interacting mechanisms, increased expression of pro-inflammatory mediators and prolongation of the life spans of cell populations such as neutrophils, which are activated to produce pro- inflammatory molecules and participate directly in acute inflammatory processes.
[0005] There are a variety of diseases, such as pneumonia, cytokine storm syndrome, respiratory distress syndrome, and organ failure that are caused by sepsis. Sepsis is a systemic inflammatory syndrome caused by activation of the innate immune system due to acute microbial infection and continues to be the main cause of mortality in intensive care units. Moreover, there are various diseases caused by sepsis that have vastly different symptoms, some of which are fatal. Unfortunately, the common treatment for Sepsis (i.e., antibiotics) may not be effective to treat the different diseases caused by sepsis, such as cytokine syndrome or damaged organs. Currently, there are no treatments available for clinical usage for diseases caused by sepsis. Thus, the development of effective alternative therapies is urgently needed. Although more than 700 inhibitors have been reported for NF-KB, no inhibitors have been approved as therapeutic agents to date. Moreover, various steroidal and nonsteroidal anti-inflammatory agents are known to block NF-KB, but their effects are not specific for inhibiting NF-KB.
[0006] Successful gene and drug delivery require the selection of an appropriate vector for delivering target molecules stably to the site of action, without causing adverse effects to the recipient. Various types of vectors, including recombinant adenoviruses, liposomes, ligand- conjugated nanoparticles, and ultrasound microbubbles, are reportedly efficient for drug delivery, because of their stability and high loading capacity. However, certain concerns and limitations were associated with their use. Though they were rapidly recognized and cleared by the reticuloendothelial system, their non-uniform particle size and non-specific uptake pattern limited their use as bio-carriers. The high immunogenicity of adenoviral vectors could provoke an immune response after administration and the virus itself can be toxic at high doses. The use of liposomes can also induce adverse immunogenic and non-IgE-mediated hypersensitivity reactions.
[0007] Current conventional strategies to deliver the therapeutic proteins include enveloping the proteins within synthetic nanoparticles. The most preferred protein delivery systems are liposomes and polymeric nanoparticles (PNPs). A liposome is a synthetic vesicle with a phospholipid membrane that self-assembles into various sizes and shapes in an aqueous environment. A PNP is a solid colloidal particle with the size between 10-1000 nm, and is made up of biodegradable polymers, in which the cargo proteins can be entrapped, encapsulated or attached to a nanoparticle matrix. However, liposomes tend to fuse or aggregate with each other resulting in premature release of the liposome cargo over time. PNPs may have better stability than liposomes, but their biocompatibility and long-term potential safety are still a concern. In addition, encapsulating proteins to liposomes and PNPs requires generation of synthetic or recombinant proteins and ex vivo encapsulation processes, whereas EXPLOR technology requires generation and maintenance of stable cells that can produce cargo-loaded exosomes via natural exosome biogenesis. In addition, exosomes have many desirable features of an ideal protein delivery system, such as biocompatibility, minimal or no inherent toxicity, long half-life in the circulation, and intrinsic ability to target tissues.
[0008] Recently, exosomes have received significant attention as novel bio-carriers for gene/drug delivery. Exosomes are extracellular vesicles (EVs) that play an important role in cell- to-cell communication, by transferring bioactive materials to recipient cells or affecting signaling pathways of target cells. Exosomes are easier to store and exhibit greater stability than other bioactive agents. Exosomes have a high capacity to overcome biological barriers, and can carry surface molecules targeting specific cell types, thus causing fewer off-target effects.
SUMMARY OF THE INVENTION
[0009] The present disclosure provides methods for treating acute kidney injury (AKI) in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-KB inhibitor to the subject.
[0010] The present disclosure also provides methods for treating a disease caused by sepsis in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-KB inhibitor to the subject.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. Characterization of engineered exosomes produced from HEK293T [0011] (A) Schematic diagram of DNA constructs used for producing super-repressor IKB- loaded exosomes (Exo-srlKB) (upper), and biogenesis of cargo protein-carrying exosomes using optically reversible protein- protein interactions, so called EXPLOR technology (lower). (B) Representative TEM images of Exo-Naive and Exo-srlKB. Scale bar: 100 nm. (C) Concentrations and size distributions of Exo-Naive (upper) and Exo-srlKB (lower) were determined by a Zetaview instrument. (D) Immunoblotting HEK293T cells and HEK293T cell- derived exosomes to analyze the expression of target proteins (srbcB, mCherry, CD9, and GFP), exosome positive markers (CD63, TSG101, Alix, and GAPDH), and exosome negative markers (Cell organelle markers; prohibitin, calnexin, GM130, and nucleoporin p62). Naive cells and Exo-naive were used as negative controls of Exo-srlKB.
Figure 2. Renal protective effects of Exo-srlKB after kidney IRI [0012] (A) Experimental scheme of kidney IRI surgery and exosome delivery. Each mouse group was intraperitoneally injected with 3 x 109 pn of Exo-naive or Exo-srlxB three times in a 1-h interval (total of 9 x 109 pn). Mice were sacrificed either 24 or 48 h after IRI surgery and serum and tissues were collected for further evaluation. (B-D) Serum levels of BUN, creatinine, and NGAL among different groups depending on treatment type (Exo-naive vs. Exo-srlxB), drug delivery timing (Pre-treatment vs. Post-treatment), and follow-up time point (24-h and 48-h), which shows the renal protective effect of Exo-srlxB treatment (Pre-treatment 24-h, n = 10; pre treatment 48-h, n = 4-5; post- treatment 24-h, n = 5-10; post-treatment 48-h, n = 4-11).
(E) Left: Representative PAS staining images of cortical tubular cells in kidney sections from each group. Normal proximal tubular brush border (*) or loss of brush border (o); chromatin condensation (black arrows); denuded basement membrane (black arrowheads); vacuolization (yellow arrows); scale bar, 100 mM. Right: Pathological tubular injury; numerous kidney samples from each group show fewer tubular injuries with Exo-srlxB treatment than those with Exo-Naive treatment (Pre-treatment 24-h, n = 5; post-treatment 24-h, n = 5). Comparisons between groups were assessed using one-way ANOVA with Bonferroni post hoc test. Data were represented as mean ± SD values. ***P < 0.001, upon comparing Exo-Naive-Sham and Exo-Naive-IRI surgery groups. #P < 0.05 and ###P < 0.001, upon comparing Exo-Naive-IRI and Exo-srlKB-IRI surgery groups.
Figure 3. Suppression of IRI-induced NF-KB activation following Exo-srlKB treatment
[0013] (A) Western blot analysis of NF-KB p65 expression using renal nuclear extracts from each experimental mouse group. Nuclear extracts were biochemically separated from cytoplasmic fractions, and NF-KB p65 and Lamin B1 expression was analyzed via western blotting. IRI-induced activation of NF-KB signaling was significantly repressed with peri operative Exo-srlKB treatment. (B) Elevated DNA-binding activity of NF-KB p65 following renal IRI was suppressed with perioperative Exo-srlxB treatment. (C) Left: Representative IHC images with NF-KB p65 antibody from each treatment group. Scale bar, 50 mM. Right: Graphical representation showing immunohistochemical detection of NF-KB p65 in each experimental group. Treatment with Exo-srlKB decreased NK-KB expression as compared with that of the control group (Pre-treatment 24-h, n=5-8; pre-treatment 48-h, n=4-5; post-treatment 24-h, n=5- 10; post-treatment 48-h, n=4-l 1). Comparisons between groups were assessed using one-way ANOVA with Bonferroni post hoc test. Data were represented as mean ± SD values. **P < 0.01, ***P < 0.001, upon comparing Exo-Naive-Sham and Exo-Naive-IRI surgery groups. #P < 0.05, ##P < 0.01, ###P < 0.001, upon comparing Exo-Naive-IRI surgery group vs. and Exo-srlKB-IRI surgery groups.
Figure 4. The expression of pro-inflammatory cytokines/chemokines and adhesion molecules is negatively affected by EXO-SITKB treatment both locally and systemically. [0014] (A) Whole kidney lysates from each experimental group were used for qRT-PCR, to measure expression levels of pro-inflammatory cytokines, including I1-1b, 11-6, Tnf-a, Ccl2,
Ccl5, and Cxcl2. The mRNA levels of those genes increased significantly after IR damage in the kidney, and this effect was alleviated with Exo-srlKB treatment (Pre-treatment 24-h, n=5-8; pre treatment 48-h, n=4-5; post- treatment 24-h, n=5-10; post- treatment 48-h, n=4-ll). (B) Multiplex cytokine studies using serum from each experimental group also showed a similar tendency of reduction in pro-inflammatory cytokine levels in the post-ischemic mouse group with Exo-srlKB treatment. (C) qRT-PCR data show increased levels of Icam-1 mRNA in post- ischemic Exo-naive treatment group and significant reduction of Icam-1 mRNA with Exo- srb<B treatment. (D) Western blot analysis results of whole kidney lysates from each group demonstrated decreased expression of ICAM-1 in IR-injured kidneys with Exo-srlKB treatment. (E) Representative kidney sections exhibiting ICAM-1 IHC staining (Left) and the graphical representation of ICAM-1 IHC (Right) show that Exo-srl KB treatment reduced ICAM-1 expression in post-ischemic kidneys of C57BL/6J mice. Scale bar, 50 mM (B-E: Pre treatment 24-h, n=5; pre-treatment 48-h, n=4-5; post-treatment 24-h, n=5; post-treatment 48-h, n=3-6). Comparisons between groups were assessed using one-way ANOVA with Bonferroni post hoc test. Data were represented as mean ± SD values. *P < 0.05, **/' < 0.01 , ***P < 0.001, upon comparing Exo-Naive-Sham and Exo-Naive-IRI surgery groups. #P < 0.05, ##P < 0.01, ###P < 0.001 upon comparing Exo-Naive-IRI and Exo-srlKB-IRI surgery groups.
Figure 5. Exo-srlKB treatment improves IR-induced kidney apoptosis.
[0015] (A) Left : Western blot analysis results that compare the expression of cleaved caspase-
3 and cleaved PARP protein in kidneys from each experimental group. Right. Graphical representation shows that Exo-srlKB treatment lowered cleaved caspase-3 levels and cleaved PARP as compared with the Exo-Naive treatment. (B) Left Representative images of TUNEL staining (FITC-labelled) are shown. Apoptotic cells are green (White arrows) and DAPI was used as a counterstain. Scale bar, 50 mM. Righ A bar graph showing TUNEL positive cells in kidney sections from each group. Exo-srlKB treatment induced a lower number of apoptotic cells (Pre-treatment 24-h, n=5; pre-treatment 48-h, n=4-5; post-treatment 24-h, n=5; post-treatment 48-h, n=3-6). Comparisons between groups were assessed using one-way ANOVA with the Bonferroni post hoc test. Data were represented as mean ± SD values. ***P < 0.001, upon comparing Exo-Naive-Sham and Exo-Naive-IRI surgery groups. #P < 0.05, ##P < 0.01, and ###P < 0.001, upon comparing Exo-Naive-IRI and Exo-srlKB IRI surgery groups.
Figure 6. Biodistribution of exosomes after renal ischemia-reperfusion injury.
[0016] (a) Experimental scheme of intravital imaging and exosome delivery (b) Sequential intravital imaging shows the uptake of DiD-labeled exosome (green) into neutrophils (Ly6G+, red) and macrophages (F4/80+, blue) in the post-ischemic kidneys treated with intravenous Exo- srlxB. White arrow indicates DiD-labeled exosomes engulfed in immune cells. White dashed line indicates renal interstitium. (c) The biodistribution of DiD-labeled exosomes (green) in spleen after kidney IRI surgery shows exosomal uptake into neutrophils (Ly6G+, red) and macrophages (F4/80+, blue) in the outer parenchyma. Elapsed time is indicated. Scale bar, 20 pm.
Figure 7. Exo-srlKB treatment modulates kidney immune cell populations following IR-induced AKI. [0017] (A) Mice were sacrificed 24 h after surgery. KMNCs were enriched using enzymatic digestion, mechanical disruption, and Percoll density gradients. Total kidney cell numbers determined using those methods showed no statistical differences among the experimental groups. (B) Graphical representation of flow cytometric analysis showed a higher percentage of kidney CD45+ cells after IRI among isolated kidney cells using enzymatic digestion, mechanical disruption, and Percoll density gradients. Exo-srlKB treatment alleviated the post-ischemic surge of renal immune cells. (C) Results of further staining with additional immune cell markers showed that the frequency of multilineage immune cells, including neutrophils (CD45+Ly6G+), pro-/anti-inflammatory mononuclear phagocytic cells (CD45+Ly6C+ / CD45+F4/80+), and T cells (CD45+CD3+) among enriched KMNCs were also decreased with Exo-srlKB treatment in post-ischemic kidneys. (D-F) Immunofluorescence studies were performed on post-ischemic kidneys from each experimental group, targeting Ly6G, Ly6C, and F4/80. Secondary antibodies conjugated with Alexa Fluor 647 were used for all immunofluorescence experiments. Data showed a decreased frequency of Alexa Fluor 647 stained cells (white arrow) in post-ischemic kidneys following Exo-srhcB treatment, suggesting that there were fewer neutrophils (Fy6G+) and pro-/anti-inflammatory mononuclear phagocytic cells (Fy6C+ / F4/80+) in Exo-srhcB treated kidneys. Proximal tubular cells were stained with FTF (green) and DAPI was used as for counterstaining. Scale bar, 50 mM. (n=5 per experimental group). Comparisons between groups were assessed using one-way ANOVA with Bonferroni post hoc test. Data were represented as mean ± SD values. *P < 0.05, **P < 0.01, ***p < 0.001, upon comparing Exo-Naive-Sham and Exo-Naive-IRI surgery groups. #P < 0.05, ##P < 0.01, upon comparing Exo-Naive-IRI and Exo- srlKB-IRI surgery groups.
Figure 8. Intravenous delivery of Exo-srlKB shows similar biologic effects compared to intraperitoneal delivery in ischemic AKI model.
[0018] (a) Experimental scheme of kidney IRI surgery and exosome delivery. Each mouse group was intravenously injected with 9 x 109 pn of Exo-Naive, Exo-srhcB, or PBS 1 h after reperfusion. Mice were killed either 24 or 48 h after IRI surgery and serum and tissues were collected for further evaluation (b-d) Serum levels of BUN, creatinine, and NGAL among different groups depending on treatment type (PBS vs. Exo-Naive vs. Exo-srhcB) and follow-up time point (24-h and 48-h), which shows the renal protective effect of Exo-srhcB treatment (n=5 per experimental group) (e) Western blot analysis of NF-KB p65 expression using renal nuclear extracts from each experimental mouse group. Nuclear extracts were biochemically separated from cytoplasmic fractions, and NF-KB p65 and Lamin B1 expression was analyzed via western blotting. IRI-induced activation of NF-KB signaling was significantly repressed with post operative Exo-srlKB treatment (f) Elevated DNA-binding activity of NF-KB p65 following renal IRI was suppressed with postoperative Exo-srlKB treatment (g) qRT-PCR data show increased levels of Icam-1 mRNA in post-ischemic Exo-Naive treatment group and significant reduction of Icam-1 mRNA with Exo-srlKB treatment (h) Western blot analysis results of whole kidney lysates from each group demonstrated decreased expression of ICAM-1 in IR-injured kidneys with Exo-srlKB treatment. Comparisons between groups were assessed using one-way ANOVA with Bonferroni post hoc test. Data were represented as mean ± SD values ns; not significant, **P<0.01, ***/'<0.001 , upon comparing PBS-Sham and Exo-Naive-IRI surgery groups.
#P<0.05, ##P< 0.01, ###P< 0.001 upon comparing Exo-Naive-IRI and Exo-srlKB-IRI surgery groups.
Figure 9 (1). Generation and characterization of engineered exosomes [0019] (A) Schematic of DNA constructs used for the production of super-repressor IKB- loaded exosome (Exo-srlKB) (upper). Schematic showing fusion proteins and their proposed activities (lower). (B) Morphological characterization of Exo-Naive and Exo-srhcB through transmission electron microscopy (TEM) (C) HEK293T cells that stably express mCherry or srhcB, and exosomes from these HEK293T cells, were lysed and immunoblotted against the indicated proteins.
Figure 10 (2). The protective effects of EXO-SITKB in endotoxemia and cecal ligation and puncture (CLP)-induced sepsis
[0020] (A) Survival curves of phosphate-buffered saline (PBS)-treated, Exo-Naive, and Exo- srlxB-treated septic mice. Lipopolysaccharide (LPS) C57BL/6 mice (n = 5-6/group), LPS BALB/c mice (n =10/group), and CLP C57BL/6 mice (n = 14-15/group). **/?<0.01, *p<0.05 compared with the PBS-treated sepsis group. (B) Levels of tumor necrosis factor (TNF)-a, interleukin (IL)-6, IL-Ib, and CCL4/macrophage inflammatory protein (MIR)-Ib in the plasma of exosome-treated mice were measured 24 h after LPS injection or CLP. **/?<0.01, *p<0.05 compared with the PBS-treated sepsis group. †/?<0.05 compared with the Exo-Naive-treated sepsis group. (C) Representative images of cortical tubular cells in kidney sections from sham, CLP with PBS, CLP with Exo-Naive, and CLP with Exo-srhcB mice. Normal brush border (*) of proximal tubules or loss of brush border (o); chromatin condensation (white arrows); denuded basement membrane (white arrow heads); vacuolization (yellow arrows); scale bar, 100 mM. (D) Pathological kidney injury scores of representative kidney samples of each group. *p<0.05 compared with the PBS-treated sepsis group. †/?<0.05 compared with the Exo-Naive-treated sepsis group.
