WO2013070079A2 - miRNA IN BLOOD BRAIN BARRIER OF MS PATIENTS - Google Patents

miRNA IN BLOOD BRAIN BARRIER OF MS PATIENTS Download PDF

Info

Publication number
WO2013070079A2
WO2013070079A2 PCT/NL2012/050793 NL2012050793W WO2013070079A2 WO 2013070079 A2 WO2013070079 A2 WO 2013070079A2 NL 2012050793 W NL2012050793 W NL 2012050793W WO 2013070079 A2 WO2013070079 A2 WO 2013070079A2
Authority
WO
WIPO (PCT)
Prior art keywords
mir
nucleic acid
acid sequence
seq
brain barrier
Prior art date
Application number
PCT/NL2012/050793
Other languages
French (fr)
Other versions
WO2013070079A8 (en
WO2013070079A3 (en
Inventor
Arie Reijerkerk
Helga Eveline DE VRIES
Original Assignee
Vereniging Voor Christelijk Hoger Onderwijs, Wetenschappelijk,
Stichting Vu-Vumc
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 Vereniging Voor Christelijk Hoger Onderwijs, Wetenschappelijk,, Stichting Vu-Vumc filed Critical Vereniging Voor Christelijk Hoger Onderwijs, Wetenschappelijk,
Publication of WO2013070079A2 publication Critical patent/WO2013070079A2/en
Publication of WO2013070079A3 publication Critical patent/WO2013070079A3/en
Publication of WO2013070079A8 publication Critical patent/WO2013070079A8/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6809Methods for determination or identification of nucleic acids involving differential detection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • the blood-brain barrier separates the blood and the central nervous system (CNS).
  • the central nervous system consists of the brain and the spinal cord.
  • This separation between the blood and CNS is created by specialized endothelial cells, brain endothelial cells, which are distinct from peripheral endothelial cells, as the blood-brain barrier is composed of high density cells restricting passage of substances from the bloodstream much more than endothelial cells elsewhere in the body (Abbott et al., Nat Rev Neurosc, 2006, 7, 41-53).
  • the BBB restricts the diffusion of large or hydrophilic molecules into the CNS, e.g. antibodies, but also of microscopic entities, e.g. bacteria.
  • small hydrophobic molecules e.g.
  • the BBB also actively transports molecules, such as for instance glucose, across the BBB utilizing transporter proteins.
  • Cells of the immune system can also pass the BBB (Pachter et al., J Neuropathol Exp Neurol. 2003 Jun;62(6):593-604; Abbott et al., Nat Rev Neurosc, 2006, 7, 41-53; ; Begley and Brightman, Prog Drug Res. 2003;61 :39-78.; Loscher and Potschka, NeuroRx. 2005 Jan;2(l):86-98.; Carvey et al., Neurochem. 2009 Oct;l 11(2):291-314.; Abbott et al., Neurobiol Dis. 2010 Jan;37(l): 13-25).
  • the function of the BBB is to allow precise control over the substances that leave or enter the central nervous system, and this function is essential for brain homeostasis and reliable functioning of the neuronal environment.
  • MS is an inflammatory demyelinating disease of the central nervous system (CNS).
  • CNS central nervous system
  • Dysregulation of the BBB and transendothelial migration of activated leukocytes are among the earliest cerebrovascular abnormalities seen in multiple sclerosis (MS) brains and parallel the release of inflammatory cytokines/chemokines.
  • MS multiple sclerosis
  • the BBB can obstruct or hamper the entry of pharmaceuticals or drugs.
  • many high potential drugs for the central nervous system (CNS) for instance biopharmaceuticals such as antibodies, are currently not or less suitable for use in the treatment of the CNS because these drugs can often not cross the blood-brain barrier, making the BBB a major hurdle for successful CNS drug development that require drugs to pass the BBB.
  • a compound that increases the blood brain barrier function may allow the (partial) reconstitution of the blood brain barrier function in MS patients that suffer from impaired BBB function.
  • An increase of the BBB function might for instance prevent bacterial infiltration of the brain or reduce seizures, or help to restore homeostasis.
  • a less complicated assay to study the BBB function is an in vitro assay in which (primary) brain endothelial cells are cultured in vitro on a membrane and passage of tracer compounds through the membrane is determined.
  • the electrical resistance (expressed as ohm) over the cultured brain endothelial cells can also be measured, i.e. the transendothelial electric resistance (Deli et al., Cell Mol Neurobiol. 2005 Feb;25(l):59-127).
  • a problem in the development of a medical treatment for the central nervous system, in particular in patients suffering form or diagnosed with multiple sclerosis, e.g. the brain, is that there is a need for compounds that can safely and effectively modulate, i.e. increase or decrease, the blood brain barrier function.
  • assays that are robust and simple to perform to allow for large scale screening for compounds (or compound libraries) that can modulate the blood brain barrier function, and would, for example be suitable in the treatment of blood brain barrier abnormalities in multiple sclerosis.
  • nucleic acid includes a plurality of molecules (e.g. lO's, 100's, 1000's, lO's of thousands, 100's of thousands, millions, or more molecules).
  • a nucleic acid or a nucleic acid sequence according to the present invention may include any polymer or oligomer of pyrimidine and purine bases, preferably cytosine, thymine, and uracil, and adenine and guanine, respectively (See Albert L. Lehninger, Principles of Biochemistry, at 793-800 (Worth Pub. 1982)).
  • the present invention contemplates any deoxyribonucleotide, ribonucleotide or peptide nucleic acid component, and any chemical variants thereof, such as methylated, hydroxymethylated or glycosylated forms of these bases, and the like.
  • the polymers or oligomers may be heterogenous or homogenous in composition, and may be isolated from naturally occurring sources or may be artificially or synthetically produced.
  • the nucleic acids may be DNA or RNA, or a mixture thereof, and may exist permanently or transitionally in single-stranded or double-stranded form, including homoduplex, heteroduplex, and hybrid states.
  • a nucleic acid or nucleic acid sequence of the invention comprises other kinds of nucleic acid structures such as for instance a DNA/RNA helix, peptide nucleic acid (PNA), morpholino, locked nucleic acid (LNA) and/or a ribozyme.
  • nucleic acid or “nucleic acid sequence” may also encompass a chain comprising non-natural nucleotides, modified nucleotides and/or non- nucleotide building blocks which can exhibit the same function as natural nucleotides.
  • non-natural nucleotides, modified nucleotides and/or non-nucleotide building blocks are referred to with the term “nucleotide analogues”.
  • % sequence identity is defined herein as the percentage of nucleotides or nucleotide analogues in a nucleic acid sequence according to the invention that is identical with the corresponding nucleotides in a reference sequence after aligning the two sequences and introducing gaps, if necessary, to achieve the maximum percent identity.
  • % sequence identity is defined herein as the percentage of nucleotides or nucleotide analogues in a nucleic acid sequence according to the invention that is identical with the corresponding nucleotides in a reference sequence after aligning the two sequences and introducing gaps, if necessary, to achieve the maximum percent identity.
  • additional nucleotides in the nucleic acid according to the invention, that do not align with the reference sequence are not taken into account for determining sequence identity.
  • complementarity is defined herein as nucleotides of a nucleic acid sequence that can bind to another nucleic acid sequence through hydrogen bonds, i.e. nucleotides that are capable of base pairing. Nucleotides that can form base pairs, that are complementary to one another, are e.g. cytosine and guanine, thymine and adenine, adenine and uracil, guanine and uracil. The percentage complementarity does not have to be calculated over the entire length of a nucleic acid sequence. The percentage of complementarity may be limited to a specific region of which the nucleic acid sequences which are base-pared, e.g.
  • a nucleic acid may comprise a nucleic acid sequence of which at least 8 adjacent nucleotides are at least 75% complementary to 8 adjacent nucleotides of the complementary nucleic acid that may comprise 8 nucleotides, 16 nucleotides, or 32 nucleotides, or more. As long as of 8 adjacent nucleotides of the nucleic acid sequence at least 6 nucleotides can base-pair with 8 adjacent nucleotides of the complementary nucleic acid sequence the nucleic acid sequence is at least 75% complementary.
  • the term comprises according to the invention is to be interpreted such that for example a nucleic acid comprising a nucleic acid sequence of 8 adjacent nucleotides may also include a nucleic acid consisting of no more than said 8 adjacent nucleotides.
  • Gene expression is defined as RNA transcription of a gene, e.g. expression of mRNA, and/or translation of mRNA into protein. Gene expression thus comprises RNA expression and/or protein expression.
  • miRNA genes may express RNA transcripts that are subsequently processed by the miRNA machinery.
  • gene expression thus comprises the transcript that is initially transcribed, the subsequent intermediate processed transcripts, and finally the miRNA molecule which may be incorporated in the RISC complex.
  • gene expression of a gene encoding a protein comprises the initially transcribed RNA molecule, but may also comprise subsequently spliced or otherwise by the cellular machinery processed transcribed RNA molecules, and/or the protein which may be translated from messenger RNA (mRNA).
  • mRNA messenger RNA
  • the expression level is to be understood as the amount of RNA transcript that is transcribed by a gene and/or the amount of protein that may be translated from an RNA transcript, e.g. mRNA.
  • the expression level may be determined through quantifying the amount of RNA transcript which is expressed, e.g.
  • the expression level may also be determined through measuring the effect of a miRNA on a target mRNA.
  • a 3 'UTR sequence comprising a target miRNA target sequence may be incorporated in a reporter gene, e.g. luciferase, this way, the expression level of a miRNA gene may be indirectly measured by measuring the amount of Luciferase expression which may be controlled by a particular miRNA.
  • the amount of Luciferase expression also correlates with the expression level of the particular gene.
  • the expression level of the miRNA when determining the expression level of a miRNA as described above (e.g. directly through qPCR or indirectly with a reporter gene construct), in addition, the expression level of the miRNA may be correlated to the expression level of a target gene of the particular miRNA.
  • the amount of protein that may be translated from an RNA transcript may also be measured to determine the expression level of a gene. Methods are known in the art, such as ELISA and Western blot.
  • the expression level of a protein may also be indirectly measured.
  • a reporter gene construct may be used which comprises the RNA transcription factor binding site. The expression level may than be subsequently determined by measuring the reporter gene construct expression, e.g. in case GFP is used, green fluorescence intensity may be determined as a measure of the expression level of the RNA transcription factor.
  • the present invention now surprisingly solves at least one of the problems as described above. It was found for the first time that expression of specific miRNA in multiple sclerosis is impaired, reduced, relative to expression of such miRNA's in healthy subjects, that these miRNA's can increase blood brain barrier function and that increasing the level of such miRNA's can improve blood brain barrier function, in particular in multiple sclerosis.
  • nucleic acids including those listed in SEQ ID No. 1-16, play an important role in modulating the BBB function, in particular in patients diagnosed with or suffering from Multiple Sclerosis.
  • the examples show that expression of miRNA with a sequence according to SEQ ID NO 1 - 16 is impaired in material obtained from patients suffering from or diagnosed with multiple sclerosis.