Figure 11 (3). The biodistribution of exosomes in LPS-injected mice [0021] (A) Intravital imaging of mCLING- labeled exosome (red) uptake into neutrophils
(LysMgfP/+, green; Ly6G+, blue) in the liver of sham mice. (B) Representative time-lapse imaging of mCLING-labeled exosomes (red) inside the Ly6G+ neutrophil cells (green) of the liver in LPS-treated C57BL/6 mice. (C) Sequential images of flowing mCLING- labeled exosomes (red) inside the spleen of sham and LPS-treated LysMGFP/+ mice. Elapsed time is indicated. Magenta, autofluorescence; scale bars, 50 pm.
Figure 12 (4). Inhibitory effect of EXO-SITKB on nuclear factor-kappa-B (NF-KB) signaling in vitro
[0022] (A) HEK293-NF-KB-luciferase cells (2x104 cells) were cultured either with Exo-Naive or with Exo-srlKB 2x105 particles. After 24 h, the cells were treated with 0.5ng/ml TNF-a for an additional 18 h. Luciferase activities were measured and normalized. (B) Exo-srlKB dose- dependently repressed NF-KB activation in NF-KB-luciferase cells. (C) THP-1 cells (5x105 cells) were stimulated with 1 pg/ml LPS and then treated with Exo-srhcBs 5x106 particles. The supernatants were collected and assayed for the production of TNF-a and monocyte chemoattractant protein (MCP)-l. JSH-23 (50 pM) was used as the positive control. **/><0.01 (D) Immunofluorescence of human umbilical vein endothelial cells (HUVECs) incubated with mCLING-labeled Exo-srlKB. Representative images are shown. Nuclei were labeled with Hoechst. (E) HUVECs were stimulated with 300 ng/ml LPS for 24 h. Cells were harvested into a single-cell suspension and assessed through flow cytometry using specific phycoerythrin (PE)- conjugated antibodies against human intercellular cell adhesion molecule (ICAM)-1. Figure 13 (SI). Exosome characterization
[0023] (A) Schematic illustration of the procedure used for generating engineered exosomes.
(B) Representative graph of nanoparticle tracking analysis (NTA) demonstrating similar size distributions in diluted samples of Exo-Naive, Exo-mCherry and Exo-srlKB.
Figure 14 (S2). Suppressive effects of Exo-srlKB in mouse models of sepsis [0024] (A) Changes in body temperature were monitored after wild-type and LPS-treated
C57BL/6 mice were injected with the indicated exosomes. **p<0.01 compared with the PBS- treated LPS group. (B) Representative images of the renal cortex area in sections from C57BL/6 (upper panels) and BALB/c (lower panels) mice treated with LPS and PBS (left), Exo-Naive (middle), and Exo-srlKB (right). Normal brush border of proximal tubules (*) or loss of brush border (o); chromatin condensation (red arrows); denuded basement membrane (red arrow heads); vacuolization (blue arrows); scale bar, 100 mM. (C) Comparison of the number of infiltrated Ly6G+ cells in each organ between the Exo-Naive and Exo-srhcB-treated groups of LPS-induced sepsis mice. (D) Levels of IL-10 were measured in the plasma of PBS-, Exo-Naive- , and Exo-srhcB-treated septic mice. NS, not significant.
Figure 15 (S3). The biodistribution of exosomes in mice with and without LPS challenge
[0025] (A) Analysis of mCLING-labeled exosomes detected in the kidney using an intravital imaging system after intravenous administration of LPS.
Figure 16 (S4). Suppressive effects of EXO-SI IKB on the LPS-induced inflammatory response in HUVECs
[0026] (A) HUVECs (7x104 cells) were cultured with Exo-srhcBs lxl O7 particles for 24 h and then stimulated with 36 ng/ml LPS. The supernatants were collected after 24 h and assayed the production of IL-8 (left) and MCP-1 (right), respectively. JSH-23 (50 DM) was used as the positive control. ***/;<0.001 compared with dimethyl sulfoxide (DMSO). #p< 0.05 compared with cells treated only with LPS. (B) HUVECs were pretreated with Exo-srhcBs for 24 h, followed by 36 ng/ml of LPS stimulation for 1 h. Western blotting was used to assess protein levels of p65 and IkBa. GAPDH and histone H3 (HH3) were used as endogenous controls for the cytosol cell lysate and nuclear fraction, respectively (left). The binding of nuclear NF-KB to its target DNA sequence, 5'-GGGACTTTCC-3', was measured through enzyme-linked immunosorbent assay (ELISA) (right).
Figures 17-20. Protective effect of Exo- srli Bs on lung injury in LPS-induced sepsis [0027] Figure 17 depicts H&E staining showing (1) Alveolar; (2) Capillary; (3) Neutrophil;
(4) Alveolar macrophages; (5) Congestion; (6) Hemorrhage; (7) Nectrosis.
[0028] Figure 18 depicts survival curve of sham, naive exosome, and Exo-srlxB-treated septic mice.
[0029] Figure 19 depicts the level of TNF-a in the plasma of Exo-srhcB-treated septic mice. [0030] Figure 20 depicts H&E staining showing (1) central vein; and (2) infiltration of immune cells.
DETAILED DESCRIPTION OF THE INVENTION
[0031] The present disclosure provides for EXPLOR technology (Yim et ah, Nature Communications 7, 12277 (2016), which was used to load srIi<B into exosomes and systemically deliver Exo-srl KB, in order to assess their effect on the course of ischemic AKI. Human embryonic kidney (HEK) 293 T cell lines that produced two recombinant proteins, CIBN-EGFP- CD9 and srlKB-mCherry-CRY2, were used for producing srlKB-containing exosomes (Exo- srlxB), by inducing the transient docking of CRY2 and CIBN using blue light illumination (Figure 1 A). Control exosomes (Exo-Naive) were generated from intact HEK293T cells. The results confirmed that optogenetically engineered exosomes could be utilized as therapeutics, and that Exo-srlKB could alleviate IR-induced kidney damage through immunologic response modulation and apoptosis.
[0032] The terms “a,” “an,” “the” and similar referents used in the context of describing the disclosure (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein is intended merely to better illuminate the disclosure and does not pose a limitation on the scope of the disclosure otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the disclosure.
[0033] As used herein, the term “about” means modifying, for example, lengths of nucleotide sequences, degrees of errors, dimensions, the quantity of an ingredient in a composition, concentrations, volumes, process temperature, process time, yields, flow rates, pressures, and like values, and ranges thereof, refers to variation in the numerical quantity that may occur, for example, through typical measuring and handling procedures used for making compounds, compositions, concentrates or use formulations; through inadvertent error in these procedures; through differences in the manufacture, source, or purity of starting materials or ingredients used to carry out the methods; and like considerations. The term “about” also encompasses amounts that differ due to aging of, for example, a composition, formulation, or cell culture with a particular initial concentration or mixture, and amounts that differ due to mixing or processing a composition or formulation with a particular initial concentration or mixture. Whether modified by the term “about” the claims appended hereto include equivalents to these quantities. The term “about” further may refer to a range of values that are similar to the stated reference value. In certain embodiments, the term “about” refers to a range of values that fall within 50, 25, 10, 9, 8,7, 6, 5,4, 3, 2, 1 percent or less of the stated reference value.
[0034] As used herein, the term "subject" refers to a human or any non-human animal (e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate).
AKI
[0035] The immune response according to NF-KB signal transduction may be involved in AKI, and Exo-srl KB may exert an effect of improving kidney damage in the kidney damage caused by sepsis. In some examples herein, it was evaluated whether administration of the inhibitor could alleviate the process of ischemic AKI. In the examples herein, SrlxB was delivered to mice before and after IRI surgery using the EXPLOR technique, and the results were compared with the control group. As a result, it was confirmed that the Exo-srl kB-treated mice exhibited lower levels of serum BUN and creatinine compared to the control group, thereby exerting prophylactic and therapeutic effects from IR-induced AKI. On the other hand, as a result of checking whether the administration of Exo-srlxB locally affects the NF-KB signal of the kidney, Exo-srlxB was found to significantly reduce the nuclear translocation and nuclear binding activity of NF-KB compared to the control group. The NF-KB pathway may play an important role in the immune response process. As a result of comparing the gene expression of Pan-inflammatory cytokines/chemokines in the Exo-srh<B treatment group and the control group, IL-Ib, IL-6, Significant reductions in a number of inflammatory mediators were identified, including CCL2, CCL5, CXCL2 and TNF-a. Finally, in the examples herein, flow cytometric analysis was conducted to compare whether NF-KB inhibitor treatment affects the renal immune cell population, and the frequency of multiple immune cell populations including neutrophils, monocytes/macrophages and T cells was observed to be significantly reduced.
[0036] IRI may induce an inflammatory cascade and oxidative stress, causing a cytokine storm, leading to apoptosis and structural damage to adjacent tissues. The NF-KB signaling pathway may regulate cell survival and inflammation in a rapid response to IRI stimulation, specifically, high levels from damaged cells during the early stages of hypoxia/reperfusion. For example, a number of endogenous factors such as mobility group box 1 (HMGB1), heat shock proteins (HSPs) and pathogen-/damage associated molecular patterns (P AMPs/DAMPs) are released. These endogenous molecules may stimulate Toll-like Receptor (TER) and Pattern Recognition Receptor (PRP), such as IF-1R. TER and IL-1R share the same intracellular domain and phosphorylate the inhibitory protein IKB residue and activate IKB kinase (IKK), which ultimately leads to degradation of IKB by the proteasome. This process allows a heterodimer (e.g., p50/p65) to move from the cytoplasm to the nucleus, which binds to DNA and promotes transcription of inflammatory mediators including TNF-a, IL-1, IL-6 and IL-8, which further accelerate NF-KB signaling. This pro-inflammatory cascade may regulate the surrounding microenvironment by inducing apoptosis and promoting leukocyte migration/activation in renal duct cells exposed to IRI. The ischemic AKI model of the present disclosure can also induce the activation of NF-KB signaling, which induces the expression of a number of inflammatory cytokines/chemokines genes, significantly affecting the renal immune cell population compared to the false surgical group. Recent studies include TLR antagonists, cytokine antagonists, IKK complex antagonists, proteasome inhibitors, decoy oligo deoxynucleotides (ODNs) specific to specific NF-kB complexes, and ODNs that inhibit Rel protein translation in various organs. It shows the effect of blocking NF-KB in IRI by inhibiting NF-KB signaling using a variety of pharmacological inhibitors, including. IkBa proteins having mutations at serine residues 32 and 36 are not phosphorylated and thus exist as multimers with NF-KB in the cytoplasm and are not degraded, which holds NF-KB nuclear translocation. This non-degradable IkBa, so-called super repressor IkBa (srlxB), is also known to have an anti-tumor effect by inducing apoptosis.
[0037] In the present disclosure, the therapeutic effect of Exo-srh<B is shown in a kidney IRI model representing acute nephritis. Exo-srh<B regulates the process of AKI by down-regulating the production of pro-inflammatory mediators that can limit leukocyte migration/activation, and by inducing early apoptosis of inflammatory cells, leading to early termination of the inflammatory phase. In the present disclosure, it was confirmed that the administration of Exo- srlxB only affects the frequency of the immune cell population in the kidney and does not affect the frequency of the immune cell population in the spleen. These results show that Exo-srl KB selectively inhibits cell proliferation or promotes apoptosis in kidney-resident immune cells. On the other hand, it may be interpreted that the in vivo delivery of ExosrlicB blocks the outflow of splenic immune cells to the damaged kidney. In the present disclosure, Exo-srlxB is shown to alleviate IR-induced AKI in mice by inhibiting the activity of NF-KB signal. In addition, in vivo delivery of EXO-SHKB can improve the overall inflammatory state of the kidney by reducing the expression of pro- inflammatory genes and reducing the renal immune cell population including neutrophils, monocytes/macrophages and T cells. Accordingly, in one aspect, the present disclosure relates to an exosome on which srlxB is mounted, and more particularly, to an exosome on which srlxB represented by the amino acid sequence of SEQ ID NO: 1 is mounted. Hereinafter, srlxB may be expressed as a cargo protein.
[0038] In some embodiments, the AKI described herein is Ischemia-reperfusion (IR)-induced AKI, lipopolysaccharide (LPS)-induced AKI or sepsis-induced AKI but not limited thereto. Ischemia-reperfusion (IR) injury (IRI) is a major cause of acute kidney injury (AKI). Ischemia- reperfusion (IR)-induced acute kidney injury (AKI) is a relatively common but severe medical condition that can happen when a subject suffers from systemic hypoperfusion that is followed by restoration of blood flow and re- oxygenation. The most common clinical examples include patients undergoing renal transplant or cardiac surgery and patients with sepsis, trauma, or myocardial infarction. Renal IR injury (IRI) is associated with a high morbidity/mortality and is known to induce dysfunction in other distant organs, including the heart, lung, and liver. In IR- induced AKI, hypoxia and reperfusion generate reactive oxygen species (ROS), followed by a cascade response, including cell death and inflammation, and subsequent renal failure. Immunologic responses may significantly affect renal IRI and repair. Nuclear factor (NF)-KB signaling, which controls cytokine production and cell survival, is significantly involved in IR- induced AKI; its inhibition can ameliorate ischemic AKI.
[0039] Using EXPLOR, an optogenetically engineered exosome technology, the exosomal super-repressor inhibitor of NF-KB (Exo-srlxB) was loaded into exosomes and administered toB6 WT mice before/after kidney IR surgery as shown in the examples herein. The outcome was compared with the results from a control exosome (Exo-Naive)-injected group. Exo-srlxB treatment resulted in lower levels of serum blood urea nitrogen, creatinine, and neutrophil gelatinase-associated lipocalin in post-ischemic mice than in the Exo-Naive treatment group. Systemic delivery of Exo-srlxB decreased NF-KB activity in post-ischemic kidneys, leading to reduced apoptosis levels. Post-ischemic kidneys showed decreased gene expression of pro- inflammatory cytokines and adhesion molecules with Exo-srlxB treatment as compared with the control. Exo-srlxB treatment also significantly affected post-ischemic renal immune cell populations, lowering neutrophil, monocyte/macrophage, and T cell frequencies than those in the control. Thus, the modulation of NF-KB signaling through exosomal delivery can be used as a novel therapeutic method for AKI, including IR-induced AKI.
[0040] In another embodiment of the present disclosure, the exosomes are engineered to contain a mutant form of the biological inhibitor of NF-KB, called super-repressor (SR) IKB (srlxB , SEQ ID NO:l) (N. Yim et al, Nature Communications 7, 12277 (2016).
[0041] As shown in the examples below, an optogenetically engineered exosome system (EXPLOR) was employed for loading a large amount of srlxB into exosomes in stably transfected cells. The below examples confirm that intraperitoneal injection of purified srlxB- loaded exosomes (Exo-srhcBs) attenuates mortality and systemic inflammation in septic mouse models. In a biodistribution study using exosomes labeled with fluorescence dye, Exo-srhcBs were mainly observed in the neutrophils, and in monocytes to a lesser extent, in the spleens and livers of mice. Moreover, the examples confirm that Exo-srlKB alleviates inflammatory responses in monocytic THP-1 cells and human umbilical vein endothelial cells ( HUVECs ) in response to lipopolysaccharide (LPS) or tumor necrosis factor ( TNF)-a . These results confirm that Exo-srlKB can protect against systemic inflammation and sepsis-induced death by inhibiting NF-KB activation.
[0042] The present disclosure provides a method for treating acute kidney injury (AKI) in a subject in need thereof, comprising administering an effective amount of a composition comprising an exosome comprising an NF-kB inhibitor to the subject. In some embodiments, the NF-KB inhibitor is selected from the group consisting of a NF-KB inhibitory protein, its fragment, and a mixture thereof.
[0043] The compositions are physiologically acceptable water-based solutions, or suspensions, of exosomes.
[0044] The NF-KB inhibitor is selected from an NF-KB inhibitory drug, an NF-KB inhibitory protein or its fragment, or a mixture thereof.
[0045] Preferably, the NF-KB inhibitor is selected from the group comprising super-repressor IKB, IkB-a, IkB-b, IkB-e, BCF-3, a mutant thereof, and a mixture thereof.
[0046] In another preferred embodiment of the present disclosure, the exosomes of the present disclosure contain a mutant form of the biological inhibitor of NF-KB, called super repressor (SR) IKB (SAKB, SEQ ID NO:l).
[0047] In one embodiment of the present disclosure, srhcB has Ser32 and Ser36 of IKB (SEQ
ID NO:2) replaced with Ala.
[0048] SEQ ID NO: 1 Homo sapiens, super-repressor-IkB (srhcB)
MFQAAERPQEWAMEGPRDGFKKERFFDDRHDAGFDAMKDEEYEQMVKEFQEI RFEPQEVPRGSEPWKQQFTEDGDSFFHFAIIHEEKAFTMEVIRQVKGDFAFFNFQNNFQ QTPFHFAVITNQPEIAEAFFGAGCDPEFRDFRGNTPFHFACEQGCFASVGVFTQSCTTPH FHSIFKATNYNGHTCFHFASIHGYFGIVEFFVSFGADVNAQEPCNGRTAFHFAVDFQNP DEV SFFFKCGAD VNRVTYQGY SPY QFTWGRPSTRIQQQFGQFTFENFQMFPESEDEES Y DTESEFTEFTEDELPYDDCVFGGQRLTL [0049] SEQ ID NO:2 Homo sapiens, IkB-a
MF Q A AERPQEW AMEGPRDGLKKERLLDDRHD S GLD SMKDEE YEQMVKELQEI RLEPQEVPRGSEPWKQQLTEDGDSFLHLAIIHEEKALTMEVIRQVKGDLAFLNFQNNLQ QTPLHLAVITNQPEIAEALLGAGCDPELRDFRGNTPLHLACEQGCLASVGVLTQSCTTPH LHSILKATNYNGHTCLHLASIHGYLGIVELLVSLGADVNAQEPCNGRTALHLAVDLQNP DLVSLLLKCGADVNRVTYQGYSPYQLTWGRPSTRIQQQLGQLTLENLQMLPESEDEESY DTESEFTEFTEDELPYDDCVFGGQRLTL [0050] SEQ ID NO: 3 recombinant srb B a peptide DRHDAGLDAMKDE
[0051] In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO: 1 or SEQ ID NO:2. In some embodiments, the NF- KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:l. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 86% sequence identity to SEQ ID NO:l. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 87% sequence identity to SEQ ID NO:l. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 88% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 89% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 91% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 92% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 93% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 94% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 95% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 96% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 97% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 98% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 99% sequence identity to SEQ ID NO: 1. In embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 1.
[0052] In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 86% sequence identity to SEQ ID NO:2. In some embodiments, the NF-kB inhibitor comprises an amino acid sequence having at least 87% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 88% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 89% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 91% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 92% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 93% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 94% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 95% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 96% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 97% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 98% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 99% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 2. [0053] In a certain embodiment of the present disclosure, the exosome further comprises a photo- specific binding protein. In some embodiments, the photo-specific binding protein comprises a first photo-specific binding protein and/or a second photo-specific binding protein, which interacts reversibly with each other upon irradiation. In some embodiments, the first photo- specific binding protein is conjugated to an exosome specific marker to form a first fusion protein (fusion protein I). In some embodiments, the second photo- specific binding protein is conjugated to the NF-KB inhibitory protein to form a second fusion protein (fusion protein II). In some embodiments, the fusion protein I and the fusion protein II are linked reversibly through the first photo-specific binding protein and the second photo-specific binding protein. In some embodiments, the first photo-specific binding protein is conjugated to the exosome specific marker to be located in the direction toward inside of the exosome.
[0054] In additional embodiments, the first photo-specific binding protein and the second photo-specific binding protein are selected from the group comprising CIB, CIBN, PhyB, PIF, FKF1, GIGANTEA, CRY or PHR, but not limited thereto.
[0055] In additional embodiments, the first photo-specific binding protein is CIB or CIBN and the second photo-specific binding protein is CRY or PHR; or the first photo-specific binding protein is CRY or PHR and the second photo-specific binding protein is CIB or CIBN.
[0056] In additional embodiments, the first photo-specific binding protein is PhyB and the second photo-specific bind protein is PIF or the first photo-specific binding protein is PIF and the second photo-specific binding protein is PhyB.
[0057] In additional embodiments, the first photo-specific binding protein is GIGANTEA and the second photo-specific binding protein is FKFl or the first photo-specific binding protein is FKFl and the second photo-specific binding protein is GIGANTEA.
[0058] In some embodiments, the exosome specific marker is selected from the group comprising CD9, CD63, CD 81 or CD 82, but not limited thereto. In additional embodiment, the exosome specific marker is CD9. In additional embodiment, the exosome specific marker is CD63. In additional embodiment, the exosome specific marker is CD81. In additional embodiment, the exosome specific marker is CD82.
[0059] In some embodiments, the exosome has a diameter of between about 50 nm and about 200 nm. In some embodiments, the exosome has a diameter of between about 50 nm and about 75 nm. In some embodiments, the exosome has a diameter of between about 75 nm and about 100 nm. In some embodiments, the exosome has a diameter of between about 100 nm and about
125 nm. In some embodiments, the exosome has a diameter of between about 125 nm and about
150 nm. In some embodiments, the exosome has a diameter of between about 150 nm and about
175 nm. In some embodiments, the exosome has a diameter of between about 175 nm and about
200 nm. In embodiments, the exosome has a diameter of between about 50 nm and about 150 nm. In some embodiments, the exosome has a diameter of about 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150 or more and/or of about 75, 80, 85,
90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185,
190, 195, 200 or less. [0060] Without listing all details, the contents of U.S. Patent No. 10,702,581 and Korean Patent No. 10-2100420 are incorporated herein by reference to provide the compositions and method of preparing the exosomes containing NF-KB inhibitors of the present disclosure.
[0061] The present disclosure also provides sustained release of the therapeutically effective drugs, while conventional sustained effects required repeated dosing of the therapeutics.
[0062] The present disclosure also further provides pharmaceutical compositions containing extracellular vesicles (exosomes) carrying NF-KB inhibitors and a pharmaceutically acceptable carrier or carriers.
[0063] Despite recent noteworthy advancements in understanding the pathophysiology of IR- induced AKI, no drug with proven clinical efficacy and safety to treat ischemic AKI has been developed; thus, the associated mortality and morbidity levels are significant. Since NF-KB signaling is deeply involved in the course of IR- induced AKI, it was assessed whether the systemic delivery of the NF-KB inhibitor using exosomes could alleviate the course of ischemic AKI. Through optogenetically controlled biogenesis of exosomes using EXPLOR technology, Exo-srlKB was delivered to mice either before/after IRI surgery and compared outcomes with those of the control group. The results showed that the mouse group receiving Exo-srlxB treatment was protected from IR-induced AKI; it showed better biochemical and histological outcomes than the control group. Systemic delivery of Exo-srhcB suppressed NF-KB signaling in post-ischemic kidneys, which led to a decreased expression of pro-inflammatory cytokines/chemokines and adhesion molecules and alleviation of apoptosis. Finally, NF-KB treatment affected the renal immune cell population and a significant reduction in frequencies of multiple immune cell populations, including neutrophils, monocytes/macrophages, and T cells, through flow cytometric and immunofluorescent analysis was observed.
[0064] During the early course of IRI, pro-inflammatory cascade and oxidative stress results in NF-KB pathway activation, subsequently leading to the regulation of cell survival and inflammation. Multiple endogenous factors are released during the early stages of hypoxia/reperfusion, including high mobility group box 1, heat shock proteins, and pathogen- /damage-associated molecular patterns, and these molecules stimulate pattern recognition receptors, such as Toll-like receptors (TLRs) and the interleukin-1 receptor (IL-1R). This process subsequently activates IKB kinase (IKK), which leads to IKB degradation by proteasomes. Next, heterodimers (e.g., p50/p65) translocate from the cytosol to the nucleus, where they promote the transcription of inflammatory mediators, including TNF-a, IL-1, IL-6, and IL-8, which in turn further promote NF-KB signaling. This proinflammatory cascade modulates surrounding microenvironments, by inducing apoptosis in renal tubular cells subjected to IRI, and promoting leukocyte migration/activation. The instant experimental ischemic AKI model could reproduce the activation of NF-KB signaling following renal IRI, leading to the increased expression of multiple inflammatory cytokines/chemokines and tissue apoptosis, which significantly affect renal immune cell populations.
[0065] Recent experimental studies have shown more detailed evidence regarding the effect of NF-KB blockade on IRI, using selective pharmacologic inhibitors of NF-KB signaling, including TLR antagonists, cytokine antagonists (33), IKK complex antagonists, proteasome inhibitors, and decoy oligodeoxynucleotides specific to a particular NF-KB complex, in various organs. srh<B is a non-degradable IkBa protein with mutations at serine residues 32 and 36. This protein prevents NF-KB nuclear translocation and subsequent NF-KB signaling. srh<B has proven protective effects in the lung IRI model, as it decreases neutrophil infiltration and pulmonary edema, and is also known to have anti-tumor effects, because it decreases chemoresistance. However, the effects of srh<B treatment in the course of kidney injury has not been fully evaluated. By using bioengineered exosomes as a vector, Choi et al. recently showed the therapeutic benefits of srh<B treatment in the septic AKI model. The below examples also demonstrate the protective effects of Exo-srlKB treatment in the renal IRI model, expanding the therapeutic potential of srh<B delivery through exosomes in various types of AKI.
[0066] Inflammatory process modulation has been a major therapeutic consideration in the field of ischemic AKI. Splenectomy as well as the use of the anti-TNF-a agent, infliximab, showed protective effects in the experimental ischemic AKI model, by lowering inflammatory cytokine expression and macrophages/monocyte accumulation. Inhibiting the migration of leukocytes and macrophages also preserved renal function and alleviated cell death the provided examples have shown that Exo-srlKB treatment downregulates the expression of pro- inflammatory cytokines/chemokines and adhesion molecules, reduces apoptosis, and alleviates multi-lineage immune cell accumulation in IR-injured kidneys. These results suggest that Exo- srh<B modifies the course of ischemic AKI through pro-inflammatory cascade downregulation, limiting subsequent leukocyte migration/activation and preventing apoptosis. [0067] Exosomes can have several benefits over the use of other types of vectors as conveyors. First, EVs are naturally protected from degradation during circulation and are non- immunogenic when used autologously. Exosomes can also overcome natural barriers such as the blood-brain barrier, through their intrinsic cell targeting properties. Additionally, exosomes use intrinsic mechanisms of recipient cells in the course of uptake, intracellular trafficking and the final delivery of cargoes to target cells. While maintaining these properties, EXPLOR technology made it possible to significantly increase intracellular levels of cargo proteins through controllable, reversible detachment from exosomes. There were several technical obstacles during the intracellular delivery of therapeutic proteins, including low purification efficiency, induction of immune responses against the host, and limited cytosolic delivery, and this novel exosomal delivery method using EXPLOR has significantly improved exosomal loading capacity, delivery efficiency, and purity, as compared with those of previous technologies. The below examples showed that efficient delivery of Exo-srlxB using EXPLOR technology could bring significant improvement in the renal outcome after experimental IRI.
[0068] Systemic Exo-srlxB treatment may only affect immune cell population frequencies in the kidney but not those in the spleen. This can be attributed to Exo-srl KB either selectively acting on renal resident immune cells alleviating proliferation or promoting apoptosis, or the systemic delivery of Exo-srlxB blocking the efflux of splenic immune cells into injured kidneys. Kidney resident immune cells and immune cells from lymphoid organs are known to have distinct origins, phenotypes, and functional characteristics. Further studies using chimeric mice or in vivo imaging would be able to differentiate resident renal immune cells from circulating immune cells, and provide more information about immune cell populations affected by systemic Exo-srlitB treatment.
[0069] Using the exosome as a carrier to deliver a cargo of active therapeutic agents is a safe and efficient method for delivering srlxB into the IR-induced AKI model. Exo-srlitB treatment alleviates IR-induced AKI in mice by down-regulating NF-KB signaling and ameliorating inflammation and apoptosis in the ischemic injured kidney. The direct intracellular delivery of immunosuppressive proteins into target cells using exosomes is a promising therapeutic tool in IR-induced AKI and the therapeutic potential of Exo-srlxB in human kidney IRI should be explored further. DISEASES CAUSED BY SEPSIS
[0070] Sepsis, which is accompanied by a large number of organ failures and excessive inflammatory reactions, has complex pathophysiological features, and it is not easy to develop a therapeutic agent that can effectively treat all symptoms. Sepsis is initiated through the strong activation of the innate immune system and is mediated by the activation of the pattern recognition receptor (PRR) by pathogens, leading to activation of the complement system, coagulation system and vascular endothelium and thus the difficulty to normal homeostasis. [0071] As shown in the examples herein, the effect of Exo-srlxB was capable of resolving the inflammatory response and reducing organ damage through inhibition of NF-KB activity in a sepsis mouse model. srhcB is a non-degradable form of IkBa that prevents NF-KB from entering the nucleus and acting as a transcription factor. For example, in order to generate an immunosuppressive exosome loaded with srlxB, a recent technology capable of mounting a functional protein on an exosome was utilized. By delivering Exo-srlxB to the whole body in a sepsis mouse model (e.g., systemic delivery), the examples herein confirmed that exosomes are delivered to neutrophils and macrophages, which are key players of the sepsis inflammatory response. In addition, in both mouse sepsis models of the present examples, Exo-srlxB treatment significantly reduced pro- inflammatory markers such as TNF-a, IF-Ib and IF-6, but did not decrease the anti-inflammatory cytokine IF- 10 Even when exosomes were injected 1 hour after EPS injection, a significant therapeutic effect of Exo-srlxB was observed. Thus, the treatment with Exo-srlxB may also be applied to the early stages of sepsis before the transition to an immune suppression state.
[0072] Pro- inflammatory cytokines may play an important role in acute kidney injury (AKI) induced by sepsis. In the mouse model with AKI caused by CEP surgery or LPS injection in the examples herein, as a result of histopathological examination, it was confirmed that the glomerular structure was destroyed, renal tubular epithelial cells were degenerated, and severe intracellular edema and congestion occurred in the renal tubules in the sepsis-induced group. However, Exo-srlxB administration reduced the severity of lesions, kidney damage, and invasion of inflammatory cells.
[0073] Tissue infiltration of neutrophils may be a major process in sepsis. For example, in some studies targeting specific adhesion molecules to deplete neutrophils and inhibit recruitment of neutrophils can reduce tissue damage in sepsis It has been reported that it can be protected. TNF-a and IL-Ib are major mediators for the expression of chemokines such as MCP-1 and IL- 8, which are important chemoattractants for neutrophils and monocytes/macrophages during the inflammatory response. These chemokines initiate local invasion of monocytes/macrophages after activation. However, in HUVECs, Exo-srlKB was shown to attenuate the release of FPS- induced MCP-1 and IL-8, thereby inhibiting monocyte/macrophage invasion. These data are consistent with results obtained from in vivo studies herein. In the present examples, Exo-srhcB treatment significantly reduced neutrophil infiltration in the spleen, kidney, lung and liver of the sepsis animal model. Considering the important role of neutrophils in the pathophysiology of sepsis, the tissue protective effect of Exo-srhcB may be related to at least a reduction in invasion of neutrophils in the kidney.