  • these nucleic acids increase blood brain barrier function. Compounds comprising such nucleic acid are thus useful in increasing blood brain barrier function, in particular in multiple sclerosis.
  • the nucleic acids according to the invention and including those selected from the group consisting of SEQ ID No. 1-16, are capable of increasing the blood brain barrier function, which is in particular useful in patients diagnosed with or suffering from multiple sclerosis.
  • blocking nucleic acids selected from the group consisting of SEQ ID No. 1-16 may result in a decrease in the blood brain barrier function, for example in healthy humans or other patients, but not necessarily suffering from multiple sclerosis (i.e. in those subjects in which expression of the disclosed miRNA is not or only to a lesser extent impaired).
  • blocking of a nucleic acid is achieved by providing a (partially) complementary nucleic acid sequence that can (partially) hybridise to the nucleic acid, such that when the (partially) complementary nucleic acid sequence is hybridised with the nucleic acid (and thus bound thereto), the nucleic acid can not perform its function, e.g. hybridizing with a target molecule, i.e. it is blocked.
  • the relative expression level of the nucleic acids listed in SEQ ID No. 1-16 can be used as a measure/indicator of blood brain barrier function, in particular in multiple sclerosis.
  • the expression level of a particular level of such nucleic acid is decreased, relative to a control setting, the BBB function is decreased.
  • Such an increase or decrease of the level of expression of a nucleic acid may be induced by the presence of a compound.
  • the invention further provides for an assay (method) for screening for compounds or compound libraries to identify compounds that are capable of modulating BBB function.
  • the assay comprises of determining the expression level(s) of at least one nucleic acid sequence as listed in SEQ ID No. 1-16 in the absence and/or presence of a compound to be tested.
  • a compound preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100%) sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis.
  • said compounds may be used in vitro or in vivo for increasing the blood brain barrier function.
  • in vivo is meant for instance in an animal model or a patient, with in vitro is meant a (primary) cell culture.
  • the use does not entail the treatment of the human or animal body by surgery or therapy.
  • the nucleic acids may be provided in the cell, for instance via transfection or other means of cell transfer, as explained below.
  • a nucleic acid according to the invention for example can comprise a nucleic acid sequence which has 95% sequence identity with SEQ ID No. 1.
  • Such a nucleic acid can be the exact length of SEQ ID No. 1, or be longer (e.g.
  • 1,2,3,4,5,6,7,8,9 nucleotides longer or shorter (e.g. 1,2,3,4,5,6,7,8,9 nucleotide shorter).
  • a skilled person will be able to test how many nucleotides he can remove or add to a nucleic acid according to SEQ ID NO 1-16, and still increase blood brain barrier function, in particular in multiple sclerosis.
  • the blood-brain barrier function can, for example, be measured by determining the transendothelial electric resistance (TEE ) of an endothelial brain cell monolayer, as is described in detail in Example 1.
  • TEE transendothelial electric resistance
  • TEER is expressed in ohm.
  • the ECISTM Model 1600R Applied BioPhysics, Troy, NY, USA
  • TEER the resistance of an endothelial brain cell monolayer to an electric current is measured, i.e. the ion permeability of the endothelial brain cell monolayer.
  • a relative high resistance means that less ions, and thus less current, can permeate the endothelial brain cell monolayer
  • a relative low resistance means that more ions, and thus more current, can pass through the endothelial brain cell monolayer.
  • Ion permeability of the endothelial brain cell monolayer correlates with the ability of compounds to passively diffuse across the blood brain barrier.
  • the TEER value measured relates to the BBB function.
  • increasing the blood-brain barrier function means that relative to a control setting, the TEER is increased when measured in the method described.
  • decreasing the blood-brain barrier function means that relative to a control setting, the TEER is decreased when measured in the method described.
  • a control setting is defined as a setting in which certain conditions (e.g. salt, nutrient and biological factor concentrations, H, temperature etc.) are selected. For instance, conditions may be selected that most closely resemble the conditions of a healthy patient, but for instance conditions may also be selected such as may occur in a diseased or injured patient in which BBB function is known to be disturbed. Whichever conditions selected, in the control setting the TEER is measured. Next, in a different setting comprising e.g. a composition for increasing or decreasing the blood-brain barrier function, the TEER is determined. In this different setting similar conditions are selected as in the control setting, but now a composition or compound is added that can, for instance, increase the BBB function.
  • certain conditions e.g. salt, nutrient and biological factor concentrations, H, temperature etc.
  • the TEER When comparing the TEER resistance value (ohm) for the latter condition, relative to the control setting, the TEER will have increased, i.e. having a higher value and thus a higher resistance, meaning that less ions, and thus less compounds, can pass through the endothelial cell layer, and the BBB function has increased.
  • the BBB function as defined is within the context of the assay.
  • An increase or decrease of the BBB function within the assay indicates that in alternative settings wherein the blood brain barrier is studied, e.g. in in vivo, or in vivo models, or in a therapeutic setting the blood brain barrier may have a similar effect and result in an increase or decrease of the blood brain barrier function in these settings, e.g. in a patient, e.g. a patient diagnosed with or suffering from Multiple Sclerosis.
  • RNA-induced silencing complex RISC
  • Protein expression is thought to be regulated through (partial) complementary base-pairing of the putative miRNA with target sequences in the 3' untranslated region of target mRNA, thereby causing degradation of the mRNA and/or translational repression, leading to improved BBB function. In patients suffering from multiple sclerosis the levels are reduced, leading to decreased blood brain barrier function.
  • putative miRNAs may also possibly act via transcriptional gene silencing or even activation of transcription and/or translation as this has also been occasionally described for (putative) miRNA sequences (Morris, Biotechniques. 2006 Apr;Suppl:7-13; Chua et al., Curr Opin Mol Ther. 2009 Apr;l 1(2): 189-99).
  • miRNA activity includes the modes of activity as described above.
  • a compound preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%, even more preferably 90%», even more preferably 95%», most preferably 100%» sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis.
  • the use does not entail the treatment of the human or animal body by surgery or therapy.
  • a compound preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%>, even more preferably 90%o, even more preferably 95%, most preferably 100%) sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for use in increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis.
  • a compound preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for the manufacture of a medicament for increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis.
  • the compound described above is or comprises miR-15a, miR-18b, miR-26a, miR-27a, miR-27b, miR-30a, miR-30b, miR-30c, miR-30d, miR-30e, miR-31, miR-106b, miR-125a-5p, miR-155, miR-195, or miR-487b.
  • the sequences of these miRNA's are given in SEQ ID NO 1 - 16.
  • These nucleic acids or use thereof according to the invention have been found to be particular advantageous for increasing the blood-brain barrier function, for example as determined according to the methods disclosed in the Examples, in particular in patients suffering from or diagnosed with multiple sclerosis.
  • a nucleic acid according to the invention has a length of at most 100, more preferably at most 75, even more preferably at most 50, most preferably at most 25 nucleotides.
  • a nucleic acid according to the invention can be transfected or transduced into a brain endothelial cell as described in example 3 and 4, and the effect on the TEER can be measured.
  • the term comprising within the context of the above is meant to also include a nucleic acid according to the invention consisting of the nucleic acid sequence having 70%», 75%», 80%», 85%», 90%», 95%, or 100%» sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID No. 1-16.
  • nucleic acid sequence which comprises a nucleic acid sequence having 70%, 75%, 80%, 85%, 90%, 95%, or 100% sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID No. 1-16, can consist of the same number of nucleotides of a nucleic acid sequence from SEQ ID No. 1-16, but may also comprise additional nucleotides, for instance 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 100 nucleotides, or any other integer, as long as the nucleic acid according to the invention is capable of increasing the BBB function.
  • the nucleic acid may also consist of a fragment of a sequence according to SEQ ID No. 1-16, such a fragment may be for instance as small as 10 nucleotides, although the fragment may be larger or smaller, as long as the fragment is capable of increasing the BBB function, as can be determined according to the methods provided by the invention.
  • the compound preferably a nucleic acid, as described above, comprising a nucleic acid sequence which has at least 70%, 80%, 85%, 90%, 95%, or 100% sequence identity with a nucleic acid sequence selected from the group of SEQ ID No 1-16 is a pre-miRNA, a pri-miRNA, a siRNA or a shRNA.
  • miRNAs are a novel class of endogenous small non-coding RNAs, which are believed to regulate gene expression via the so-called RNA interference pathway (RNAi).
  • RNAi RNA interference pathway
  • the primary transcripts of miRNAs can be transcribed by RNA polymerase II and III which generally have a length of several kilobases, and are called primary miRNAs (pri-miRNA).
  • pri-miRNA primary miRNAs
  • a typical animal pri-miRNA comprises an imperfectly paired stem of 30-35 bp, with a terminal loop and flanking segments (Bartel (2004) Cell 116, 281-297).
  • pri-miRNAs which are capped and polyadenylated (Kim (2005) Nature Rev. Mol. Cell. Biol. 6 376-385), are subsequently processed by the miRNA biogenesis pathway (reviewed in Winter et al., Nature Cell Biol 2009, vol 11, nr 3, p.228-234), in the cell nucleus to shorter, 70-100 nucleotide stem- loop structures known as pre -miRNAs.
  • This processing can be performed in animals by the RNase III endonuclease Drosha.
  • Pre-miRNAs can subsequently be transported into the cytoplasm by exportin-5, where they can be processed to double-stranded miRNAs, a miRNA duplex, with a length of 21-25 nucleotides by the RNase III endonuclease DICER.
  • the miRNA duplex comprises the miRNA nucleic acid sequence, which may be subsequently incorporated into the RNA-induced silencing complex (RISC).
  • RISC RNA-induced silencing complex
  • this activated RISC complex i.e. a RISC complex loaded with a miRNA, can inhibit gene expression of a target gene by inhibiting translation and/or by cleaving mRNA through complementary base-pairing, although other regulatory mechanisms can be envisaged as disclosed above.
  • miRNAs small interfering RNA
  • siRNAs small interfering RNA
  • the pre-miRNA or pri-miRNA according to the invention may comprise the natural sequences from which such a miRNA is processed, i.e. the endogenous sequence as it is expressed in nature.
  • non-natural nucleic acid sequences i.e. exogenous artificial pre-miRNA, pri- miRNA or a miRNA duplex RNA structures are also possible (Aagaard et al., Gene Ther. 2008 Dec; 15(23): 1536-49; Liu et al., Nucleic Acids Res. 2008 May;36(9):2811-24).
  • small interfering RNA (Elbashir et al., Nature. 2001 May 24;411(6836):494-8) or short hairpin RNA (shRNA) (Brummelkamp et al., 2002 Apr 19;296(5567):550-3).
  • siRNA small interfering RNA
  • shRNA short hairpin RNA
  • a short hairpin RNA can comprise two short complementary RNA sequences of 19-35 nucleotides long with a loop sequence in between such that a hairpin structure can be formed.
  • a small interfering RNA may comprise two complementary strands of 19-25 nucleotides.
  • a shRNA may also be processed by the RNA interference pathway, by the DICER enzyme, resulting in an siRNA.
  • RNA interference pathway by the DICER enzyme
  • siRNA As with a duplex miRNA, one of the strands of the siRNA duplex has been suggested to be incorporated into RISC, resulting in an activated RISC complex.
  • a nucleic acid comprising a nucleic acid sequence which has at least 70%, 80%, 85%, 90%», 95%», 100%» sequence identity with a nucleic acid sequence selected from the group of SEQ ID No 1-16, is preferably a pre-miRNA, pri-miRNA, siRNA, shRNA, or is hybridised to a complement nucleic acid sequence thus forming a siRNA duplex or miRNA duplex, such that the nucleic acid sequence with at least 70%, 80%, 85%, 90%, 95%, 100% sequence identity with a nucleic acid sequence selected from the group of SEQ ID No. 1-16 is compatible with the RNA interference pathway.
  • the provided nucleic acid may be recognized and processed by the RNA interference pathway or miRNA biogenesis pathway (Winter et al, Nature Cell Biol 2009, vol 11, nr 3, p.228-234) such that the nucleic acid sequence which has at least 70%, 80%, 85%, 90%, 95%, 100% sequence identity with a nucleic acid sequence selected from the group of SEQ ID No 1-16 can have its action for increasing the blood-brain barrier function.
  • a compound, preferably a nucleic acid according to the invention as disclosed above, is for use in a treatment for increasing blood-brain barrier function. It has been found that in particular these nucleic acids can advantageously be used to increase the blood-brain barrier function.
  • such a treatment may be a medical treatment of a patient suffering from a neurological condition, preferably multiple sclerosis.
  • a neurological condition preferably multiple sclerosis.
  • BBB damage is an early event, and thus increasing BBB function as an early intervention for treating such conditions may slow down, stop or even reverse such neurological conditions.
  • a compound, preferably a nucleic acid according to the invention as disclosed above is for use in treatment of multiple sclerosis, in particular to increase blood brain barrier function. It is beneficial to provide patients with a treatment that increases the blood-brain barrier such that homeostasis may be (partially) restored and neuronal damage reduced.
  • the use of a single compound, preferably nucleic acid according to the invention may suffice for increasing the blood-brain barrier function, however, it is also well within the scope of the invention to combine multiple compounds, preferably nucleic acids according to the invention such that an enhanced effect for increasing the blood-brain barrier is obtained.
  • the invention provides for the use of at least one nucleic acid according to the invention as described above, for use in a medical treatment, in particular for increasing the BBB function in a patient, in particular a patient diagnosed with or suffering from multiple sclerosis.
  • nucleic acids and the use thereof, that can bind, hybridize and/or base-pair to a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID No 1-16, thereby blocking the function thereof, for decreasing the blood brain barrier function according to the invention.
  • a nucleic acid binds, hybridizes and/or base-pairs (i.e. forms base pairs) with (part of) the a nucleic acid that comprises or consists of a nucleic acid sequence selected from the group consisting of SEQ ID No 1-16.
  • a compound preferably a nucleic acid
  • a compound preferably a nucleic acid, comprising a nucleic acid sequence of which at least 8 (or at least 10, 15, or 12) adjacent nucleotides are at least 70%, preferably 75%, more preferably 80%, even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% complementary to 8 (or at least 10, 15, or 20) adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1- 16, for decreasing blood -brain barrier function.
  • the use does not entail the treatment of the human or animal body by surgery or therapy.
  • nucleic acid has for example 8, 9, or 10 adjacent nucleotides that are complementary to 8, 9 , or 10 adjacent nucleotides of a nucleic acid sequence from the group of SEQ ID No 1-16.
  • These nucleic acids can in particular be used for decreasing the blood brain barrier function.
  • the additional nucleotides do not necessarily have to base pair, i.e. be complementary, with the nucleic acid sequence from SEQ ID No. 1-16.
  • Such a compound, comprising such complementary nucleic acid is capable of binding to a nucleic acid selected from the group of SEQ ID No. 1-16, to thereby blocking said nucleic acid.
  • the term blocking herewith is being defined as preventing the nucleic acid sequence of SEQ ID No. 1-16 from performing its function as described herein, e.g. having a miRNA activity or increasing the BBB function.
  • nucleic acid sequences from the group of SEQ ID No. 1-16 are involved in the BBB function, i.e. an increased concentration of said nucleic acid results in an increased BBB function, blocking the said nucleic acid sequences with a complementary nucleic acid sequence has been found to block its normal function, e.g. may be used for decreasing the BBB function.
  • a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID No 1-16 is involved in maintaining or increasing BBB function
  • blocking a nucleic acid comprising a nucleic acid sequence selected from the group of SEQ ID No 1-16 may result in a decrease of BBB function.
  • a decrease in BBB function means that relative to a control, the TEER is decreased, as described above.
  • a compound preferably a nucleic acid, comprising a nucleic acid sequence of which at least 8 (or at least 10, 15, or 20) adjacent nucleotides are at least 70%, preferably 75%, more preferably 80%, even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% complementary to 8 (or at least 10, 15, or 20) adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16, for use in decreasing blood-brain barrier function.
  • nucleic acid comprising a nucleic acid sequence of which at least 8 (or at least 10, 15, or 20) adjacent nucleotides are at least 70%, preferably 75%, more preferably 80%, even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% complementary to 8 (or at least 10, 15, or 20) adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16, for the manufacture of a medicament for decreasing blood-brain barrier function.
  • nucleic acid comprising a nucleic acid sequence of which at least 8 adjacent nucleotides are at least 85%, most preferably 100% complementary to 8 adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16 for decreasing the blood brain-brain barrier function further comprises nucleotide analogues.
  • Nucleotide analogous such as for instance Locked Nucleic Acids (Chan et al., Clin Exp Pharmacol Physiol. 2006 May-Jun;33(5-6):533-40)
  • nucleic acid sequences comprising the nucleotide analogue are capable of complementary base-pairing with its target sequence and/or reducing the blood brain barrier function, the skilled person may select such analogues, however, it is preferred to select analogues that result in an improvement of the stability and/or binding affinity.
  • Other nucleotide analogues are well known in the art, such as for instance morpholinos (Summerton and Weller Antisense Nucleic Acid Drug Dev.
  • nucleotide analogue(s) may result in an increased stability of the nucleic acid, and may improve the binding of the nucleic acid to its target, thereby improving its capacity to block its target, i.e. improving the blocking of a nucleic acid sequence selected from the group of SEQ ID No 1-16, for decreasing the blood brain-brain barrier function.
  • polyoma viruses e.g. JCV, HIV-1, or Japanese encephalitis virus may passage through the blood brain barrier.
  • the viral infiltration of the CNS may be more effectively treated with antiviral medication, as the antiviral medicine may more easily penetrate the brain to have its action.
  • such a treatment preferably further comprises the use of a medicament that would benefit from decreasing the blood-brain barrier function, thereby improving the medicaments ability to cross the blood-brain barrier and entering the CNS and thus the spinal cord and/or brain.
  • nucleic acid sequence may be sufficient for decreasing the blood-brain barrier function; however, it is also within the scope of the invention to combine multiple nucleic acids for decreasing BBB function according to the invention such that an improved decrease of the BBB function can be obtained.
  • nucleic acid that encodes a nucleic acid according to the invention, for instance DNA constructs that drive expression of an siRNA duplex or of a nucleic acid (partially) complementary to a nucleic acid sequence selected from SEQ ID No 1-16.
  • the nucleic acid may also be synthesised de novo, for example by the cell itself.
  • a nucleic acid according to the invention may be coupled to a ligand for delivery into the brain endothelial cells.
  • the ligand can mediate the uptake of the nucleic acid by the brain endothelial cells, upon which the nucleic acids can execute its function.
  • a cholesterol conjugate may be used as a ligand and can be used as follows: a nucleic acid is provided, chemically modified and labelled with a cholesterol moiety (Soutschek et al., J Nature.
  • nucleic acids can then for instance be injected intravenously, where the cholesterol group enables the siRNAs to be taken up by cells.
  • peptide or nucleotide aptamers may also be used as ligands, and the nucleic acid according to the invention may be coupled thereto (as reviewed e.g. in Cerchia et al. Methods Mol Biol. 2009;535:59-78 and Borghouts et al.,Comb Chem High Throughput Screen. 2008 Feb;l 1(2): 135-45).
  • Aptamers are molecules that are capable of binding to a specific target molecule, for instance a receptor on a cell, resulting in cellular uptake. Aptamers are usually created by selecting them from a large random sequence pool, but natural aptamers also exist in riboswitches. Aptamers are used for both basic research and clinical purposes as macromolecular drugs. These compound molecules have additional research, industrial and clinical applications. Nucleotide aptamers normally contain (usually short) strands of oligonucleotides. Peptide aptamers normally contain a short variable peptide domain, which are often attached at both ends to a protein scaffold.
  • nucleic acid may be coupled to a ligand which enables the delivery of the nucleic acid to brain endothelial cells, such that the nucleic acid sequences can have its modulating effect, i.e. increasing or decreasing the blood brain barrier function.
  • delivery of a nucleic acid according to the invention may be achieved through a delivery vehicle comprising the nucleic acid.
  • a delivery vehicle may be a gene therapy vector.
  • Suitable vectors for instance comprise adenoviral vectors, adeno-associated viral (AAV) vectors, gamma retroviral vectors and lentiviral vectors.
  • a delivery vehicle according to the present invention may also comprise a plasmid delivery system, a virus like particle (VLP), a stable nucleic acid lipid particle (SNALP), a cationic delivery system, a cationic liposomal delivery system, a cationic polymer, a lipoplex, or a liposome.
  • VLP virus like particle
  • SNALP stable nucleic acid lipid particle
  • cationic delivery system a cationic liposomal delivery system
  • a cationic polymer a lipoplex, or a liposome.
  • Virus like particles contain viral protein(s) derived from the structural proteins of a virus. In some cases these proteins are embedded within a lipid bilayer. These particles resemble the virus from which they were derived but lack viral nucleic acid, meaning that they are not infectious. Also VLPs are suitable for packaging plasmids or any other nucleic acid, such as for instance pre-miRNAs, pri-miRNAs, shRNAs, miRNA duplexes, siRNA duplexes. By modifications of the viral proteins and linkage of other molecules to the surface of the particle their tropism can be modulated. Cationic macromolecules can also be used for delivery vehicles, the two major classes being lipids and polymers. Examples of cationic vectors are liposomes (lipoplexes) and polyethelyenimine (PEI) (polyplexes).
  • PEI polyethelyenimine
  • the invention also includes the use of a nucleic acid according to the invention, the use of a nucleic acid coupled to a ligand according to the invention, or the use of a delivery vehicle according to the invention, for modulating the blood-brain barrier function.