[0074] In some embodiments, the spleen shows higher levels of neutrophils compared to other organs such as kidney and liver. Pro-inflammatory cytokines may occur in the spleen and affect other organs. Most of the genes encoding pro-inflammatory cytokines may be under the control of NF-KB, one of the important transcription factors in the septic inflammatory response caused by LPS. Expression of the NF-KB response reporter protein according to the presence or absence of Exo-srhcB treatment was investigated in the examples herein, and the translocation of p65 was confirmed. Exo-srlKB inhibited NF-KB signal activation by blocking nuclear translocation of the NF-KB subunit p65.
[0075] In some embodiments, exosomes may be used as a mechanism for therapeutic delivery of immunosuppressive proteins in a sepsis mouse model. Exosomes may be excellent carriers capable of delivering srhcB into cells, providing a new option for the treatment of sepsis. A method of directly delivering an immunosuppressive protein into a target cell in vivo has not been established before, but Exo-srhcB according to the present disclosure acts as an inhibitor of NF-KB, thereby inhibiting the expression of inflammation-related genes and directly inflammatory response. The examples herein shows it can be suppressed, thereby alleviating the pro-inflammatory cytokine storm of sepsis and the resulting organ damage.
[0076] Recent studies in animal models of sepsis induced either by lipopoly saccharide (EPS; endotoxin model) or cecal ligation and puncture (CEP, polymicrobial model) have demonstrated that NF-KB inhibitors with diverse chemical properties and mechanisms of action protect animals from septic lethality. Although more than 700 inhibitors of NF-KB have been reported, no NF-KB blocker has been approved for human use to date. Various steroids and non- steroid anti-inflammatory drugs have been found to block NF-KB, but their effects are highly pleiotropic and lack specificity. Novel therapeutic strategies aimed at specific inhibition of key elements in the NF-KB activation pathway have been under development in recent years, and expectations are high regarding their potential efficacy as sepsis treatments and treatments for one or more diseases caused by sepsis. Genetic constructs expressing an engineered IkBa protein without sites for phosphorylation, super-repressor IKB (srlicB), have also been used. IkBa mutation at specific phosphorylation sites (Ser32 and Ser36 replaced to Ala) results in a dominant active form of IkBa with a prolonged half-life. These srlxBs lead to a stable cytoplasmic pool of IkBa, thereby preventing nuclear NF-KB activation.
[0077] Here, exosomes were utilized as a therapeutic conveyor to deliver srlxB to a therapeutic target. Exosomes have been recognized as potent therapeutic vehicles for transferring various proteins and regulatory genes to target cells. They function as non-immunogenic nanovesicles, and can protect their cargo from serum proteases and immune responses. Loading of soluble proteins into exosomes has been enabled by a technology called EXPLOR, which exploits the natural exosome biogenesis process and reversible protein-protein interactions controlled by optogenetics. To load specific target proteins into exosomes, a HEK293T cell line was generated that stably expresses two recombinant proteins, CIBN-EGFP-CD9 and srlxB- mCherry-CRY2 (Figure 1 A). Consistent with previous studies, EXPLOR technology was used to produce srlxB-loaded exosomes. The srlicB loading exosomes were designated as EXO-SHKB, and exosomes generated from intact HEK293T cells as Exo-Naive. By applying EXO-SAKB to sepsis models for therapeutic purposes, sepsis-induced organ injury was ameliorated and inhibited the secretion of pro-inflammatory cytokines, thereby improving the overall survival of sepsis patients.
[0078] The present disclosures provides for methods for treating a disease caused by sepsis in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-KB inhibitor to the subject.
[0079] In embodiments, the disease is selected from the group consisting of pneumonia, cytokine storm syndrome, respiratory distress syndrome, and organ failure but not limited thereto. In some embodiments, the disease is pneumonia. In some embodiments, the disease is cytokine storm syndrome. In some embodiments, the disease is respiratory distress syndrome. In some embodiments, the disease is organ failure. [0080] In embodiments, the NF-KB inhibitor is selected from the group consisting of a NF-KB inhibitory protein, its fragment, and a mixture thereof. In embodiments, the NF-KB inhibitor is selected from the group consisting of super-repressor (SR)-IKB (srlicB), IkB-a, IkB-b, IkB-e, BCL-3, a mutant thereof, and a mixture thereof. In embodiments, the NF-KB inhibitor is super repressor (SR)-IKB (srlicB).
[0081] In embodiments, the composition of is administered via oral, transdermal, intraperitoneal, intravenous, intramuscular, subcutaneous, or mixed routes. In some embodiments, the composition is administered via oral routes. In some embodiments, the composition is administered via transdermal routes. In some embodiments, the composition is administered via intraperitoneal routes. In some embodiments, the composition is administered via intravenous routes. In some embodiments, the composition is administered via intramuscular routes. In some embodiments, the composition is administered via subcutaneous routes. In some embodiments, the composition is administered via mixed routes.
[0082] In embodiments, the method further comprises administering an anti-inflammatory agent to the subject.
[0083] In embodiments, the exosome comprising a NF-KB inhibitor further comprises a photo- specific binding protein. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:l or SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 86% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 87% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 88% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 89% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 91% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 92% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 93% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 94% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 95% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 96% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 97% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 98% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 99% sequence identity to SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 1.
[0084] In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 86% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 87% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 88% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 89% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 91% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 92% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 93% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 94% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 95% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 96% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 97% sequence identity to SEQ ID NO:2. In some embodiments, the NF-KB inhibitor comprises an amino acid sequence having at least 98% sequence identity to SEQ ID NO:2. In some embodiments, the NF-kB inhibitor comprises an amino acid sequence having at least 99% sequence identity to SEQ ID NO:2. In embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 1. In some embodiments, the NF-KB inhibitor is represented by SEQ ID NO: 2.
[0085] In some embodiments, the photo-specific binding protein is a first photo-specific binding protein and/or a second photo-specific binding protein. In embodiments, the first photo specific binding protein is conjugated to an exosome specific marker to form fusion protein I; and the second photo-specific binding protein is conjugated to the NF-KB inhibitor to form fusion protein II. In embodiments, the fusion protein I and the fusion protein II are linked reversibly through the first photo-specific binding protein and the second photo-specific binding protein. In embodiments, the first photo-specific binding protein is conjugated to an exosome specific marker to be located in the direction toward inside of the exosome.
[0086] In some embodiments, the first photo-specific binding protein and the second photo specific binding protein are selected from the group consisting of CIB, CIBN, PhyB, PIF, FKF1, GIGANTEA, CRY and PHR. In embodiments, the first photo-specific binding protein is CIB and the second photo-specific binding protein is CRY. In some embodiments, the first photo specific binding protein is CIBN and the second photo-specific binding protein is CRY. In some embodiments, the first photo-specific binding protein is CIB and the second photo-specific binding protein is PHR. In some embodiments, the first photo-specific binding protein is CIBN and the second photo-specific binding protein is PHR.
[0087] In some embodiments, the first photo-specific binding protein is CRY or PHR and the second photo- specific binding protein is CIB or CIBN. In some embodiments, the first photo specific binding protein is CRY and the second photo-specific binding protein is CIB. In some embodiments, the first photo-specific binding protein is CRY and the second photo-specific binding protein is CIBN. In some embodiments, the first photo-specific binding protein is PHR and the second photo-specific binding protein is CIB. In some embodiments, the first photo specific binding protein is PHR and the second photo-specific binding protein is CIBN.
[0088] In some embodiments, the first photo-specific binding protein is PhyB and the second photo- specific binding protein is PIF. In embodiments, the first photo-specific binding protein is PIF and the second photo-specific binding protein is PhyB. [0089] In some embodiments, the first photo-specific binding protein is GIGANTEA and the second photo-specific binding protein is FKF1. In embodiments, the first photo-specific binding protein is FKF1 and the second photo- specific binding protein is GIGANTEA.
[0090] In some embodiments, the exosome specific marker is selected from the group consisting of CD9, CD63, CD81 and CD82. In some embodiments, the exosome specific marker is CD9. In some embodiments, the exosome specific marker is CD63. In some embodiments, the exosome specific marker is CD81. In some embodiments, the exosome specific marker is CD82. [0091] In some embodiments, the exosome has a diameter of between about 50 nm and about 200 nm. In some embodiments, the exosome has a diameter of between about 50 nm and about 75 nm. In some embodiments, the exosome has a diameter of between about 75 nm and about 100 nm. In some embodiments, the exosome has a diameter of between about 100 nm and about
125 nm. In some embodiments, the exosome has a diameter of between about 125 nm and about
150 nm. In some embodiments, the exosome has a diameter of between about 150 nm and about
175 nm. In some embodiments, the exosome has a diameter of between about 175 nm and about
200 nm. In embodiments, the exosome has a diameter of between about 50 nm and about 150 nm. In some embodiments, the exosome has a diameter of about 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150 or more and/or of about 75, 80, 85,
90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200 or less.
[0092] In some embodiments, the composition is a pharmaceutical composition further comprising a physiologically acceptable carrier or carriers.
EXPLOR® Technology
[0093] Recently, exosomes received significant attention as novel bio-carriers for gene/drug delivery. Exosomes are extracellular vesicles (EVs) that play an important role in cell-to-cell communication, by transferring bioactive materials to recipient cells or affecting signaling pathways of target cells. Exosomes are easier to store and exhibit greater stability. Exosomes have a high capacity to overcome biological barriers, and can carry surface molecules targeting specific cell types, thus causing fewer off-target effects. However, in the clinical application of exosomes, it was a significant challenge to obtain high amounts of pure exosomes and load soluble proteins into exosomes. Using “exosomes for protein loading via optically reversible protein-protein interactions” (EXPLOR), a novel, optogenetically engineered exosome technology developed by Yim etal, significant advancements in the production efficiency and biological compatibility of exosomes were achieved. This EXPLOR technology has been recently adopted in the experimental sepsis model by Choi et al. ; they delivered exosomes containing the super-repressor IKB (srlxB) to mice. This was a non-degradable form of inhibitor of kappa B (IKB) that prevented the nuclear translocation of NF-KB. Their study showed that treatment with Exo-srlKB ameliorated the systemic pro- inflammatory response and subsequent organ dysfunction in the mouse sepsis model.
EXAMPLES
[0094] Animals: Male C57BL/6J mice were bred under specific pathogen-free conditions at the central animal facility of the Yonsei Medical Center. 6-7-week-old male mice were used for all experiments. Additionally, C56BL/6, C57BL/6N, BALB/c mouse were purchased from Orientbio (Seong-Nam, Republic of Korea). LysMGFP/+ mice intrinsically expressing GFP in neutrophils and macrophages were generously provided by Dr. M. Kim (University of Rochester, Rochester, NY, USA).
[0095] Exosome isolation: The HEK293T cell line, which expresses two recombinant proteins, CIBN-EGFP-CD9 and srlKB-mCherry-CRY2, were used to produce srhcB-loaded exosomes. Intact HEK293T cells were used for producing Exo-Naive. Stable cells were seeded into T175 flasks for 24 h at 37 °C. After removing the medium, cells were washed and resuspended with exosome-depleted medium. Cells were then exposed to continuous blue light illumination from a 460-nm light emitting diode in a CO2 incubator. After 72 h, cell culture supernatants were harvested and centrifuged, to remove cells and debris, and filtered through a 0.22- pm poly ether sulfone filter; exosomes were isolated using a combination of tangential flow filtration and size-exclusion chromatography. In some examples, to produce Exo-srhcB, the stable cells were seeded into T175 flasks. After 1 day, the medium was carefully removed, the cells were rinsed with PBS, and exosome-depleted medium was added. Then, the cells were exposed to continuous blue light illumination from a 460-nm light emitting diode in a CO2 incubator. After 72 h, the cell culture supernatant was harvested and centrifuged at 1,000 g for 15 min to remove cells and cell debris, and then filtered through a 0.22-um polyethersulfone (PES) filter to remove large particles. The exosomes were isolated using molecular weight cutoff (MWCO)- based membrane filtration. The isolated exosomes were purified through SEC.
[0096] Transmission Electron Microscopy: To observe the morphology of EVs via TEM, 5 mΐ of EVs were absorbed to carbon-coated copper grids (Electron Microscopy Sciences, Hatfield, PA) for 15 s. After removing excess liquids, samples were negatively stained with 2% uranyl acetate (Electron Microscopy Sciences). TEM images were obtained using a Tecnai G2 Retrofit electron microscope (FEI company, Hillsboro, OR) operating at 200 kV. Extracellular vesicles (EVs) were evaluated morphologically through negative staining. First, 5 mΐ of EV suspended in PBS was loaded onto glow-discharged carbon-coated copper grids (Electron Microscopy Sciences, Hatfield, PA, USA). After sample adsorption for 3-5 s, the grid was blotted with filter paper and stained with 2% uranyl acetate (UA). Next, samples were dried for 20 s using a dryer. EVs were viewed with Tecnai G2 Retrofit (FEI Company, Hillsboro, OR, USA) at a voltage of 200 kV.
[0097] Nanoparticle tracking analysis: The particle number and size distribution of EVs were measured by NTA using the Zetaview instrument (PMX120, Particle Metrix, Germany) with an embedded 488 nm laser. Samples were diluted (serum 1:100-1:10000) in particle-free PBS according to the manufacturer’s instructions. Samples were analyzed under constant flow conditions at 25 °C with a camera level of 78 and shutter of 80. Size distributions determined by NTA corresponded to hydrodynamic diameters of EVs in the suspension. Particle number was calculated from the rate of Brownian motion and size was determined using the two-dimensional Stokes-Einstein equation based on the velocity of particle movement. Certain samples were diluted in 0.2-mih filtered PBS between 1 : 100 and 1 : 10,000. EV concentrations were measured based on counts of 50-200 particles per frame. For each measurement, two cycles of scanning at 11 cell positions were performed with the following settings: focus, autofocus; camera sensitivity for all samples, 78.0; shutter, 70; and cell temperature, 25°C. The EV concentration was expressed in particle numbers per ml (pn/ml).
[0098] Immunoblotting: Cells were lysed in RIPA buffer containing the Halt™ Protease and Phosphatase Inhibitor Cocktail (100X) (Thermo Fisher Scientific, Waltham, MA) and kidneys were lysed in Triton X-100 lysis buffer. Lysates were centrifuged at 9,000-16,000 x g for 10-20 min at 4 °C and supernatants were stored at -70 °C for subsequent immunoblotting. Protein concentrations were measured using the BCA assay (Pierce™ BCA Protein Assay, Thermo Scientific™). Protein extracts were lysed in Laemmli sample buffer (Bio-Rad, Hercules, CA) and heated for 5 min at 100 °C. Proteins were electrophoresed in an acrylamide-denaturating SDS-polyacrylamide gel and transferred onto a nitrocellulose (NC) membrane. Transfer membranes were incubated in blocking buffer A (PBS, 0.1% Tween-20, and 5% nonfat milk) for 1 h at 22 °C, and then incubated with primary antibodies against srb B (customized antibody, Abclon, Seoul, Korea), mCherry, Alix, TSG101, GM130, calnexin (Abeam, Cambridge, UK), CD9, CD63 (SBI, Tokyo, Japan), GAPDH (Santa Cruz Biotechnology, Dallas, TX), prohibitin (NOVUSBIO, Centennial, CO), nucleoporin p62 (BD bioscience, San Jose, CA), NF-KB (Clone 8242S, Cell signaling technology, Danvers, MA), Lamin B1 (Clone abl33741), ICAM-1 (Clone AF796, R&D Systems), cleaved caspase-3 (Clone 9661L, Cell signaling technology), cleaved PARP (Clone 9544S, Cell signaling technology), and b-actin (A5441, Sigma- Aldrich) overnight at 4 °C. After washing and incubating membranes with specific secondary antibodies, they were developed using chemiluminescent agents [Clarity and Clarity Max ECL Western Blotting Substrates (Bio-Rad) or West-Q Pico Dura ECL solution (GenDEPOT, Katy, TX)]. Image J software (NIH) was used to measure the relative optical densities of bands for each group.
[0099] Mouse renal ischemia-reperfusion model: Mice were anesthetized via the intraperitoneal administration of a tiletamine/zolazepam (Zoletil®)-xylazine (Rompun®) mixture (1 mg/kg), and placed on a heated surgical pad (37 °C). Bilateral IRI was performed using the dorsal approach. Renal pedicles were dissected, and a micro clamp (Jeungdo Bio & Plant, Seoul, South Korea) was placed on each renal pedicle for 30 min. Sham animals received anesthesia and bilateral dorsal incisions alone. During the procedure, mice were hydrated with 1 ml warm normal saline, to prevent dehydration. After 30 min of ischemia, both clamps were removed and blood flow restoration was confirmed before closing wounds with the Autoclip® Wound Closure System (Harvard apparatus, Holliston, MA). The animals were allowed to recover with free access to food and water. Mice were sacrificed 24 or 48 h after IRI surgery. Serum, spleens, and kidneys were collected from each mouse for further analysis. Serum BUN and creatinine levels were analyzed using the Cobas C502 automatic analyzer (Roche diagnostics, Basel,
Switzerland).
[0100] Enzyme-linked immunosorbent assay (ELISA): Serum NGAL levels were measured using mouse ELISA kits, according to the manufacturers’ protocols (Mouse Lipocalin-2/NGAL Quantitative ELISA kit) (R&D Systems, Minneapolis, MN) after diluting samples 1:100-1000 times.
[0101] The TNF-a, IL-8 and MCP-1 levels in supernatants collected from THP-1 and HUYEC cultures were measured using commercial ELISA kits (R&D Systems, Minneapolis, MN, USA), and NF-KB activity in HUVECs was measured using the Trans AM NF-KB p65 kit (Active Motif, Carlsbad, CA, USA). TNF-a, CCL-4/macrophage inflammatory protein (MIP)- 1 b, IL-6, and IL-1 b levels in mouse plasma collected through cardiac puncture at the indicated time points after the high-grade CLP procedure were measured using the Mouse Magnetic Luminex eScreening Assay kit (R&D Systems). The assay was performed according to the manufacturer’s instructions. All samples and standards were assayed in duplicate using the Luminex 200TM System (Merck Millipore, Darmstadt, Germany). To measure serum TNF-a, IL-Ib, IL-6, and CCL4 at the indicated time points, blood samples were collected through cardiac puncture after the LPS endotoxemia procedure, allowed to clot for 2 h at room temperature, and then centrifuged at 2,000 g for 20 min at 4 °C.
[0102] NE-PER nuclear and cytoplasmic extraction: Kidneys were collected, cut into small pieces, washed with PBS, and homogenized with beads in certain volumes of cytoplasmic extraction reagent (CER) I, obtained from the NE-PER Nuclear and Cytoplasmic Extraction Reagent kit (ThermoFisher, Waltham, MA), according to the kidney weight. After 15 s of vigorous vortexing and 10 min of incubation on ice, CER II was added to each kidney sample, followed by incubation on ice for 1 min and centrifugation at 16,000 x g for 5 min. The supernatant was collected as a cytoplasmic extract, and the insoluble fraction was suspended in nuclear extraction reagent. After centrifuging for 10 min at 16,000 x g, all supernatants were collected and used as nuclear extracts.
[0103] Histopathologic assessment of kidney injury score and immunohistochemistry: Five micrometer-thick sections of formalin-fixed and paraffin- embedded kidney tissue samples were processed via hematoxylin-eosin (HE) and IHC staining. For histologically evaluating kidney injury, each slide was stained with HE. A renal pathologist (JIY) scored the percentage of necrotic tubules out of total tubules in cortical areas in a blind manner. Kidney injury was scored according to the percentage of damaged tubules among total tubules: 0, normal; 1, < 25% damage; 2, 25-50% damage; 3, 50-75% damage, 4, 75-90% damage; and 5, > 90% damage. Histological criteria for injured tubules included the loss of tubular brush border, vacuolization, chromatin condensation, and denuded tubular basement membrane. Renal tubules in five randomly selected high-power fields, typically including 50-100 tubules, were evaluated and scored.
[0104] For IHC, tissue sections were deparaffinized, rehydrated in ethyl alcohol, and washed in tap water. Antigens were retrieved in 10 mM sodium citrate buffer for 20 min, using a Black & Decker vegetable steamer. Slides were blocked with 10% donkey serum for 30 min at 22 °C, and washed using PBS. Primary antibodies for NF-KB (Clone 8242S, Cell signaling technology, Danvers, MA) and ICAM-1 (Clone AF796, R&D Systems) were diluted to appropriate concentrations using 2% casein in bovine serum albumin, added to the slides, and incubated overnight at 4 °C. After washing, secondary antibodies (Dako, Carpinteria, CA) were applied for 1 h at 22 °C. Diaminobenzidine was added for 2 min, and hematoxylin was used to counterstain slides. A semi-quantitative score for staining intensity was obtained by examining at least five fields in each section under a 400 x magnification and via digital image analysis (MetaMorph version 4.6r5, Universal Imaging Corp., Downingtown, PA).
[0105] Measurement of DNA-binding activity of NF-KB: Twenty micrograms of kidney nuclear extracts were used to measure the binding activity of p65/c-Rel (NF-KB) to DNA, using the TransAM NF-KB assay kit (ActiveMotif, Carlsbad, CA), according to the manufacturer’s protocol.
[0106] Quantitative real-time polymerase chain reaction: The established method for whole kidney RNA extraction was used. Briefly, whole kidney samples were homogenized with 700 mΐ of RNAiso reagent (Takara Bio Inc., Otsu, Shiga, Japan). Next, 160 mΐ of chloroform was added into homogenized samples of kidney cells. Next, the mixture was shaken vigorously for 30 s, kept at 22 °C for 3 min, and centrifuged at 16,000 x g for 15 min at 4 °C. The aqueous phase, located on the top of three phases, was transferred to a fresh tube carefully, to prevent contamination with other phases. Extracted RNA was precipitated by adding 400 mΐ of isopropanol and centrifuged at 16,000 x g for 30 min at 4 °C. RNA pellets were washed with 70% ethanol, air-dried for 2 min, and dissolved in sterile diethyl pyrocarbonate-treated distilled water. Quantity and quality of extracted RNA were assessed by spectrophotometric measurements at wavelengths of 260 and 280 nm. Isolated RNA was reverse transcribed to cDNA using cDNA synthesis kits (Takara Bio Inc, Shiga Prefecture, Japan). Five microliters of each cDNA were mixed with 10 mΐ of the SYBR Green PCR Master Mix (Applied Biosystems, Foster City, CA) and 5 pmol of sense/antisense primers. Primer concentrations were determined after preliminary experiments confirming optimal concentrations for each primer. Expression levels of //-7a, II- 1b, 11-2, 11-4, 11-6, II- 10, Il-17a, Ccl2, Ccl3, Ccl5, Cxcl2, Ifii-g, Tnf-a, and Icam-1 were measured using the ABI PRISM 7700 Sequence Detection System (Applied Biosystems). PCR conditions were as follows: the initial heating step was performed for 9 min at 95 °C, followed by 35 cycles of denaturation for 30 min at 94.5 °C, annealing for 30 s at 60 °C, and extension for 1 min at 72 °C, and final extension for 7 min at 72 °C. Sequence data for each primer are shown in Table 1.
[0107] TABLE 1. PRIMER SEQUENCES
Figure imgf000037_0001
Figure imgf000038_0001
[0109] Each sample was run in triplicate in separate tubes and a negative control without cDNA was also run in parallel with each assay. After real-time PCR, the temperature was increased from 60 °C to 95 °C at a rate of 2 °C/min, to construct a melting curve. The expression value for each gene was normalized to 18S rRNA and relative fold expression values were calculated using a comparative CT method with 2
Figure imgf000039_0001
[0110] Multiplex cytokine assay in serum: Thirteen inflammatory cytokines and chemokines, including IL-la, IL-Ib, IL-2, IL-4, IL-6, IL-10, IL-17A, CCL2, CCL3, CCL5, CXCL2, IFN-g, and TNF-a were analyzed using Milliplex Mouse Cytokine/Chemokine Magnetic bead Panel - Multiplex Assay kits, according to the manufacturer’s protocol (EMD Millipore, Billerica, MA), using 1 :2 diluted serum. Quantification was performed using a Fuminex 200 microplate reader (EMD Millipore).
[0111] Terminal deoxynucleotidyl transferase dUTP nick-end labeling assay: Cell death was evaluated by the FITC-labelled TUNEL assay, using a commercially available kit (S7110, Merck Millipore, Billerica, MA). TUNEL-positive cells in formalin-fixed renal tissue were identified by examining at least five fields in each section under a 400 x magnification and with digital image analysis (MetaMorph version 4.6r5, Universal Imaging Corp.).
[0112] Isolation of kidney mononuclear cells and splenocytes from mice: KMNCs were isolated according to an established protocol. Both kidneys were removed after exsanguination, decapsulated, finely minced, and incubated in collagenase type 1 (1 mg/ml) (Worthington Biochemical, Lakewood, NJ) for 30 min at 37 °C. After enzymatic digestion, a single-cell kidney suspension was achieved by mechanical disruption of the tissue through a 70 pm strainer (BD bioscience, San Jose, CA). KMNCs were collected following centrifugation at RT using isotonic Percoll density gradient (GE Healthcare, Chicago, IL) (1,500 x g for 30 min in brake off mode), as per the manufacturer’s instructions. Collected cells were washed and re-suspended in RPMI media containing 5% FBS and automatically counted using the Countess® II FL Automated Cell Counter (Life Technologies, Waltham, MA).
[0113] Splenocytes were collected through mechanical disruption of the spleen using a 70 pm strainer (BD bioscience) and incubated with RBC lysis solution (Qiagen, Hilden, Germany) for 3 min, to remove red blood cells.
[0114] FACS sorting of kidney CD4+ T cells: For FACS sorting, KMNCs were pre-incubated with anti-CD 16/CD32 Fc block (Clone 93, Biolegend, San Diego, CA) for 10 min on ice and stained with monoclonal Ab anti-CD45 (APC, Clone 30-Fl 1, Biolegend), anti-Ly6G (APC- Fire750, Clone 1A8, Biolegend), anti-Ly6C (FITC, Clone HK1.4, Biolegend), anti-F4/80 (PerCP-Cy5.5, Clone BM8, Biolegend), and anti-CD3 (PE, Clone 17A2, Biolegend) for 30 min at 4 °C. After washing and resuspension in FACS buffer (PBS with 5% FBS), stained cells were analyzed with LSRII using FACS Diva software (BD Biosciences) and FlowJo software (Version 10.2). After excluding debris/dead cells and doublets, each sample was analyzed based on CD45, Ly6G, Ly6C, F4/80, and CD3 expression. OneComp eBeads (ThermoFisher) were used for compensation.
[0115] Immunofluorescence: After performing deparaffinization, rehydration, and antigen retrieval, slides were blocked with 10% goat serum for 30 min at 22 °C, and washed using phosphate buffered saline with Tween-20. Primary antibodies for Ly-6G (Clone ab25377,
Abeam, Cambridge, UK), Ly-6C (Clone abl5627, Abeam), F4/80 (Clone ab6640, Abeam) and fluorescein (FITC)-conjugated Lotus tetragonolobus lectin (LTL) (Clone FL-1321, Vector laboratories, Burlingame, CA) were diluted to proper concentrations using 2% casein in bovine serum albumin, added to slides, and incubated overnight at 4 °C. After washing, goat anti-rat IgG Alexa Fluor 647 (abl50167, Abeam) was applied for 1 h at 22 °C. Next, 4’,6-diamidino-2- phenylindole (DAPI) (MBD0015, Sigma- Aldrich, St. Louis, MO) was used to counterstain slides. Staining intensity was semi-quantitatively scored by examining at least five fields in each section under a 400/ magnification via digital image analysis (MetaMorph version 4.6r5, Universal Imaging Corp.).
[0116] Statistics: Data were expressed as means ± standard deviation (SD) values. Statistical differences were analyzed using one-way ANOVA with Bonferroni post hoc test using Prism 8 (GraphPad, San Diego, CA). If P < 0.05, results were statistically significant.
[0117] LPS endotoxemia model: To generate the LPS endotoxemia mouse model, LPS derived from Escherichia coli (Sigma-Aldrich, Milwaukee, WI, USA) was injected into male mice.
[0118] High-grade CLP sepsis model and treatment regiment: Male C57BL/6 mice purchased from ORIENTBIO (Seongnam-si, Gyeonggi-do, Republic of Korea) were subjected to high- grade CLP at 9-10 weeks of age, using a previously described procedure with minor modification. All mice were housed, and experiments were performed, in a specific-pathogen- free area of Yonsei Biomedical Research Institute, Yonsei University College of Medicine, which was certified by the Association for Assessment and Accreditation of Laboratory Animal Care International. Mice were anesthetized through i.p. injection of a mixture of 80 mg/kg ketamine and 10 mg/kg xylazine prior to all procedures. After anesthesia, the peritoneum was opened in a sterile manner and the cecum was ligated using 4-0 black silk thread 1 cm from the distal end and punctured with a 23 -gauge needle. After puncturing, the needle was removed and a small amount of stool was extruded through both punctures to ensure patency. Then, the cecum was placed back into the abdominal cavity and the abdominal incision was closed with 6-0 nylon sutures, with stainless steel removable wound clips used on the skin. After this procedure, 1 ml of prewarmed saline per 20 g body weight was administered subcutaneously. Sham-treated mice underwent the same procedure except for the ligation and puncture of the cecum. After this procedure, 1.0 x 109 particles of Exo-naive or Exo-srhcB were administered to mice via i.p. injection at 0, 6, 12 and 18 h. As a control, an equivalent volume of phosphate buffer solution (PBS) was injected in the same manner. The animals were assessed every 2 h during the initial 48 h after CLP, and then every 4 h for 5 days. Samples were collected to evaluate the outcome of the procedure within 18 h after CLP.
[0119] Intravital imaging: A laser-scanning intravital confocal microscope (IVM-C; IVIM Technology, Daejeon, South Korea) was used to visualize the biodistribution and anti-septic effects of Exo-srlKB. During intravital imaging, mouse body temperature was maintained at 37°C using a homoeothermic controller. Mice were anaesthetized with intramuscular injections of Zoletil (30 mg/kg) and xylazine (10 mg/kg). To access the internal organs, including the liver, spleen and kidney, a small incision of 10 mm was made in both the skin and peritoneum.
Exposed organs were kept hydrated through repeated application of saline during imaging. The biodistribution of exosomes was visualized through wide-area z-stack imaging before and after the injection of Exo-Naive or Exo-srhcB. To fluorescently label neutrophils of C57BL/6N mice in vivo, anti-Ly6G antibody (BD Bioscience, San Jose, CA, USA) conjugated with Alexa Fluor 555 (A20009; Thermo Fisher Scientific, Waltham, MA, USA) was injected intravenously naive or srlxB exosomes labeled with mCLING ATTO 647N (Synaptic System, Gottingen, Germany) were intravenously injected via a tail vein catheter or 31-G insulin syringe. To generate the sepsis model, high-dose LPS (Sigma- Aldrich, St. Louis, MO, USA) was intravenously injected 1-16 h prior to imaging. [0120] Confocal microscopy: For exosome uptake analysis, exosomes were diluted in 0.5 ml of Dulbecco’s PBS (DPBS). Subsequently, the suspension was incubated with 647N-labeled mCLING-ATTO (Synaptic System) according to the manufacturer's instructions. Then, a pellet of the mixture was obtained through centrifugation and 300 mΐ of the resuspended pellet was incubated with the HUYECs. After 24 h, cells were washed with DPBS and fixed in 4% paraformaldehyde solution. Hoechst was used for nuclear staining. Images were recorded using a Zeiss 710 confocal microscope (Zeiss, Oberkochen, Germany).