  • nucleic acids according to the invention i.e. for increasing or decreasing the blood-brain barrier function
  • a pharmaceutical composition suitable for administration may comprise a pharmaceutically acceptable carrier, which is intended to include any and all solvents, dispersion media, coatings, isotonic and adsorption delaying agents and the like compatible with pharmaceutical administration.
  • the pharmaceutical composition may also comprise medicaments that are suitable for use in combination with a nucleic acid according to the invention in a treatment for increasing and/or decreasing the blood-brain barrier.
  • Routes of administration of pharmaceutical compositions according to the invention comprise oral administration, application on the skin, or injection, but are not limited thereto.
  • a method is provided for modulating the blood brain barrier by providing the nucleic acids as described above.
  • the blood brain barrier can be modulated in vitro or in vivo.
  • a method involving modulation of the blood brain barrier is in a human and in vivo, such a method does not involve a method for treatment of the human or animal body practised on the human or animal body.
  • the nucleic acid, or delivery vehicle is provided in vitro, the method may comprise providing a vessel or a dish which contains or comprises the nucleic acid, or delivery vehicle.
  • the nucleic acid, or delivery vehicle that is freeze dried in a well (e.g. Reinisalo et al., J Control Release. 2006 Jan 10;110(2):437-43 and del Pozo- Rodriguez et al., Eur J Pharm Biopharm. 2009 Feb;71(2): 181-9).
  • the nucleic acid or delivery vehicle is introduced in a cell, preferably a brain endothelial cell.
  • Said brain endothelial cell can be a primary cell or a cell line, preferably HCMEC/D3 (Weksler et al., the FASEB Journal. 2005;19: 1872-1874).
  • the current invention now provides for the above methods, and now makes it possible to modulate, e.g. in vitro, the blood-brain barrier by the provision of said nucleic acid, protein or delivery vehicle. These methods may in particular be useful in the study of the blood-brain barrier, in particular in multiple sclerosis.
  • the methods of the invention also provides for the modulation of the blood brain barrier in animal models that would be of particular interest for studying the effect of modulating BBB function.
  • animal models exist for multiple sclerosis. Studying the effect of the nucleic acids, and/or delivery vehicles according to the invention in these models may be of interest for the development of novel therapies, but is not limited thereto.
  • the nucleic acids and proteins according to the invention may be used.
  • the invention further also provides for an assay (method) for screening for compounds that may modulate the blood brain barrier function, preferably that increases blood brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis, comprising the steps of incubating a cell in the presence of a compound to be tested (i.e. screened), and determining the expression level of a nucleic acid sequence selected from the group of SEQ ID No. 1-16, wherein the expression level of said nucleic acid sequence is compared with the expression level of said nucleic acid sequence determined in a cell incubated without the presence of said compound to be tested (i.e. screened).
  • an assay for screening for compounds that may modulate the blood brain barrier function, preferably that increases blood brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis
  • Cells may for instance be contained in multi-well plates either in suspension or attached.
  • the cells may be incubated in the presence of a compound, for instance by adding the compound to the cell culturing medium.
  • Cell culturing medium is defined as a solution which provides the cell with nutrients such that the cells may be sustained.
  • the compound may also be provided in a suitable standard buffer, such as for instance phosphate buffered saline (PBS), for a period of time such that the cells are contacted with the compound. Any method incubating the cells with the compounds are allowed, as long as the compound may have its effect on the cells.
  • PBS phosphate buffered saline
  • modulation means either increasing or decreasing.
  • modulation means either increasing or decreasing.
  • the expression level of a said nucleic acid sequence is upregulated in a cell incubated in the presence of a compound as compared to a cell incubated in the absence of said compound, such a compound is capable of increasing the BBB function
  • the expression level of a said nucleic acid sequence is downregulated in the presence of a compound as compared to a cell incubated without the presence of said compound to be tested, such a compound is capable of decreasing the BBB function.
  • 1-16 of at least 2-fold may be considered as compounds that are identified to have the potential to modulate the BBB.
  • Not only single compounds may be tested, but it is also envisaged that combinations of compounds may be tested for their potential effect on modulation of the BBB function.
  • compounds able to increase the expression of at least one of the miRNA's with a sequence according to SEQ ID NO 1- 16 are useful in improving blood brain barrier function, in particular in patients diagnose with or suffering from multiple sclerosis.
  • the assay as disclosed above comprises in the step of incubating a cell in the presence of a compound to be tested, astrocyte conditioned medium in addition and/or wherein the assay further comprises the step of: incubating the cell in the presence of astrocyte conditioned medium, before and/or after the step of incubating a cell in the presence of a compound to be tested.
  • Astrocyte conditioned medium ACM is defined as cell-free medium in which primary human astrocytes have been cultured (see Example 1).
  • the astrocyte conditioned medium which may preferably be fetal astrocyte medium, has an effect on the level of expression of SEQ ID No 1-16, i.e. the level of expression is increased, and thus the BBB function is increased.
  • ACM increases the expression of a nucleic acid sequence selected from the group of SEQ ID No. 1-16, and the said medium thus has an effect on the expression said nucleic acid sequences. Accordingly, compounds may interact with the ACM, or influence the interaction between ACM and the endothelial brain cells. Thus, when compounds interact with ACM or influence the interaction between ACM and the endothelial brain cells, the effect of ACM with regard to increasing the expression of a nucleic acid from SEQ ID No. 1-16 may be modulated. Thus, the expression of a nucleic acid sequence selected from the group of SEQ ID No.
  • 1-16 may be further increased, or is decreased (or even completely blocked), in a cell incubated in the presence of astrocyte conditioned medium and a compound, as compared to a cell incubated in the presence of ACM and the absence of the said compound.
  • compounds may have an effect on the expression of a nucleic acid from SEQ ID No. 1-16 induced by the astrocyte conditioned medium, for instance compounds may interfere, inhibit, enhance pathways/receptors and the like involved in the regulation of the expression of a nucleic acid sequence from the group of SEQ ID No. 1-16 by the astrocyte conditioned medium and thereby increase or decrease the effect of the ACM on the expression level of a nucleic acid sequence from SEQ ID No. 1-16.
  • Such an increasing or decreasing effect may not be observed as effectively without the step of incubating the cell in the presence of astrocyte conditioned medium, although it is to be understood that the use of the astrocyte medium is optional.
  • the expression level of said nucleic acid sequence is upregulated in a cell incubated in the presence of a compound and in the presence of ACM (which may be present before, during and/or after the incubation in the presence of said compound) as compared to a cell incubated without the presence of a compound, such a compound is capable of increasing the BBB function.
  • nucleic acid sequence When the expression level of said nucleic acid sequence is downregulated in a cell incubated in the presence of a compound and in the presence of ACM (which may be present before, during and/or after the incubation in the presence of said compound) as compared to a cell incubated without the presence of a compound, such a compound is capable of decreasing the BBB function.
  • ACM which may be present before, during and/or after the incubation in the presence of said compound
  • compounds that result in either an up- or downregulation of (a) nucleic acid(s) sequence(s) selected from the group of SEQ ID No. 1-16 of at least 2-fold may be considered as compounds that are identified to have the potential to modulate the BBB function, i.e. increase or decrease the BBB function.
  • the cell in the assays as described is a brain endothelial cell.
  • the brain endothelial cell can be a primary cell, but a cell line is preferred.
  • Human primary brain endothelial cells may be obtained from subjects from biopsy material, and thus supply might be limited and restricted by regulations as consent is required, furthermore, primary cells have a limited life span. Therefore, it is preferred to use an immortalized human brain endothelial cell line, such as for instance HCMEC/D3.
  • the expression level of a nucleic acid sequence from SEQ ID No 1-16 is determined. However, it is to be noted that it is also possible to determine the expression level of several nucleic acid sequences from SEQ ID No 1-16, for example simultaneously.
  • the expression level of the nucleic acid sequences in the above assays can be determined with a microarray (Li and Ruan, Anal Bioanal Chem. 2009 Jun;394(4): 1117-24).
  • a microarray is a multiplex technology used in molecular biology and in medicine which consists of an arrayed series of thousands of microscopic spots of oligonucleotides, each containing picomoles of a specific oligonucleotide sequence.
  • This sequence is complementary to a sequence from SEQ ID No 1-16 that are used as probes to hybridize to a target nucleic acid sequence under suitable, preferably high-stringency conditions.
  • the target nucleic acid sequences can be labelled for example with a fluorescent label, and probe-target hybridization can thus be detected and quantified by measuring the fluorescence intensity. For instance, in example 2 it is described in close detail how a microarray may be performed.
  • the expression level is determined with a PCR reaction, preferably a quantitative PCR reaction.
  • the expression level of a nucleic acid sequence selected from the group consisting of SEQ ID No 1-16 may be determined with the use of a reporter construct.
  • a reporter gene construct expresses a reporter gene, for instance Green Fluorescent Protein (GFP) or luciferase.
  • the reporter gene construct thus expresses a mRNA transcript that encodes GFP.
  • GFP Green Fluorescent Protein
  • a complementary target sequence, or multiple complementary target sequences can be introduced, such that a nucleic acid sequence from the group consisting of SEQ D No. 1-16 can bind to the complementary target sequence (Brown BD, et al. Nat Med. 2006 May;12(5):585-91).
  • the reporter gene construct can be introduced in a cell transiently, for instance via transfection, or may be stably introduced, for instance via recombination or transduction.
  • the nucleic acid sequence from the group consisting of SEQ ID No. 1-16 targets the reporter gene construct via complementary base paring and/or RNA interference, the level of expression of the reporter gene is controlled by level of expression of the said nucleic acid sequence, because binding and/or cleavage of the nucleic acid sequence to the reporter transcript reduces mRNA translation into protein.
  • the GFP expression level is inversely proportional to the level of expression of a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID No. 1-16.
  • a reduction in the GFP expression corresponds to an increase in the said nucleic acid sequence expression and thus an increase in the BBB function
  • an increase in GFP expression corresponds to a decrease in the said nucleic acid sequence expression and thus a decrease in the BBB function.
  • the expression level of a nucleic acid sequence selected from the group of SEQ ID No. 1-16 is determined with a microarray, a PCR reaction, a quantitative PCR or a reporter gene construct.
  • HCMEC/D3 cells a brain endothelial cell line
  • EGM-2 medium a brain endothelial cell line
  • astrocyte conditioned medium cell free medium from human astrocytes cultured for 7 days in standard DMEM (Gibco) medium
  • EGM-2 medium containing 2.5% foetal calf serum
  • the ECISTM Model 1600R (Applied BioPhysics, Troy, NY, USA) was used to measure the transendothelial electrical resistance (TEER) for 60-80 hours using ECIS according to the manufacturer's instructions and impedance was measured at 6000 Hz in real time.
  • the TEER values of a control well, in which the endothelial brain cells were incubated without astrocyte conditioned medium (ACM) was about 1250 ohm.
  • the cells incubated with ACM had a value of about 1700 ohm.
  • the endothelial brain cells incubated in ACM had an increased TEER value, representing an increased BBB function. It is concluded that ACM improves BBB function.
  • miRNAs were studied for their involvement in the blood-brain barrier function. Large-scale expression analysis was performed of miRNAs in HCMEC/D3s either or not treated with astrocyte conditioned medium. 13 - 18 ⁇ g total RNA was isolated using Trizol from 1 x 10 6 brain endothelial cells which were cultured in 50% astrocytes-conditioned medium / 50% complete EGM2 for 48 h. Before mRNA and miRNA profiling the RNA samples underwent quality control by Agilent Bioanalyzer Picochip analysis (RIN>9).
  • RNA from sample and reference was labeled with Hy3TM and Hy5TM fluorescent label, respectively, using the miRCURYTM LNA Array power labeling kit (Exiqon, Denmark) following the procedure described by the manufacturer.
  • the Hy3TM-labeled samples and a Hy5TM-labeled reference RNA sample were mixed pair-wise and hybridized to the miRCURYTM LNA array version 11.0 (Exiqon, Denmark), which contains capture probes targeting all miRNAs for human, mouse or rat registered in the miRBASE version 12.0 at the Sanger Institute (Sanger Institute details: http://www.mirbase.org/ ).
  • the hybridization was performed according to the miRCURYTM LNA array manual using a Tecan HS4800 hybridization station (Tecan, Austria). After hybridization the microarray slides were scanned and stored in an ozone free environment (ozone level below 2.0 ppb) in order to prevent potential bleaching of the fluorescent dyes.
  • the miRCURYTM LNA array microarray slides were scanned using the Agilent G2565BA Microarray Scanner System (Agilent Technologies, Inc., USA) and the image analysis was carried out using the ImaGene 8.0 software (BioDiscovery, Inc., USA). Generation and statistical analysis of miRNA expression profiles were performed similarly using R/Bioconductor.
  • HCMEC/D3s Exposure of HCMEC/D3s to astrocyte conditioned medium changed the expression level of miRNAs (Table I; SEQ ID No. 1-16) at least 2-fold as compared to HCMEC/D3s not exposed to astrocyte conditioned medium. It was concluded that an increase of the nucleic acid sequences of SEQ ID No. 1-16 increases the BBB function. Furthermore, it was also concluded that these (putative) miRNA sequences can be used as markers for BBB function, and thus are suitable for use in assays in which compounds are screened for that modulate, i.e. increase or decrease, the BBB function.
  • nucleic acid sequences or nucleic acids comprising nucleic acid sequences with some degree of identity therewith, are suitable for increasing the BBB function, and that nucleic acids with some degree of complementarity with the nucleic acid sequences from SEQ ID No. 1-16 (See Table 1) would be suitable for downregulating the BBB function.
  • Table 1
  • Table 1 should be considered as the true original with the information as intended.
  • RNAi pathway via knock-down of DICER results in a decrease in BBB function
  • the effect of Dicer knock-down with an siRNA against Dicer was studied. Knocking down Dicer should result in blocking the processing of pri-miRNAs and hence prevent incorporation of endogenous miRNAs into the RISC complex. Endothelial brain cells were transfected with an siRNA against DICER (sc-40489, Santa Cruz) or a silencing control siRNA (also obtained from Santa Cruz) and trans fections were performed with DharmaFECT-4 according to the manufacturer's instructions (Dharmacon).
  • results show knocking down the DICER enzyme reduced the resistance of the in endothelial brain cells, confirming that indeed the RNA interference pathway and/or miRNA biogenesis pathway is involved and required for BBB function.
  • MiRNAs were cloned from human genomic DNA and ligated between the Smal and EcoRI restriction sites of pRRL-cPPT-CMV-X2-PRE-SIN-IRES-eGFP vector (kindly provided by Dr. J. Seppen, Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands).
  • Recombinant lentiviruses were produced by co-transfecting subconfluent HEK 293T cells with the specific expression plasmids and packaging plasmids (pMDLg/pRRE, pRSV-Rev and pMD2G) using calcium phosphate as a transfection reagent.
  • HEK 293T cells were cultured in DMEM supplemented with 10% FCS, 1% penicillin/streptomycin, in a 37°C incubator with 5% C02. Infectious lentiviruses were collected 48 h after transfection. The supernatant was centrifuged to remove cell debris and stored at -80°C.
  • hCMEC/D3 human brain endothelial cells were transduced with the lentiviruses containing the microRNAs.
  • the ECISTM Model 1600R (Applied BioPhysics, Troy, NY, USA) was used to measure the transendothehal electrical resistance (TEER) for 60-80 hours using ECIS according to the manufacturer's instructions and impedance was measured at 6000 Hz in real time.
  • hCMEC/D3 Transendothelial migration of primary monocytes Another functional aspect of the BBB is the prevention of monocyte transmigration.
  • Human blood monocytes were isolated from buffy coats of healthy donors by Ficoll gradient and CD14-positive beads (Elkord et al. 2005, Immunology 114, 204-212).
  • Primary monocyte transmigration through a monolayer of hCMEC/D3 was assayed as described previously (Reijerkerk et al. 2008 J. Immunol. 181, 3567-3574). Briefly, hCMEC/D3 cells were seeded onto 96 well plates and infected with lentivirus expressing miR-30a,b,c,d or e for two days.
  • hCMEC/D3 cells were immune activated with TNF alpha (5 ng/ml) for 24 h. Subsequently, 7.5 x 10 4 monocytes were added to 96-well plates containing hCMEC/D3 monolayers. Monocytes were allowed to settle and migrate over a 4-h period. To monitor monocyte migration, cocultures were placed in an inverted phase-contrast microscope (Nikon Eclipse TE300) housed in a temperature-controlled (37°C), 5% C0 2 gassed chamber. A field (220 x 220 ⁇ ) was randomly selected and recorded for 10 min at 50 times by using a digital video camera using Cell F imaging software (Olympus, Heidelberg, Germany).
  • Monocyte diapedesis was assessed by enumerating the number of monocytes within the field that had either adhered or migrated through the monolayer. Transmigrated monocytes could be readily distinguished from those remaining on the cell surface by their highly refractive morphology. The results confirm the data presented above.
  • Tissues were obtained from The Netherlands Brain Bank (NBB), Netherlands Institute for Neuroscience, Amsterdam. All material has been collected from donors from whom a written informed consent for brain autopsy and the use of the material and clinical information for research purposes had been obtained by the NBB.
  • Brain capillaries were isolated from periventricular MS lesions from post-mortem MS patients or non-neurological controls. After resection, the tissues were homogenized using pestles and capillaries were separated from myelin and single cells using DEAE-dextran gradient (15%; Sigma Life Science) centrifuged 25 minutes 2500 rpm without brake. Isolated capillaries were washed on a 40 ⁇ meshed filter (BD Falcon) to remove red blood cells. Finally, RNA was extracted with Trizol and stored in -80°C. qPCR analysis of miRs
  • RNA spike-in RNA spike-in synthesis
  • the samples were incubated at 42oC for one hour followed by heat-inactivation at 95oC for 5 minutes. Samples were cooled to 4oC and thereafter stored at -20oC.
  • the qPCR was performed using the Exiqon "pick and mix" plates (cat# 203801). Three independent samples per plate were tested.
  • the cDNA was diluted 1 : 100 in RNAse free water. Per well, 5 ⁇ of sample and 5 ⁇ of 2x SYBRgreen mastermix were added. Plates were sealed and centrifuged. qPCR was performed on an ABI VILA 7 machine which performed 40 cycles of 95 °C 10 sec, 60 °C 1 minute followed by a melt curve.
  • Isolated brain capillaries were predominantly comprised of brain endothelial cells as indicated by qPCR analysis of ZO-1, claudin-5, P- glycoprotein and VE-cadherin expression at contained minor amounts of astrocytes (glial fibrillary acidic protein a and ⁇ ) and pericytes (platelet-derived growth factor receptor- ⁇ ). Of these, 16 barrier-enhancing micro RN As were diminished in brain capillaries of MS patients (see Figure 1). We conclude that therapeutic application of miRNAs, could re-establish normal functioning of the brain vasculature in endothelial cell based neurological diseases, in particular MS.

Abstract

The blood-brain barrier (BBB) separates the blood and the central nervous system (CNS). Here it is disclosed that blood-brain barrier function can be increased with particular nucleic acid sequences, in particular in patients suffering from or diagnosed with Multiple Sclerosis. The use of compounds comprising such nucleic acid sequences for increasing blood-brain barrier function is described.

Description

miRNA IN BLOOD BRAIN BARRIER OF MS PATIENTS
BACKGROUD ART
The blood-brain barrier (BBB) separates the blood and the central nervous system (CNS). The central nervous system consists of the brain and the spinal cord. This separation between the blood and CNS is created by specialized endothelial cells, brain endothelial cells, which are distinct from peripheral endothelial cells, as the blood-brain barrier is composed of high density cells restricting passage of substances from the bloodstream much more than endothelial cells elsewhere in the body (Abbott et al., Nat Rev Neurosc, 2006, 7, 41-53). The BBB restricts the diffusion of large or hydrophilic molecules into the CNS, e.g. antibodies, but also of microscopic entities, e.g. bacteria. On the other hand, small hydrophobic molecules, e.g. hormones and oxygen, can diffuse over the BBB. The BBB also actively transports molecules, such as for instance glucose, across the BBB utilizing transporter proteins. Cells of the immune system can also pass the BBB (Pachter et al., J Neuropathol Exp Neurol. 2003 Jun;62(6):593-604; Abbott et al., Nat Rev Neurosc, 2006, 7, 41-53; ; Begley and Brightman, Prog Drug Res. 2003;61 :39-78.; Loscher and Potschka, NeuroRx. 2005 Jan;2(l):86-98.; Carvey et al., Neurochem. 2009 Oct;l 11(2):291-314.; Abbott et al., Neurobiol Dis. 2010 Jan;37(l): 13-25). In summary, the function of the BBB is to allow precise control over the substances that leave or enter the central nervous system, and this function is essential for brain homeostasis and reliable functioning of the neuronal environment.
MS is an inflammatory demyelinating disease of the central nervous system (CNS). Dysregulation of the BBB and transendothelial migration of activated leukocytes are among the earliest cerebrovascular abnormalities seen in multiple sclerosis (MS) brains and parallel the release of inflammatory cytokines/chemokines. Mechanisms for breakdown of the BBB in MS are incompletely under stood (Minagar et al. 2003 Multiple Sclerosis 9:540).
On the other hand, in brain abnormalities in which the BBB function is maintained, the BBB can obstruct or hamper the entry of pharmaceuticals or drugs. For instance, many high potential drugs for the central nervous system (CNS), for instance biopharmaceuticals such as antibodies, are currently not or less suitable for use in the treatment of the CNS because these drugs can often not cross the blood-brain barrier, making the BBB a major hurdle for successful CNS drug development that require drugs to pass the BBB.