[0121] Luciferase assay: HEK293 cells were stably transfected with a luciferase reporter construct regulated under the NF-KB response element (SL-0012; Signosis, Santa Clara, CA, USA) and grown according to the manufacturer’s instructions. Cells were treated with exosomes for 24 h at 37°C, and luciferase activity was measured (El 501; Promega, Madison, WI, USA) using a SpectraMax ID3 microplate reader (Molecular Devices, Sunnyvale, CA, USA).
[0122] Flow cytometry: The levels of surface markers expressed on HUYECs were assessed using flow cytometry. The cells were separated and harvested through centrifugation, labeled with phycoerythrin (PE)-conjugated antibodies specific for human ICAM-1 (BD Biosciences) on ice for 30 min in the dark, and then washed extensively. All samples were analyzed with a BD Celesta flow cytometer (BD Biosciences). The data were analyzed using BD FACSDiva software. PE-conjugated antibodies specific to IgG l k (BD Biosciences) were used as the isotype control.
Example 1. Characterization and analysis of engineered exosomes [0123] The production, collection, and purification of exosomes have been thoroughly described in previous studies as shown in Figure 13. The size of particles mostly ranged from 30 to 120 nm, with a mean size of 101 nm. Transmission electron microscopy (TEM) revealed intact cup-shaped membrane vesicles with sizes in accordance with nanoparticle tracking analysis (NT A) results (Figure 1, B and C). Immunoblotting analysis results for Exo-srlxB revealed the robust expression of target proteins, including srlxB, mCherry, CD9, and GFP with positive exosome markers, such as CD63, TSG101, Alix, and GAPDH. Exo-srlKB lacked the expression of cell organelle markers, including prohibitin, calnexin, GM130, and nucleoporin p62. Exo-Naive did not express any markers except exosome markers (Figure ID). Example 2. Injection of Exo-srliiB ameliorates renal ischemic-reperfusion injury [0124] Firstly, the role of EXO-SHKB in the course of renal IRI was investigated. Each experimental group was intraperitoneally injected with 3 x 109 pn of Exo-srlicB or with Exo- Naive three times in 1 -h intervals (total of 9 x 109 pn), either before or after renal IRI. (Pre treatment: 3, 2, and 1 h before IRI surgery; Post-treatment: 1, 2, and 3 h after IRI surgery) Mice were sacrificed either 24 or 48 h after surgery (Figure 2A).
[0125] Interestingly, the mice group that received Exo-srlxB showed a significantly lower level of serum blood urea nitrogen (BUN) and creatinine than the Exo-Naive-injected group after IRI surgery, both in pre-treatment and post-treatment models (24-h/48-h BUN and creatinine in both models, P < 0.001) (Figure 2B and 2C). Serum neutrophil gelatinase-associated lipocalin (NGAL) levels were significantly reduced in the Exo-srlKB-injected group, in the pre- and post treatment models as compared with those in the Exo-Naive group ( P < 0.001) (Figure 2D). Histologic evaluation results also revealed lower tubular injury scores in the mouse group receiving EXO-SHKB treatment as compared with those in the Exo-naive group, in pre-/post- treatment models (P < 0.05 vs. Exo-Naive-injected group) (Figure 2E). Collectively, these data demonstrate that the systemic delivery of Exo-srl KB directly prevents the course of ischemic AKI.
Example 3. Systemic Exo-srliiB treatment represses renal NF-KB signaling in IR- injured kidney
To understand the underlying mechanism by which systemic Exo-srlxB treatment affects the course of IR- induced AKI, the systemic delivery of Exo-srlxB was first studied to determined if it represses local NF-KB signaling in kidneys. The expression of NF-KB p65 protein in renal nuclear extracts from each different experimental group was measured via western blotting. Systemic treatment with 9 x 109 pn of Exo-srlxB significantly decreased IR-induced NF-KB nuclear translocation, as compared with that seen in the Exo-Naive-treated group, in the pre-IRI (24-h after IRI, P < 0.05; 48-h, P < 0.001) and post-IRI (24-/48-h after IRI, P < 0.01) (Figure 3A) treatment model. The finding was reconfirmed by measuring the DNA binding activity of p65 using renal nuclear extracts from each experimental group. Exo-srlKB treatment significantly downregulated the DNA binding activity of NF-KB after kidney IRI as compared with that in the Exo-Naive group, regardless of treatment timings (Pre-treatment: 24-/48-h after IRI, P < 0.05; Post-treatment: 24-/48-h after IRI, P < 0.01) (Figure 3B). Further validation through NF-KB immunohistochemical (IHC) staining showed a significant reduction in NK-KB expression in the Exo-srlKB-treated group as compared with that of the control treatment group (Pre-treatment: 24- h after IRI, P < 0.05; 48-h, P < 0.001 and post-treatment: 24-/48-h after IRI, P < 0.05) (Figure 3C). Thus, systemic Exo-srlicB treatment can reduce renal NF-KB signaling in post-IR kidneys.
Example 4. EXO-SITKB ameliorates inflammation in post-ischemic kidneys [0126] To further investigate whether systemic exosomal srlxB treatment locally alleviates renal IRI-induced inflammation, gene expression levels of pivotal inflammatory mediators was determined, including //-7a, II- 1b, 11-2, 11-4, 11-6, II- 10, II- 17a, Ccl2, Ccl3, Ccl5, Cxcl2, Ifii-g, and Tnf-a through quantitative real-time polymerase chain reaction (qRT-PCR). Treatment with 9 x 109 pn of Exo-srlxB before IRI significantly suppressed the expression levels of II- 1b, 11-6, Tnf- a, Ccl2, Ccl5, and Cxcl2 as compared with those of the Exo-Naive-treated group in IR- injured kidneys (Figure 4A). Multiplex cytokine analysis was also performed using serum from different experimental groups. Results were less significant than kidney transcriptional data, but there was an obvious tendency for decreased expression of inflammatory cytokines, including IL-6, TNF-a, CCL2, CCL5, and CXCL2 in the Exo-srlxB treatment group, as compared with that in the control group (Figure 4B).
[0127] Additionally, the effect of exosomal srlxB treatment on the expression of adhesion molecules was evaluated, by comparing transcriptomic/translational intercellular adhesion molecule 1 (ICAM-1) levels among treatment groups. qRT-PCR results revealed the significantly lower level of Icam-1 expression in the Exo-srhcB treatment group than that in the Exo-Naive treatment, in both pre-/post-treatment models (Figure 4C). This result was reproduced in the translational level via western blotting and IHC staining (Figure 4, D and E). Thus, systemic Exo-srlxB delivery can downregulate the expression of pro-inflammatory cytokines/chemokines and adhesion molecules in IR- induced AKJ.
Example 5. EXO-SITKB alleviates apoptosis in post-ischemic kidneys [0128] Based on the well-known role of NF-KB in modulating programmed cell death, how Exo-srlKB affected apoptosis in post-ischemic kidneys was investigated. Kidney IRI surgery induced significant apoptosis in renal cells, which showed an abrupt increase in cleaved caspase- 3 and cleaved Poly (ADP-ribose) polymerase (P RP) levels in western blot analysis. Systemic delivery of Exo-srlxB either before/after the injury could substantially downregulate the expression level of cleaved caspase-3 and cleaved PARP, suggesting that Exo-srl KB has protective effects on apoptosis in post-ischemic kidneys (Figure 5A). Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining was used to determine the extent of renal cell injury. Compared with the controls, Exo-srlxB -treated kidneys showed a significantly lower number of TUNEL-positive cells in pre-/post-treatment models (Figure 5B).
Example 6. Biodistribution of exosomes after renal ischemia- reperfusion injury [0129] The biodistribution of Exo-srlxB in renal IRI model was investigated to better understand which cell types are orchestrating the protective effect of Exo-srlxB in post- ischemic kidneys. Mice were intravenously injected with fluorochrome-labeled Ly6G and F4/80 antibodies 1 h before IRI surgery and intravenously injected with 9 x 109 pn of Exo-srlxB 1 hr after reperfusion. Intravital imaging was performed at 10 min, 5 h, and 24 h after reperfusion (Figure 6A). Intravital imaging following injection of DiD-labeled exosomes as well as anti- Ly6G antibody to visualize neutrophils (Fluor 555) and anti-F4/80 antibody to visualize macrophages (Fluor 488) confirmed that both Exo-srlxB and Exo-Naive are taken up by neutrophils (Ly6G+) and macrophages (F4/80+) in post-ischemic kidneys (Figure 6B). We observed mild increase in DiD (green) signal in renal tubules after IRI, raising a possibility of exosome uptake by renal tubular cells but the signal intensity was not significantly different compared to the one before the DiD-labeled exosome injection (data not shown). Spleens were observed at the same time which showed exosomal uptake in the neutrophils and macrophages in the outer parenchymal area (Figure 6C). Inner parenchymal area did not show any significant uptake (data not shown).
Example 7. Systemic delivery of Exo-srlnB affects the renal immune cell population after kidney IRI
[0130] Immune cells are known to serve important roles in the course of ischemic AKJ; hence, it was evaluated whether providing exosomal srlxB treatment before IRI surgery affected the population of each immune cell type. After 24 h of ischemic AKJ, there was no difference in the total number of renal cells isolated through multiple steps of mechanical disruption, enzymatic digestion, and Percoll density gradient method between the experimental groups, but the Exo-srl kB-treated group had significantly lower frequency of kidney mononuclear cells (KMNCs) than those of the Exo-Naive-treated group ( P < 0.05) (Figure 7, A and B). Further analysis showed that the Exo-srlKB-injected group had significantly lower frequency of neutrophil (0)45P^60+) ( P < 0.01), pro-/anti-inflammatory mononuclear phagocytic cells (0)45¾t6u7 CD45+F4/80+) (P < 0.01 IP < 0.05, respectively), and T cells (CD45+CD3+) (P < 0.05) among total KMNCs than those in the Exo-Naive-injected group (Figure 7C). Results of immunofluorescence of IR-damaged kidneys also showed decreased neutrophil and mononuclear phagocytic cell frequencies in Exo-srlKB-injected kidneys (Figure 7D). However, Exo-srlxB treatment did not have significant effects on the total immune cell numbers or frequencies in the spleen after ischemic AKI. There were no differences in total cell numbers and frequencies of total immune cells (CD45+), neutrophils (CD45+Fy6G+), and pro-/anti-inflammatory mononuclear phagocytic cells (CD45+Fy6C+/ CD45+F4/80+), except T cells (CD45+CD3+) between the Exo-srlxB and Exo-Naive treatment groups. This confirms that systemic Exo-srlxB treatment before ischemic AKI has significant local effects on renal immune cell proliferation/trafficking, but not on splenic immune cells.
Example 8. Intravenous delivery of Exo-srlKB
[0131] To validate whether the similar protective effects from intraperitoneal exosomal delivery is found with intravenous delivery, some experiments were repeated by using intravenous injection method. The improved bio-chemical outcomes as well as the decreased level of NF-KB signaling and ICAM-1 expression with intravenous delivery of Exo-srlxB were reproduced as shown in Figure 8. Thus, intravenous delivery of Exo-srlxB results in similar biologic effects compared to intraperitoneal delivery in ischemic AKI model.
Example 9. More characterization and analysis of srli B-loaded exosomes [0132] To produce sufficient exosomes, culture medium was collected from each cell type. [0133] HUVECs were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA) and cultured in F-12K medium (ATCC) containing 10% fetal bovine serum (FBS; Atlas Biologicals, Fort Worth, CO, USA), heparin (Sigma-Aldrich), endothelial cell growth supplement (BD Biosciences) and 1% penicillin/streptomycin (Thermo Fisher). HUVECs between the third and sixth passages were used in all experiments. Human monocytes (THP-1) were purchased from ATCC and cultured in RPMI 1640 medium (Welgene, Daegu, Korea) with 10% FBS, 1% penicillin/streptomycin and 0.05 mM 2-mercaptoethanol (Sigma-Aldrich).
[0134] Exosomes were isolated using a combination of tangential flow filtration (TFF) and size-exclusion chromatography (SEC) (Figure 13 A). To reduce the risk of loading the SEC column with impurities that would exceed its binding capacity, the samples were subjected to diafiltration and concentration through TFF. After TFF, the concentrated medium was loaded onto an SEC column for further purification. A second TFF was then performed to concentrate the exosomes. Each type of exosome isolated through sequential purification was analyzed via nanoparticle tracking analysis (NTA), which characterizes the size and concentration of exosomes (Figure 13B). Particles with diameters from 30 to 120 nm accounted for over 80% of all particles, with a mean size as 101 nm, which is consistent with the characteristic size range (30-120 nm) of exosomes. Additionally, transmission electron microscopy (TEM) showed intact cup-shaped membrane vesicles with sizes corresponding to the NTA results obtained from Exo-Naive and EXO-SHKB (Figure 9B). To further characterize the exosomes in our preparations, two common exosome markers were investigated. These were the tetraspanins CD63 and TSG101, through western blotting.
[0135] Western blotting was performed as described in a previous study. Antibodies targeting the following proteins were used: IkBa (CST4812; Cell Signaling Technology, Danvers, MA, USA), p65 (CST6956S; Cell Signaling Technology), CD 9 (NBP2-22187; NOVUSBIO, Centennial, CO, USA), CD63 (EXOAB-CD63A-1 ; SBI, Tokyo, Japan), TSG101 (ab228013; Abeam, Cambridge, UK), GM130 (ab52649; Abeam), GAPDH (sc-47724; Santa Cruz Biotechnology, Santa Cruz, CA, USA), Histone H3 (abl791; Abeam), mCherry (abl25096; Abeam), and GFP (CST2555; Cell Signaling Technology).
[0136] The presence of CD63 and TSG101 was clearly observed in the samples, whereas a Golgi-derived contaminant, GM130, was only detected in the cell lysate (Figure 9C). This analysis of the exosome preparations showed that they exhibit the characteristics of exosomes. Furthermore, robust loading of srlKB-mCherry-CRY2 (130 kDa) into exosomes isolated from SHKB stable cell lines was observed. Example 10. Exo-srlKB improves survival and ameliorates acute organ injury in septic mice
[0137] It was investigated to determine whether intraperitoneal (i.p.) injection with Exo-srhcB protected against endotoxic shock. Animals received a single i.p injection with a lethal dose of LPS (40 mg/kg body weight for C57BL/6, 20 mg/kg body weight for BALB/c), followed by a single i.p. injection of Exo-srhcB (6h apart). Compared with control C57BL/6 mice, which showed 100% mortality due to LPS-induced sepsis, mice treated with Exo-srlxB were remarkably resistant to LPS-induced mortality; the majority of the mice were rescued from sepsis and showed prolonged survival (Figure 10A, upper). Exo-srlKB -mediated effects were associated with modest protection from the LPS-induced temperature drop (Figure 14A). A mouse mouse model of LPS-induced sepsis was developed using BALB/c mice. Exo-srhcB treatment significantly improved the survival rate of LPS-induced sepsis in BALB/c mice (Figure 10A, middle). The role of Exo-srhcB in survival was examined next, using a model of CLP-induced sepsis, as a standardized murine model of intra-abdominal sepsis. The animals survived the 7-day monitoring period, indicating that Exo-srhcB provides lasting protection against sepsis mortality (Figure 10A, lower). To further investigate whether Exo-srhcB treatment alleviated the inflammation induced by sepsis, enzyme-linked immunosorbent assay (ELISA) was used to determine the concentrations of key inflammation factors in the serum. Significant reductions significant reductions were observed in the expression levels of the pro- inflammatory cytokines TNFa, IL-Ib, and IL-6, and the chemokine carbon tetrachloride (CCL4), in the Exo-srhcB treatment group with LPS-induced sepsis (Figure 10B, upper). Serum levels of TNF-a and CCL4 in the CLP group were also observed to be significantly higher than those in sham-operation control mice, but were not elevated in mice treated with both CLP and Exo- srhcB (Figure 10B, lower). These results suggest that Exo-srhcB alleviates inflammation induced by LPS or CLP and protects septic mice against the acute inflammatory process.
[0138] Numerous studies have shown that acute kidney injury (AKI) is a frequent and serious complication of sepsis, occurring in 50% or more cases, and is associated with very high mortality. To determine the role of Exo-srhcB in CLP-induced AKI in the septic mouse model, renal histological examination was performed. In comparison with those that underwent a sham operation, the renal tubular cells of mice treated with phosphate-buffered saline (PBS) and Exo- Naive mice showed significant damage in terms of vacuolization, loss of the brush border, and nuclear condensation of tubular epithelial cells (Figure IOC). Notably, Exo-srlKB treatment markedly decreased damage to tubular areas, preserving proximal tubules and demonstrating the therapeutic potential of Exo-srhcB for sepsis-induced renal injury. These results are consistent with those for LPS-induced AKI (Figure 14B). The severity of histological kidney damage was further quantified, and found that Exo-srhcB treatment significantly reduced the injury score by 50% in both sepsis models (Figure 10D). Exo-srlxB ameliorated degenerative changes in the renal tubules, although some damaged areas were present. These data suggest the critical importance of Exo-srhcB in improving renal physiological structure and function in septic mice.
Example 11. Biodistribution of exosomes after LPS injection
[0139] The changes in exosome biodistribution in the LPS-injected sepsis septic mouse model were investigated. The in vivo biodistribution was analyzed using a custom-made intravital video-rate laser scanning confocal microscopy system. mCLING fluorescence dye-labeled exosomes were injected intravenously into LysM8^ transgenic C57BL/6 mice that had also been injected with anti-Ly6G antibody to visualize neutrophils (green fluorescent protein [GFP] and Alexa 555) and macrophages (GFP). It was observed that most exosomes were taken up by neutrophils and macrophages rapidly, i.e., within a few minutes of intravenous exosome injection (Figure 11 A).
[0140] Exosomes were delivered mainly to the neutrophils of the liver (Figure 1 IB), and increased recruitment of neutrophils and macrophages was observed, as well as increased uptake of exosomes, in the spleens of LPS-induced mice (Figure 11C). mCLING- labeled exosomes were also delivered to neutrophils in the kidneys of LPS-injected mice (Figure 15A). These observations suggest that therapeutic exosomes were successfully delivered to target neutrophils and macrophages within 30 minutes in the septic mouse model.
Example 12. EXO-SITKB alleviates inflammation associated with sepsis [0141] The target specificity and efficiency of Exo-srhcB in vitro was identified. Exo-srlKB, but not Exo-Naive, significantly blocked TNF-a induced NF-KB activation in an HEK293 cell stably expressing an NF-KB luciferase reporter gene (Figure 12A). Increasing doses of Exo- srlxB decreased NF-KB reporter activity in a dose-dependent manner, indicating that srhcB released from exosomes prevents NF-KB transcriptional activity (Figure 12B). [0142] Among all sepsis-responsive cells, monocytes/macrophages play the most critical role in promoting the immune response and depletion of these cells in septic mice increases mortality. THP-l cells are widely employed to represent monocytes in cell culture models. When THP-1 cells were stimulated with LPS, treatment with Exo-srlKB led to decreased secretion of the inflammatory cytokines TNF-a and monocyte chemoattractant protein (MCP)-l, which are under NF-KB transcriptional control, compared with Exo-Naive (Figure 12C). The effects of Exo-srlKB was also compared with those of the commonly used NF-KB inhibitor JSH-23, which interferes with the binding of NF-KB to its target DNA. The example data shows that Exo-srl B and JSH- 23 had comparable effects with respect to inhibiting EPS-induced NF-KB activation and cytokine production in THP-1 cells (Figure 12C, lane 4 versus lane 6).
[0143] Sepsis is characterized by monocyte adherence to the endothelium. Finder normal conditions, endothelial cells are quiescent and do not interact with monocytes. However, in an inflammatory environment, activated endothelial cells express adhesion molecules, such as intercellular cell adhesion molecule (ICAM)-1, which bind to monocytes. The expression of adhesion molecules is widely considered to be activated during the development of sepsis. [0144] To explore the effect of Exo-srlxB on the expression of cell adhesion molecules in human umbilical vein endothelial cells (HUVECs), the examples first examined whether Exo- srlxBs are internalized in the cells. mCFING-labeled ECO-SHKBS were detectable in HFTVECs, with maximum internalization observed after 24 h (Figure 12D). It was found that ICAM-1 expression in HUVECs was significantly inhibited by EXO-SHKB treatment (Figure 12E). IF-8 and MCP-1 also triggered the migration and adhesion of monocytes to the vascular endothelium and led to extravasation of these cells into the surrounding tissues. Expression of IF-8 and MCP- 1 has been observed previously in various tissues during acute inflammation caused by bacterial and viral infections, and is associated with severe sepsis. It was found that Exo-srlxB suppressed EPS-induced IF-8 and MCP-1 production (Figure 16A) by inhibiting NF- KB transcriptional activity in HUVECs (Figure 16B). These results confirm that EXO-SHKB reduces the NF-KB-mediated inflammatory response during sepsis.
Example 13. Protective effect of Exo- srli Bs on lung injury in LPS-induced sepsis [0145] Animals received a single intraperitoneal injection with a lethal dose of EPS (30 mg/kg body weight for 30 min, followed by a single Intravenous injection of Exo-srlkB (lxlO10) for 10 hrs. Lung tissues were analyzed by H&E staining as shown in Figure 17. The group injected with LPS for 10 hrs showed the neutrophil infiltration and alveolar macrophage proliferation, intra-alveolar and external hyperemia, and bleeding proceeded. Alveolar endothelial cell damage as well as alveolar necrosis are observed. Exosome administration alleviated alveolar wall expansion, neutrophil infiltration and alveolar macrophage proliferation even if congestion is slightly observed.
[0146] 30mg/kg LPS was administered to LPS -administered group (negative control) and 3- dose Exo-srlkB-administered group, consisting of 10 mice per group. After 30 minutes of the administration, Exo-srlkB was administered i.v once, and the survival rate for 72 hours was evaluated as shown in Figure 18. Compared with control C57BL/6 mice, mice treated with high dose of Exo-srhcB were resistant to LPS-induced mortality and showed prolonged survival.
[0147] 30mg/kg LPS was administered to LPS -administered group (negative control) and 3- dose Exo-srlkB-administered group, consisting of 5 mice per group. After 30 minutes of the administration, Exo-srlkB was administered i.v once, and blood plasma was obtained by autopsy at each time point. The obtained blood plasma level of TNF-a was measured by ELISA method (Figure 19). The expression levels of the proinflammatory cytokines TNF- a was reduced in the Exo-srhcB treatment group with LPS-induced sepsis in a time-dependent manner.
[0148] Animals received a single intraperitoneal injection with a lethal dose of LPS (30 mg/kg body weight for 30 min, followed by a single Intravenous injection of Exo-srlkB (lxl 010). Liver tissues were analyzed by H&E staining (Figure 20). After 1 hour of LPS administration, infiltration of Kupffer and immune cells around the central vein was observed slightly, but infiltration of inflammatory cells was alleviated in the Exosome-administered group. After 10 hours of LPS administration, infiltration of Kupffer and immune cells around the central vein was significantly increased, and damage to the connective tissue between liver cells was observed. In the exosome-administered group, infiltration was reduced, but necrosis and swelling process of hepatocytes were expected to gradually relieve. Therefore, repeated administration of Exosome is expected to enhance the protective effect.
EMBODIMENTS FOR TREATING AKI [0149] Embodiment 1. A method for treating acute kidney injury (AKJ) in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-kB inhibitor to the subject.
[0150] Embodiment 2. The method of any of the above embodiments, wherein the NF-KB inhibitor is selected from the group consisting of a NF-KB inhibitory protein, its fragment, and a mixture thereof.
[0151] Embodiment 3. The method of any of the above embodiments, wherein the NF-KB inhibitor is selected from the group consisting of super-repressor (SR)-IKB (srlicB), IkB-a, IkB-b, IkB-e, BCF-3, a mutant thereof, and a mixture thereof
[0152] Embodiment 4. The method of any of the above embodiments, wherein the NF-KB inhibitor is super-repressor (SR)-IKB (srlKB).
[0153] Embodiment 5. The method of any of the above embodiments, wherein the composition of is administered via oral, transdermal, intraperitoneal, intravenous, intramuscular, subcutaneous, or mixed routes.
[0154] Embodiment 6. The method of any of the above embodiments, wherein the subject is human.
[0155] Embodiment 7. The method of any of the above embodiments, wherein the method further comprises administering an anti-inflammatory agent to the subject.
[0156] Embodiment 8. The method of any of the above embodiments, wherein the exosome comprising a NF-KB inhibitor further comprises a photo-specific binding protein.
[0157] Embodiment 9. The method of any of the above embodiments, wherein the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:l or SEQ ID NO:2.
[0158] Embodiment 10. The method of any of the above embodiments, wherein the NF-KB inhibitor is represented by SEQ ID NO: 1.
[0159] Embodiment 11. The method of any of the above embodiments, wherein the NF-KB inhibitor is represented by SEQ ID NO: 2.
[0160] Embodiment 12. The method of any of the above embodiments, wherein the photo specific binding protein is a first photo-specific binding protein and/or a second photo-specific binding protein; the first photo-specific binding protein is conjugated to an exosome specific marker to form fusion protein I; and the second photo-specific binding protein is conjugated to the NF-KB inhibitor to form fusion protein II.
[0161] Embodiment 13. The method of embodiment 12, wherein the fusion protein I and the fusion protein II are linked reversibly through the first photo-specific binding protein and the second photo-specific binding protein.
[0162] Embodiment 14. The method of embodiment 12 or 13, wherein the first photo-specific binding protein is conjugated to an exosome specific marker to be located in the direction toward inside of the exosome.
[0163] Embodiment 15. The method of any of embodiments 12-14, wherein the first photo specific binding protein and the second photo-specific binding protein are selected from the group consisting of CIB, CIBN, PhyB, PIF, FKFl, GIGANTEA, CRY and PHR.
[0164] Embodiment 16. The method of embodiment 15, wherein the first photo-specific binding protein is CIB or CIBN and the second photo-specific binding protein is CRY or PHR. [0165] Embodiment 17. The method of embodiment 15, wherein the first photo-specific binding protein is CRY or PHR and the second photo-specific binding protein is CIB or CIBN. [0166] Embodiment 18. The method of embodiment 15, wherein the first photo-specific binding protein is PhyB and the second photo- specific binding protein is PIF.
[0167] Embodiment 19. The method of embodiment 15, wherein the first photo-specific binding protein is PIF and the second photo-specific binding protein is PhyB.
[0168] Embodiment 20. The method of embodiment 15, wherein the first photo-specific binding protein is GIGANTEA and the second photo-specific binding protein is FKFl.
[0169] Embodiment 21. The method of embodiment 15, wherein the first photo-specific binding protein is FKFl and the second photo-specific binding protein is GIGANTEA.
[0170] Embodiment 22. The method of embodiments 12-21, wherein the exosome specific marker is selected from the group consisting of CD9, CD63, CD81 and CD82.
[0171] Embodiment 23. The method of any of the above embodiments, wherein the exosome has a diameter of between about 50 nm and about 200 nm.
[0172] Embodiment 24. The method of any of the above embodiments, wherein the exosome has a diameter of between about 50 nm and about 150 nm. [0173] Embodiment 25. The method of any of the above embodiments, wherein the composition is a pharmaceutical composition further comprising a physiologically acceptable carrier.
[0174] Embodiment 26. A composition comprising an exosome comprising an NF-KB inhibitor as described in Embodiments 1-16 for treating AKX.
[0175] Embodiment 27. Else of a composition comprising an exosome comprising an NF-KB inhibitor as described in Embodiments 1-16 for treating AKI.
EMBODIMENTS FOR TREATING OTHER DISEASES
[0176] Embodiment 1. A method for treating a disease caused by sepsis in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF-KB inhibitor to the subject.
[0177] Embodiment 2. The method of any of the above embodiments, wherein the disease is selected from the group consisting of pneumonia, cytokine storm syndrome, respiratory distress syndrome, and organ failure. In some embodiments, the disease is pneumonia. In some embodiments, the disease is cytokine storm syndrome. In some embodiments, the disease is respiratory distress syndrome. In some embodiments, the disease is organ failure.
[0178] Embodiment 3. The method of any of the above embodiments, wherein the NF-KB inhibitor is selected from the group consisting of a NF-KB inhibitory protein, its fragment, and a mixture thereof.
[0179] Embodiment 4. The method of any of the above embodiments, wherein the NF-KB inhibitor is selected from the group consisting of super-repressor (SR)-IKB (srlxB), IkB-a, IkB-b, IkB-e, BCL-3, a mutant thereof, and a mixture thereof.
[0180] Embodiment 5. The method of any of the above embodiments, wherein the NF-KB inhibitor is super-repressor (SR)-IKB (srbcB).
[0181] Embodiment 6. The method of any of the above embodiments, wherein the composition of is administered via oral, transdermal, intraperitoneal, intravenous, intramuscular, subcutaneous, or mixed routes.
[0182] Embodiment 7. The method of any of the above embodiments, wherein the subject is human. [0183] Embodiment 8. The method of any of the above embodiments, wherein the method further comprises administering an anti-inflammatory agent to the subject.
[0184] Embodiment 9. The method of any of the above embodiments, wherein the exosome comprising a NF-KB inhibitor further comprises a photo-specific binding protein.
[0185] Embodiment 10. The method of any of the above embodiments, wherein the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:l or SEQ ID NO:2.
[0186] Embodiment 11. The method of embodiment 10, wherein the NF-KB inhibitor is represented by SEQ ID NO: 1.
[0187] Embodiment 12. The method of embodiment 10, wherein the NF-KB inhibitor is represented by SEQ ID NO: 2.
[0188] Embodiment 13. The method of any of the above embodiments, wherein the photo specific binding protein is a first photo-specific binding protein and/or a second photo-specific binding protein; the first photo-specific binding protein is conjugated to an exosome specific marker to form fusion protein I; and the second photo-specific binding protein is conjugated to the NF-KB inhibitor to form fusion protein II.
[0189] Embodiment 14. The method of embodiment 13, wherein the fusion protein I and the fusion protein II are linked reversibly through the first photo-specific binding protein and the second photo-specific binding protein.
[0190] Embodiment 15. The method of embodiment 13 or 14, wherein the first photo-specific binding protein is conjugated to an exosome specific marker to be located in the direction toward inside of the exosome.
[0191] Embodiment 16. The method of any one of embodiments 13-15, wherein the first photo- specific binding protein and the second photo- specific binding protein are selected from the group consisting of CIB, CIBN, PhyB, PIF, FKF1, GIGANTEA, CRY and PHR.
[0192] Embodiment 17. The method of embodiment 16, wherein the first photo-specific binding protein is CIB or CIBN and the second photo-specific binding protein is CRY or PHR. [0193] Embodiment 18. The method of any of the above embodiments, wherein the first photo- specific binding protein is CRY or PHR and the second photo- specific binding protein is CIB or CIBN. [0194] Embodiment 19. The method of embodiment 16, wherein the first photo-specific binding protein is PhyB and the second photo- specific binding protein is PIF.
[0195] Embodiment 20. The method of embodiment 16, wherein the first photo-specific binding protein is PIF and the second photo-specific binding protein is PhyB.
[0196] Embodiment 21. The method of embodiment 16, wherein the first photo-specific binding protein is GIGANTEA and the second photo-specific binding protein is FKF1.
[0197] Embodiment 22. The method of embodiment 16, wherein the first photo-specific binding protein is FKF1 and the second photo-specific binding protein is GIGANTEA.
[0198] Embodiment 23. The method of any one of embodiments 13-22, wherein the exosome specific marker is selected from the group consisting of CD9, CD63, CD81 and CD82.
[0199] Embodiment 24. The method of any of the above embodiments, wherein the exosome has a diameter of between about 50 nm and about 200 nm.
[0200] Embodiment 25. The method of any of the above embodiments, wherein the exosome has a diameter of between about 50 nm and about 150 nm.
[0201] Embodiment 26. The method of any of the above embodiments, wherein the composition is a pharmaceutical composition further comprising a physiologically acceptable carrier.
[0202] Embodiment 27. A composition comprising an exosome comprising an NF-KB inhibitor as described in Embodiments 1-16 for treating a disease caused by sepsis..
[0203] Embodiment 28. Use of a composition comprising an exosome comprising an NF-KB inhibitor as described in Embodiments 1-16 for treating a disease caused by sepsis.
[0204] While the present disclosure has been described and illustrated herein by references to various specific materials, procedures and examples, it is understood that the disclosure is not restricted to the particular combinations of materials and procedures selected for that purpose. Numerous variations of such details can be implied as will be appreciated by those skilled in the art. It is intended that the specification and examples be considered as exemplary only, with the true scope and spirit of the disclosure being indicated by the following claims. All references, patents, and patent applications referred to in this application are herein incorporated by reference in their entirety.