Current strategies for drug delivery in the brain involve invasive measures such as for instance stereotactic injections in the brain or intrathecal injections, which both means the BBB is physically disrupted and which always involves a risk of neural injuries. Other means include disruption of the BBB by osmotic means, or biochemically by the use of vasoactive substances such as bradykinin or even by localized exposure to high intensity focused ultrasound (HIFU), but success has been limited (Daffertshofer and Fatar, Eur J Ultrasound. 2002 Nov; 16(1- 2): 121-30; Doolittle et al., J Neurosci Nurs. 1998 Apr;30(2):81-90; Wahl et al., Acta Physiol Hung. 1999;86(2): 155-60). As such, there remains a need for methods and compounds that can safely and effectively modulate, i.e. increase or decrease the blood brain barrier function. With an increase of the blood brain barrier function is meant that the blood brain barrier becomes less permeable for the passive transfer of compounds across the BBB, for example (therapeutic) antibodies or antibiotics. With a decrease of the blood brain barrier function is meant that the blood brain barrier becomes more permeable for the passive transfer of compounds such as for example (therapeutic) antibodies or antibiotics across the BBB. Such compounds or methods that would influence the BBB function would be beneficial, as for instance co-administration of a compound that decreases the blood brain barrier with a drug, e.g. therapeutic antibodies or antibiotics, which normally is less capable or incapable of passing the BBB, might result in the improved delivery of said drug to the brain. In addition, a compound that increases the blood brain barrier function may allow the (partial) reconstitution of the blood brain barrier function in MS patients that suffer from impaired BBB function. An increase of the BBB function might for instance prevent bacterial infiltration of the brain or reduce seizures, or help to restore homeostasis.
Unfortunately, the discovery of compounds that may modulate the BBB has been hampered by a lack of understanding of the functioning of the BBB and the pathways involved, as well by the absence of easy methods to determine BBB function. For instance, in vivo animal knockout models were used in which the involvement of certain transmembrane proteins involved in the BBB function such as for instance the G protein-coupled receptor 124 was observed (GPR124) (WO2008147528). Downregulating GPR124 with an antagonist was suggested to decrease the BBB function. On the other hand, increasing expression of GPR124 may provide for increased BBB function. In addition, it has also been suggested that inhibiting claudin-5 expression using RNA interference with hydrodynamic tail vein injections may be used to "open up" the BBB (WO2009047362). In such animal experiments the permeability of the BBB is determined by measuring the uptake by the brain of tracer molecules that normally have difficulty or can not pass the BBB. Obviously, in vivo animal models or knock-out studies do not allow large scale screening for new candidate compounds.
A less complicated assay to study the BBB function is an in vitro assay in which (primary) brain endothelial cells are cultured in vitro on a membrane and passage of tracer compounds through the membrane is determined. Alternatively, the electrical resistance (expressed as ohm) over the cultured brain endothelial cells can also be measured, i.e. the transendothelial electric resistance (Deli et al., Cell Mol Neurobiol. 2005 Feb;25(l):59-127). Importantly, in many of these in vitro assays the cells have to be grown to full confluency in order to ensure that the tracer compounds (or current) has to pass the brain endothelial cell layer and does not simply bypass the membrane without encountering the brain endothelial cells. Although these in vitro models are a major improvement over the in vivo experiments, they are laborious and technically challenging, making large scale screening to identify compounds that influence BBB function still a very difficult task.
In summary, a problem in the development of a medical treatment for the central nervous system, in particular in patients suffering form or diagnosed with multiple sclerosis, e.g. the brain, is that there is a need for compounds that can safely and effectively modulate, i.e. increase or decrease, the blood brain barrier function. In addition, there is a need for assays that are robust and simple to perform to allow for large scale screening for compounds (or compound libraries) that can modulate the blood brain barrier function, and would, for example be suitable in the treatment of blood brain barrier abnormalities in multiple sclerosis.
DETAILED DESCRIPTION
Definitions
In the description and examples, a number of terms are used. In order to provide a clear and consistent understanding of the specification and claims, including the scope to be given to such terms, the following definitions are provided. Unless otherwise defined herein, all technical and scientific terms used have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The disclosures of all publications, patent applications, patents and other references are incorporated herein in their entirety by reference.
As used herein, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. For example, "a" nucleic acid as mentioned below, includes a plurality of molecules (e.g. lO's, 100's, 1000's, lO's of thousands, 100's of thousands, millions, or more molecules).
A nucleic acid or a nucleic acid sequence according to the present invention may include any polymer or oligomer of pyrimidine and purine bases, preferably cytosine, thymine, and uracil, and adenine and guanine, respectively (See Albert L. Lehninger, Principles of Biochemistry, at 793-800 (Worth Pub. 1982)). The present invention contemplates any deoxyribonucleotide, ribonucleotide or peptide nucleic acid component, and any chemical variants thereof, such as methylated, hydroxymethylated or glycosylated forms of these bases, and the like. The polymers or oligomers may be heterogenous or homogenous in composition, and may be isolated from naturally occurring sources or may be artificially or synthetically produced. In addition, the nucleic acids may be DNA or RNA, or a mixture thereof, and may exist permanently or transitionally in single-stranded or double-stranded form, including homoduplex, heteroduplex, and hybrid states. In other embodiments a nucleic acid or nucleic acid sequence of the invention comprises other kinds of nucleic acid structures such as for instance a DNA/RNA helix, peptide nucleic acid (PNA), morpholino, locked nucleic acid (LNA) and/or a ribozyme. Hence, the term "nucleic acid" or "nucleic acid sequence" may also encompass a chain comprising non-natural nucleotides, modified nucleotides and/or non- nucleotide building blocks which can exhibit the same function as natural nucleotides. As used herein, such non-natural nucleotides, modified nucleotides and/or non-nucleotide building blocks are referred to with the term "nucleotide analogues".
The term "% sequence identity" is defined herein as the percentage of nucleotides or nucleotide analogues in a nucleic acid sequence according to the invention that is identical with the corresponding nucleotides in a reference sequence after aligning the two sequences and introducing gaps, if necessary, to achieve the maximum percent identity. Hence, in case a nucleic acid according to the invention is longer than a reference sequence, additional nucleotides in the nucleic acid according to the invention, that do not align with the reference sequence, are not taken into account for determining sequence identity. Methods and computer programs for alignment are well known in the art. One computer program which may be used or adapted for determining the percentage of sequence identity is "Align 2", authored by Genentech, Inc., which was filed with user documentation in the United States Copyright Office, Washington, D.C. 20559, on Dec. 10, 1991.
The term "complementarity" is defined herein as nucleotides of a nucleic acid sequence that can bind to another nucleic acid sequence through hydrogen bonds, i.e. nucleotides that are capable of base pairing. Nucleotides that can form base pairs, that are complementary to one another, are e.g. cytosine and guanine, thymine and adenine, adenine and uracil, guanine and uracil. The percentage complementarity does not have to be calculated over the entire length of a nucleic acid sequence. The percentage of complementarity may be limited to a specific region of which the nucleic acid sequences which are base-pared, e.g. starting from a first base-paired nucleotide and ending at a last base-paired nucleotide. For example, and in an embodiment according to the current invention, a nucleic acid may comprise a nucleic acid sequence of which at least 8 adjacent nucleotides are at least 75% complementary to 8 adjacent nucleotides of the complementary nucleic acid that may comprise 8 nucleotides, 16 nucleotides, or 32 nucleotides, or more. As long as of 8 adjacent nucleotides of the nucleic acid sequence at least 6 nucleotides can base-pair with 8 adjacent nucleotides of the complementary nucleic acid sequence the nucleic acid sequence is at least 75% complementary. The term comprises according to the invention is to be interpreted such that for example a nucleic acid comprising a nucleic acid sequence of 8 adjacent nucleotides may also include a nucleic acid consisting of no more than said 8 adjacent nucleotides.
The term "gene expression" as used throughout, is defined as RNA transcription of a gene, e.g. expression of mRNA, and/or translation of mRNA into protein. Gene expression thus comprises RNA expression and/or protein expression. For example, miRNA genes may express RNA transcripts that are subsequently processed by the miRNA machinery. For a miRNA gene, gene expression thus comprises the transcript that is initially transcribed, the subsequent intermediate processed transcripts, and finally the miRNA molecule which may be incorporated in the RISC complex. Furthermore, gene expression of a gene encoding a protein comprises the initially transcribed RNA molecule, but may also comprise subsequently spliced or otherwise by the cellular machinery processed transcribed RNA molecules, and/or the protein which may be translated from messenger RNA (mRNA). When reference is made to the "expression level" of a gene, the expression level is to be understood as the amount of RNA transcript that is transcribed by a gene and/or the amount of protein that may be translated from an RNA transcript, e.g. mRNA. For example, for genes which encode a miRNA, the expression level may be determined through quantifying the amount of RNA transcript which is expressed, e.g. using standard methods such as quantitative PCR of a mature miRNA, microarray, or Northern blot. Alternatively, the expression level may also be determined through measuring the effect of a miRNA on a target mRNA. For example, a 3 'UTR sequence comprising a target miRNA target sequence may be incorporated in a reporter gene, e.g. luciferase, this way, the expression level of a miRNA gene may be indirectly measured by measuring the amount of Luciferase expression which may be controlled by a particular miRNA. Furthermore, in case the 3 'UTR sequence is from a particular gene, the amount of Luciferase expression also correlates with the expression level of the particular gene. As such, when determining the expression level of a miRNA as described above (e.g. directly through qPCR or indirectly with a reporter gene construct), in addition, the expression level of the miRNA may be correlated to the expression level of a target gene of the particular miRNA. The amount of protein that may be translated from an RNA transcript may also be measured to determine the expression level of a gene. Methods are known in the art, such as ELISA and Western blot. Alternatively, the expression level of a protein may also be indirectly measured. For example, in case the expression level of an RNA transcription factor needs to be determined, a reporter gene construct may be used which comprises the RNA transcription factor binding site. The expression level may than be subsequently determined by measuring the reporter gene construct expression, e.g. in case GFP is used, green fluorescence intensity may be determined as a measure of the expression level of the RNA transcription factor.
Description
The present invention now surprisingly solves at least one of the problems as described above. It was found for the first time that expression of specific miRNA in multiple sclerosis is impaired, reduced, relative to expression of such miRNA's in healthy subjects, that these miRNA's can increase blood brain barrier function and that increasing the level of such miRNA's can improve blood brain barrier function, in particular in multiple sclerosis.
In the current invention it was found for the first time that particular nucleic acids, including those listed in SEQ ID No. 1-16, play an important role in modulating the BBB function, in particular in patients diagnosed with or suffering from Multiple Sclerosis. The examples show that expression of miRNA with a sequence according to SEQ ID NO 1 - 16 is impaired in material obtained from patients suffering from or diagnosed with multiple sclerosis. As the same time it is disclosed these nucleic acids increase blood brain barrier function. Compounds comprising such nucleic acid are thus useful in increasing blood brain barrier function, in particular in multiple sclerosis. As can be witnessed in detail from the examples, it was found that the nucleic acids according to the invention, and including those selected from the group consisting of SEQ ID No. 1-16, are capable of increasing the blood brain barrier function, which is in particular useful in patients diagnosed with or suffering from multiple sclerosis.