Claims

1. A method for treating acute kidney injury (AKJ) in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF- KB inhibitor to the subject.
2. The method of claim 1, wherein the NF-KB inhibitor is selected from the group consisting of a NF-KB inhibitory protein, its fragment, and a mixture thereof.
3. The method of any one of the preceding claims, wherein the NF-KB inhibitor is selected from the group consisting of super-repressor (SR)-IKB (srl B), IkB-a, IkB-b, IkB-e, BCL-3, a mutant thereof, and a mixture thereof.
4. The method of any one of the preceding claims, wherein the composition of is administered via oral, transdermal, intraperitoneal, intravenous, intramuscular, subcutaneous, or mixed routes.
5. The method of any one of the preceding claims, further comprising administering an anti inflammatory agent to the subject.
6. The method of any one of the preceding claims, wherein the exosome comprising a NF- KB inhibitor further comprises a photo- specific binding protein.
7. The method of any one of the preceding claims, the NF-KB inhibitor comprises an amino acid sequence having at least 85% sequence identity to SEQ ID NO:l or SEQ ID NO:2.
8. The method of any one of the preceding claims, wherein the exosome comprising a NF-KB inhibitor further comprises a first photo-specific binding protein and/or a second photo-specific binding protein, the first photo-specific binding protein is conjugated to an exosome specific marker to form fusion protein I; and the second photo-specific binding protein is conjugated to the NF-KB inhibitor to form fusion protein II.
9. A method for treating a disease caused by sepsis in a subject in need thereof, comprising: administering an effective amount of a composition comprising an exosome comprising an NF- KB inhibitor to the subject.
10. The method of claim 9, wherein the disease is selected from the group consisting of pneumonia, cytokine storm syndrome, respiratory distress syndrome, and organ failure.
PCT/IB2021/052708 2020-03-31 2021-03-31 Use of exosome-based delivery of nf-kb inhibitors WO2021198961A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
KR1020217014385A KR20210124180A (en) 2020-03-31 2021-03-31 Use of exosome-based delivery of NF-κB inhibitors
AU2021247253A AU2021247253A1 (en) 2020-03-31 2021-03-31 Use of exosome-based delivery of NF-κB inhibitors
JP2022559527A JP2023519700A (en) 2020-03-31 2021-03-31 Use of exosome-based delivery of NF-κB inhibitors
CA3173845A CA3173845A1 (en) 2020-03-31 2021-03-31 Use of exosome-based delivery of nf-kb inhibitors
CN202180024099.7A CN115335079A (en) 2020-03-31 2021-03-31 Use of exosome-based delivery of NF-KB inhibitors
EP21781109.0A EP4127136A4 (en) 2020-03-31 2021-03-31 Use of exosome-based delivery of nf-kb inhibitors
KR1020227034736A KR20220149781A (en) 2020-03-31 2021-03-31 USE OF EXOSOME-BASED DELIVERY OF NF-κB INHIBITORS
US17/227,359 US20210299215A1 (en) 2020-03-31 2021-04-11 Use of Exosome-Based Delivery of NF-KB Inhibitors

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
KR20200039011 2020-03-31
KR10-2020-0039011 2020-03-31
US202063112154P 2020-11-10 2020-11-10
US63/112,154 2020-11-10
US202063112155P 2020-11-11 2020-11-11
US63/112,155 2020-11-11

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/227,359 Continuation-In-Part US20210299215A1 (en) 2020-03-31 2021-04-11 Use of Exosome-Based Delivery of NF-KB Inhibitors

Publications (1)

Publication Number Publication Date
WO2021198961A1 true WO2021198961A1 (en) 2021-10-07

Family

ID=77928093

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2021/052708 WO2021198961A1 (en) 2020-03-31 2021-03-31 Use of exosome-based delivery of nf-kb inhibitors

Country Status (7)

Country Link
EP (1) EP4127136A4 (en)
JP (1) JP2023519700A (en)
KR (2) KR20220149781A (en)
CN (1) CN115335079A (en)
AU (1) AU2021247253A1 (en)
CA (1) CA3173845A1 (en)
WO (1) WO2021198961A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115212234A (en) * 2021-10-22 2022-10-21 武汉大学中南医院 Application of fibroreticulocyte-derived exosome in preparation of medicine for treating and/or preventing sepsis acute kidney injury

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014168548A2 (en) * 2013-04-12 2014-10-16 El Andaloussi, Samir Therapeutic delivery vesicles
KR20180036134A (en) * 2016-09-30 2018-04-09 한국과학기술원 Process for preparing exosome loading super-repressor-IκB protein, and pharmaceutical composition for use in preventing or treating inflammatory diseases containing the same as an active ingredient
WO2018102397A1 (en) * 2016-11-29 2018-06-07 PureTech Health LLC Exosomes for delivery of therapeutic agents

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3002520A1 (en) * 2016-09-30 2018-04-05 Cellex Life Sciences, Incorporated Compositions containing protein loaded exosome and methods for preparing and delivering the same
CN107158390A (en) * 2017-07-12 2017-09-15 上海市东方医院 Purposes of the histon deacetylase (HDAC) HDAC6 inhibitor in preventing and treating acute injury of kidney medicine is prepared

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014168548A2 (en) * 2013-04-12 2014-10-16 El Andaloussi, Samir Therapeutic delivery vesicles
KR20180036134A (en) * 2016-09-30 2018-04-09 한국과학기술원 Process for preparing exosome loading super-repressor-IκB protein, and pharmaceutical composition for use in preventing or treating inflammatory diseases containing the same as an active ingredient
WO2018102397A1 (en) * 2016-11-29 2018-06-07 PureTech Health LLC Exosomes for delivery of therapeutic agents

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHOI HOJUN, KIM YOUNGEUN, MIRZAAGHASI AMIN, HEO JAENYOUNG, KIM YU NA, SHIN JU HYE, KIM SEONGHUN, KIM NAM HEE, CHO EUNAE SANDRA, YO: "Exosome-based delivery of super-repressor IB relieves sepsis-associated organ damage and mortality", SCIENCE ADVANCES, vol. 6, no. 15, 8 April 2020 (2020-04-08), XP055854716, DOI: 10.1126/sciadv.aaz6980 *
LIU SHU FANG, MALIK ASRAR B.: "NF-κB activation as a pathological mechanism of septic shock and inflammation", AMERICAN JOURNAL OF PHYSIOLOGY - LUNG CELLULAR AND MOLECULAR PHYSIOLOGY, AMERICAN PHYSIOLOGICAL SOCIETY, US, vol. 290, no. 4, 1 April 2006 (2006-04-01), US, pages 622 - 645, XP055854713, ISSN: 1040-0605, DOI: 10.1152/ajplung.00477.2005 *
See also references of EP4127136A4 *
YIM NAMBIN, RYU SEUNG-WOOK, CHOI KYUNGSUN, LEE KWANG RYEOL, LEE SEUNGHEE, CHOI HOJUN, KIM JEONGJIN, SHAKER MOHAMMED R., SUN WOONG,: "Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein–protein interaction module", NATURE COMMUNICATIONS, vol. 7, no. 1, 1 November 2016 (2016-11-01), XP055783136, DOI: 10.1038/ncomms12277 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115212234A (en) * 2021-10-22 2022-10-21 武汉大学中南医院 Application of fibroreticulocyte-derived exosome in preparation of medicine for treating and/or preventing sepsis acute kidney injury
CN115212234B (en) * 2021-10-22 2023-11-10 武汉大学中南医院 Application of exosomes derived from reticulocytes in preparation of drugs for treating and/or preventing sepsis acute kidney injury

Also Published As

Publication number Publication date
EP4127136A1 (en) 2023-02-08
JP2023519700A (en) 2023-05-12
EP4127136A4 (en) 2024-04-03
CA3173845A1 (en) 2021-10-07
AU2021247253A1 (en) 2022-10-20
CN115335079A (en) 2022-11-11
KR20220149781A (en) 2022-11-08
KR20210124180A (en) 2021-10-14

Similar Documents

Publication Publication Date Title
JP7275193B2 (en) Cardiomyocyte-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
CN113694075A (en) Use of exosomes for treating diseases
Poppelaars et al. Critical role for complement receptor C5aR2 in the pathogenesis of renal ischemia‐reperfusion injury
US11779600B2 (en) Methods of treating or preventing alzheimer&#39;s disease and associated conditions
Winkler et al. Blood-brain barrier resealing in neuromyelitis optica occurs independently of astrocyte regeneration
AU2018386215A1 (en) Methods and compositions for treating cancer using exosomes-associated gene editing
WO2019213686A1 (en) Therapeutic compositions and uses therefor
WO2021198961A1 (en) Use of exosome-based delivery of nf-kb inhibitors
JP2021517811A (en) Immunoexosomes and how to use them
US20210299215A1 (en) Use of Exosome-Based Delivery of NF-KB Inhibitors
Sun et al. FcRn-targeting and ROS-responsive Fedratinib-incorporated nanoparticles alleviate asthma by inducing eosinophil apoptosis
ES2843724T3 (en) NMDAR antagonists for the treatment of tumor angiogenesis
Shin et al. Inflammation‐Targeting Mesenchymal Stem Cells Combined with Photothermal Treatment Attenuate Severe Joint Inflammation
US20220387512A1 (en) Methods for treating pain
US20220280428A1 (en) Tunable leukocyte-based biomimetic nanoparticles and methods of use
WO2023183855A1 (en) Delivery methods using platelets
MASUM Studies on morphofunctional interactions between epithelial cells composing blood-urine barrier in mice: as a role for immunological gate regulating renal pathology
WO2019146805A1 (en) Therapeutic agent for frontotemporal lobar degeneration, method for screening therapeutic agent for frontotemporal lobar degeneration and method for treating frontotemporal lobar degeneration
KR20230159847A (en) Compositions and methods for targeting inflammatory or activated cells and treating or ameliorating inflammatory conditions and pain
JP2022534786A (en) Oligodendrocyte-derived extracellular vesicles for multiple sclerosis therapy
WO2023089151A1 (en) Materials and methods for treatment of macular degeneration
CN112789030A (en) Pharmaceutical formulations of peptide inhibitors
Chun et al. SAP330 THE TUBULAR EXPRESSION OF MESENCHYMAL MARKERS INCLUDES EXTRACELLULAR MATRIX PROTEINS
EA043103B1 (en) APPLICATION OF EXOSOMES FOR THE TREATMENT OF DISEASE

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: 21781109

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3173845

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2022559527

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2021247253

Country of ref document: AU

Date of ref document: 20210331

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021781109

Country of ref document: EP

Effective date: 20221031