Furthermore, blocking nucleic acids selected from the group consisting of SEQ ID No. 1-16, may result in a decrease in the blood brain barrier function, for example in healthy humans or other patients, but not necessarily suffering from multiple sclerosis (i.e. in those subjects in which expression of the disclosed miRNA is not or only to a lesser extent impaired). In the current invention, blocking of a nucleic acid is achieved by providing a (partially) complementary nucleic acid sequence that can (partially) hybridise to the nucleic acid, such that when the (partially) complementary nucleic acid sequence is hybridised with the nucleic acid (and thus bound thereto), the nucleic acid can not perform its function, e.g. hybridizing with a target molecule, i.e. it is blocked.
Finally, the relative expression level of the nucleic acids listed in SEQ ID No. 1-16 can be used as a measure/indicator of blood brain barrier function, in particular in multiple sclerosis. When the expression level of a particular level of such nucleic acid is decreased, relative to a control setting, the BBB function is decreased. Such an increase or decrease of the level of expression of a nucleic acid may be induced by the presence of a compound. Thus, the invention further provides for an assay (method) for screening for compounds or compound libraries to identify compounds that are capable of modulating BBB function. The assay comprises of determining the expression level(s) of at least one nucleic acid sequence as listed in SEQ ID No. 1-16 in the absence and/or presence of a compound to be tested. Compounds that would increase expression of at least one of the miRNA with a sequence according to any one of SEQ ID NO 1 - 16 would be useful in the treatment of blood brain barrier function, in particular in multiple sclerosis. In a first aspect of the invention there is provided for the use of a compound, preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100%) sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis.
For instance, said compounds may be used in vitro or in vivo for increasing the blood brain barrier function. With in vivo is meant for instance in an animal model or a patient, with in vitro is meant a (primary) cell culture. In certain embodiments, the use does not entail the treatment of the human or animal body by surgery or therapy. The nucleic acids may be provided in the cell, for instance via transfection or other means of cell transfer, as explained below. A nucleic acid according to the invention for example can comprise a nucleic acid sequence which has 95% sequence identity with SEQ ID No. 1. Such a nucleic acid can be the exact length of SEQ ID No. 1, or be longer (e.g. 1,2,3,4,5,6,7,8,9 nucleotides longer) or shorter (e.g. 1,2,3,4,5,6,7,8,9 nucleotide shorter). A skilled person will be able to test how many nucleotides he can remove or add to a nucleic acid according to SEQ ID NO 1-16, and still increase blood brain barrier function, in particular in multiple sclerosis.
The blood-brain barrier function can, for example, be measured by determining the transendothelial electric resistance (TEE ) of an endothelial brain cell monolayer, as is described in detail in Example 1. TEER is expressed in ohm. For instance, the ECISTM Model 1600R (Applied BioPhysics, Troy, NY, USA) can be used to measure the TEER according to the manufacturers instructions. By measuring TEER the resistance of an endothelial brain cell monolayer to an electric current is measured, i.e. the ion permeability of the endothelial brain cell monolayer. A relative high resistance means that less ions, and thus less current, can permeate the endothelial brain cell monolayer, a relative low resistance means that more ions, and thus more current, can pass through the endothelial brain cell monolayer. Ion permeability of the endothelial brain cell monolayer correlates with the ability of compounds to passively diffuse across the blood brain barrier. Thus, the TEER value measured relates to the BBB function. Within the context of the current invention, increasing the blood-brain barrier function means that relative to a control setting, the TEER is increased when measured in the method described. Also, decreasing the blood-brain barrier function means that relative to a control setting, the TEER is decreased when measured in the method described. A control setting is defined as a setting in which certain conditions (e.g. salt, nutrient and biological factor concentrations, H, temperature etc.) are selected. For instance, conditions may be selected that most closely resemble the conditions of a healthy patient, but for instance conditions may also be selected such as may occur in a diseased or injured patient in which BBB function is known to be disturbed. Whichever conditions selected, in the control setting the TEER is measured. Next, in a different setting comprising e.g. a composition for increasing or decreasing the blood-brain barrier function, the TEER is determined. In this different setting similar conditions are selected as in the control setting, but now a composition or compound is added that can, for instance, increase the BBB function. When comparing the TEER resistance value (ohm) for the latter condition, relative to the control setting, the TEER will have increased, i.e. having a higher value and thus a higher resistance, meaning that less ions, and thus less compounds, can pass through the endothelial cell layer, and the BBB function has increased. The BBB function as defined is within the context of the assay. An increase or decrease of the BBB function within the assay indicates that in alternative settings wherein the blood brain barrier is studied, e.g. in in vivo, or in vivo models, or in a therapeutic setting the blood brain barrier may have a similar effect and result in an increase or decrease of the blood brain barrier function in these settings, e.g. in a patient, e.g. a patient diagnosed with or suffering from Multiple Sclerosis.
Surprisingly it was found that, when comparing (control vs. astrocyte conditioned medium) the expression level of putative miRNA sequences, a selection of putative miRNA sequences was at least two-fold upregulated in the blood-brain barrier, i.e. in brain-endothelial cells, see Example 2 and SEQ ID No. 1-16. Furthermore, it was found that an increase in the expression of said nucleic acid sequences in the blood brain barrier, i.e. in brain endothelial cells, was found to result in an increase in the BBB function. Support for the involvement of miRNA sequences in the BBB function was further provided as it was found that knocking down an essential factor from the miRNA biogenesis pathway, the DICER enzyme (Winter et al., Nature Cell Biol 2009, vol 1 1 , nr 3, p.228-234), resulted in a decrease in BBB function (example 3). Thus, the miRNA biogenesis pathway, and consequently, miRNA function is implicated in BBB function. Next it was found that in material derived from multiple sclerosis patient's specific miRNA are reduced or expressed at a lower level in comparison to subject not suffering from or diagnosed with multiple sclerosis. With respect to these miRNA's, the discovery of miRNAs, a new class of endogenous small non-coding RNAs, has dramatically changed the perspective on gene regulation. With non- coding RNAs is meant RNAs that do not encode for a protein, i.e. are not translated into a protein such as coding RNAs, e.g. mRNA. Without being bound by theory, it is the inventors' belief that these small non-coding RNAs according to the invention may be involved in the regulation of gene expression and are activated through incorporation into an enzyme complex called the RNA-induced silencing complex (RISC), via the RNA interference pathway. Protein expression is thought to be regulated through (partial) complementary base-pairing of the putative miRNA with target sequences in the 3' untranslated region of target mRNA, thereby causing degradation of the mRNA and/or translational repression, leading to improved BBB function. In patients suffering from multiple sclerosis the levels are reduced, leading to decreased blood brain barrier function. These putative miRNAs may also possibly act via transcriptional gene silencing or even activation of transcription and/or translation as this has also been occasionally described for (putative) miRNA sequences (Morris, Biotechniques. 2006 Apr;Suppl:7-13; Chua et al., Curr Opin Mol Ther. 2009 Apr;l 1(2): 189-99). These other possible mechanisms all can depend on base-pairing of putative miRNAs with for instance chromosomal DNA, thereby inducing chromatin remodelling that can result in transcriptional activation or silencing. A (putative) miRNA can also interact with a messenger RNA in order to stabilize it thereby preventing degradation and/or processing. Furthermore, other modes of activity of the putative miRNA sequences according to the invention, and leading to improved BBB functioning are not excluded by the invention. Hence, the term miRNA activity includes the modes of activity as described above.
Thus, according to the invention, there is provided for use of a compound, preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%, even more preferably 90%», even more preferably 95%», most preferably 100%» sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis. In certain embodiments, the use does not entail the treatment of the human or animal body by surgery or therapy.
Alternatively, there is provided for a compound, preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%>, even more preferably 90%o, even more preferably 95%, most preferably 100%) sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for use in increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis.
Alternatively, there is provided for use of a compound, preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for the manufacture of a medicament for increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis. Preferably, the compound described above (and the use thereof) is or comprises miR-15a, miR-18b, miR-26a, miR-27a, miR-27b, miR-30a, miR-30b, miR-30c, miR-30d, miR-30e, miR-31, miR-106b, miR-125a-5p, miR-155, miR-195, or miR-487b. The sequences of these miRNA's are given in SEQ ID NO 1 - 16. These nucleic acids or use thereof according to the invention have been found to be particular advantageous for increasing the blood-brain barrier function, for example as determined according to the methods disclosed in the Examples, in particular in patients suffering from or diagnosed with multiple sclerosis. Preferably, a nucleic acid according to the invention has a length of at most 100, more preferably at most 75, even more preferably at most 50, most preferably at most 25 nucleotides.
The skilled person is well capable to determine whether a nucleic acid according to the invention increases the BBB function. For instance, a nucleic acid according to the invention can be transfected or transduced into a brain endothelial cell as described in example 3 and 4, and the effect on the TEER can be measured. It is to be understood that the term comprising within the context of the above is meant to also include a nucleic acid according to the invention consisting of the nucleic acid sequence having 70%», 75%», 80%», 85%», 90%», 95%, or 100%» sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID No. 1-16. Thus, the nucleic acid sequence which comprises a nucleic acid sequence having 70%, 75%, 80%, 85%, 90%, 95%, or 100% sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID No. 1-16, can consist of the same number of nucleotides of a nucleic acid sequence from SEQ ID No. 1-16, but may also comprise additional nucleotides, for instance 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 100 nucleotides, or any other integer, as long as the nucleic acid according to the invention is capable of increasing the BBB function. Furthermore, the nucleic acid may also consist of a fragment of a sequence according to SEQ ID No. 1-16, such a fragment may be for instance as small as 10 nucleotides, although the fragment may be larger or smaller, as long as the fragment is capable of increasing the BBB function, as can be determined according to the methods provided by the invention.
In a preferred embodiment, the compound, preferably a nucleic acid, as described above, comprising a nucleic acid sequence which has at least 70%, 80%, 85%, 90%, 95%, or 100% sequence identity with a nucleic acid sequence selected from the group of SEQ ID No 1-16 is a pre-miRNA, a pri-miRNA, a siRNA or a shRNA.
Without being bound by any theory, the sequences as listed, are in the art referred to as microRNA or miRNA sequences (Bartel D. P. (2004) Cell 116, 281-297). miRNAs are a novel class of endogenous small non-coding RNAs, which are believed to regulate gene expression via the so-called RNA interference pathway (RNAi). The primary transcripts of miRNAs can be transcribed by RNA polymerase II and III which generally have a length of several kilobases, and are called primary miRNAs (pri-miRNA). A typical animal pri-miRNA comprises an imperfectly paired stem of 30-35 bp, with a terminal loop and flanking segments (Bartel (2004) Cell 116, 281-297). These pri-miRNAs , which are capped and polyadenylated (Kim (2005) Nature Rev. Mol. Cell. Biol. 6 376-385), are subsequently processed by the miRNA biogenesis pathway (reviewed in Winter et al., Nature Cell Biol 2009, vol 11, nr 3, p.228-234), in the cell nucleus to shorter, 70-100 nucleotide stem- loop structures known as pre -miRNAs. This processing can be performed in animals by the RNase III endonuclease Drosha. Pre-miRNAs can subsequently be transported into the cytoplasm by exportin-5, where they can be processed to double-stranded miRNAs, a miRNA duplex, with a length of 21-25 nucleotides by the RNase III endonuclease DICER. The miRNA duplex comprises the miRNA nucleic acid sequence, which may be subsequently incorporated into the RNA-induced silencing complex (RISC). When the miRNA is incorporated into RISC, this activated RISC complex, i.e. a RISC complex loaded with a miRNA, can inhibit gene expression of a target gene by inhibiting translation and/or by cleaving mRNA through complementary base-pairing, although other regulatory mechanisms can be envisaged as disclosed above. One of the key differences between miRNAs and most siRNAs (small interfering RNA) is that most species of a miRNA have a highly conserved end, although there is variation (Carthew and Sontheimer (2009) Cell 136, 642-655). For example, such highly conserved end consists of 5- 9 bases that are identical within a species of a miRNA. In contrast, siRNAs tend to be much more heterogeneous in end composition.
As the nucleic acid sequences of SEQ ID No 1-16 may represent human miRNA sequences, the pre-miRNA or pri-miRNA according to the invention may comprise the natural sequences from which such a miRNA is processed, i.e. the endogenous sequence as it is expressed in nature. However, non-natural nucleic acid sequences, i.e. exogenous artificial pre-miRNA, pri- miRNA or a miRNA duplex RNA structures are also possible (Aagaard et al., Gene Ther. 2008 Dec; 15(23): 1536-49; Liu et al., Nucleic Acids Res. 2008 May;36(9):2811-24). In that respect, it is also be possible to incorporate the nucleic acid sequences according to the invention in so-called small interfering RNA (siRNA) (Elbashir et al., Nature. 2001 May 24;411(6836):494-8) or short hairpin RNA (shRNA) (Brummelkamp et al., 2002 Apr 19;296(5567):550-3). A short hairpin RNA can comprise two short complementary RNA sequences of 19-35 nucleotides long with a loop sequence in between such that a hairpin structure can be formed. A small interfering RNA may comprise two complementary strands of 19-25 nucleotides. A shRNA may also be processed by the RNA interference pathway, by the DICER enzyme, resulting in an siRNA. As with a duplex miRNA, one of the strands of the siRNA duplex has been suggested to be incorporated into RISC, resulting in an activated RISC complex.
Thus, a nucleic acid comprising a nucleic acid sequence which has at least 70%, 80%, 85%, 90%», 95%», 100%» sequence identity with a nucleic acid sequence selected from the group of SEQ ID No 1-16, is preferably a pre-miRNA, pri-miRNA, siRNA, shRNA, or is hybridised to a complement nucleic acid sequence thus forming a siRNA duplex or miRNA duplex, such that the nucleic acid sequence with at least 70%, 80%, 85%, 90%, 95%, 100% sequence identity with a nucleic acid sequence selected from the group of SEQ ID No. 1-16 is compatible with the RNA interference pathway. Thus, according to this embodiment, the provided nucleic acid may be recognized and processed by the RNA interference pathway or miRNA biogenesis pathway (Winter et al, Nature Cell Biol 2009, vol 11, nr 3, p.228-234) such that the nucleic acid sequence which has at least 70%, 80%, 85%, 90%, 95%, 100% sequence identity with a nucleic acid sequence selected from the group of SEQ ID No 1-16 can have its action for increasing the blood-brain barrier function. In a further embodiment, a compound, preferably a nucleic acid according to the invention as disclosed above, is for use in a treatment for increasing blood-brain barrier function. It has been found that in particular these nucleic acids can advantageously be used to increase the blood-brain barrier function. In particular, such a treatment may be a medical treatment of a patient suffering from a neurological condition, preferably multiple sclerosis. For many neurological conditions, BBB damage is an early event, and thus increasing BBB function as an early intervention for treating such conditions may slow down, stop or even reverse such neurological conditions. Preferably, a compound, preferably a nucleic acid according to the invention as disclosed above is for use in treatment of multiple sclerosis, in particular to increase blood brain barrier function. It is beneficial to provide patients with a treatment that increases the blood-brain barrier such that homeostasis may be (partially) restored and neuronal damage reduced. It is to be noted that the use of a single compound, preferably nucleic acid according to the invention may suffice for increasing the blood-brain barrier function, however, it is also well within the scope of the invention to combine multiple compounds, preferably nucleic acids according to the invention such that an enhanced effect for increasing the blood-brain barrier is obtained. Thus, the invention provides for the use of at least one nucleic acid according to the invention as described above, for use in a medical treatment, in particular for increasing the BBB function in a patient, in particular a patient diagnosed with or suffering from multiple sclerosis. According to another aspect of the current invention, there is provided for nucleic acids, and the use thereof, that can bind, hybridize and/or base-pair to a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID No 1-16, thereby blocking the function thereof, for decreasing the blood brain barrier function according to the invention. Preferably such a nucleic acid binds, hybridizes and/or base-pairs (i.e. forms base pairs) with (part of) the a nucleic acid that comprises or consists of a nucleic acid sequence selected from the group consisting of SEQ ID No 1-16. Such blocking nucleic acids are also known in the art for instance as antagomirs (Kriitzfeldt et al., Nature, 2005 Dec l;438(7068):685-9). In a further embodiment a compound, preferably a nucleic acid, is provided comprising a nucleic acid sequence of which at least 8 (or at least 10, 15, or 20) adjacent nucleotides are at least 70%, preferably 75%, more preferably 80%,even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% complementary to 8 (or at least 10, 15, or 20) adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16.
Also provide is use of a compound, preferably a nucleic acid, comprising a nucleic acid sequence of which at least 8 (or at least 10, 15, or 12) adjacent nucleotides are at least 70%, preferably 75%, more preferably 80%, even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% complementary to 8 (or at least 10, 15, or 20) adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1- 16, for decreasing blood -brain barrier function. In certain embodiments, the use does not entail the treatment of the human or animal body by surgery or therapy.
In other words, such nucleic acid has for example 8, 9, or 10 adjacent nucleotides that are complementary to 8, 9 , or 10 adjacent nucleotides of a nucleic acid sequence from the group of SEQ ID No 1-16. These nucleic acids can in particular be used for decreasing the blood brain barrier function. For example, a nucleic acid according to this embodiment of exactly 8 nucleotides, or 16 nucleotides, or 30 nucleotides in size, or even larger, and said nucleic acid comprises a nucleic acid sequence of 8 nucleotides that are (partially) complementary to a nucleic acid from the group of SEQ ID No. 1-16. In case the nucleic acid is longer than 8 nucleotides, the additional nucleotides do not necessarily have to base pair, i.e. be complementary, with the nucleic acid sequence from SEQ ID No. 1-16.
Such a compound, comprising such complementary nucleic acid, is capable of binding to a nucleic acid selected from the group of SEQ ID No. 1-16, to thereby blocking said nucleic acid. The term blocking herewith is being defined as preventing the nucleic acid sequence of SEQ ID No. 1-16 from performing its function as described herein, e.g. having a miRNA activity or increasing the BBB function.
As nucleic acid sequences from the group of SEQ ID No. 1-16 are involved in the BBB function, i.e. an increased concentration of said nucleic acid results in an increased BBB function, blocking the said nucleic acid sequences with a complementary nucleic acid sequence has been found to block its normal function, e.g. may be used for decreasing the BBB function. Thus, as a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID No 1-16 is involved in maintaining or increasing BBB function, blocking a nucleic acid comprising a nucleic acid sequence selected from the group of SEQ ID No 1-16 may result in a decrease of BBB function. A decrease in BBB function means that relative to a control, the TEER is decreased, as described above.
As an alternative, there is provided a compound, preferably a nucleic acid, comprising a nucleic acid sequence of which at least 8 (or at least 10, 15, or 20) adjacent nucleotides are at least 70%, preferably 75%, more preferably 80%, even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% complementary to 8 (or at least 10, 15, or 20) adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16, for use in decreasing blood-brain barrier function. As an alternative, there is provided for the use of compound, preferably a nucleic acid, comprising a nucleic acid sequence of which at least 8 (or at least 10, 15, or 20) adjacent nucleotides are at least 70%, preferably 75%, more preferably 80%, even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% complementary to 8 (or at least 10, 15, or 20) adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16, for the manufacture of a medicament for decreasing blood-brain barrier function.
In an another embodiment, there is provided that the nucleic acid comprising a nucleic acid sequence of which at least 8 adjacent nucleotides are at least 85%, most preferably 100% complementary to 8 adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16 for decreasing the blood brain-brain barrier function further comprises nucleotide analogues. Nucleotide analogous, such as for instance Locked Nucleic Acids (Chan et al., Clin Exp Pharmacol Physiol. 2006 May-Jun;33(5-6):533-40)„ are in particular advantageous as nucleotide analogous can increase the stability of the nucleic acids, i.e. they may provide resistance to nucleases, but can also increase the strength of the complementary base-pairing (binding affinity) between the nucleic acid sequence and the target sequence, i.e. a nucleic acid sequence selected from the group of SEQ ID No 1-16. As long as the nucleic acid sequence comprising the nucleotide analogue is capable of complementary base-pairing with its target sequence and/or reducing the blood brain barrier function, the skilled person may select such analogues, however, it is preferred to select analogues that result in an improvement of the stability and/or binding affinity. Other nucleotide analogues are well known in the art, such as for instance morpholinos (Summerton and Weller Antisense Nucleic Acid Drug Dev. 1997 Jun;7(3): 187-95). Accordingly, (a) nucleotide analogue(s) may result in an increased stability of the nucleic acid, and may improve the binding of the nucleic acid to its target, thereby improving its capacity to block its target, i.e. improving the blocking of a nucleic acid sequence selected from the group of SEQ ID No 1-16, for decreasing the blood brain-brain barrier function. For instance, it is known in the art that polyoma viruses, e.g. JCV, HIV-1, or Japanese encephalitis virus may passage through the blood brain barrier. By decreasing the BBB function, the viral infiltration of the CNS may be more effectively treated with antiviral medication, as the antiviral medicine may more easily penetrate the brain to have its action. Similarly, in the above neurological conditions, it may be beneficial in a relatively early stage of disease to decrease the blood brain barrier function, such that the brain may become accessible to pharmaceuticals that would otherwise not be effective. By combining the drug with the nucleic acid according to the invention for reducing the BBB function, the drug may now effectively reach the brain cancer. Accordingly, such a treatment preferably further comprises the use of a medicament that would benefit from decreasing the blood-brain barrier function, thereby improving the medicaments ability to cross the blood-brain barrier and entering the CNS and thus the spinal cord and/or brain. It is to be noted that it is envisaged that a single nucleic acid sequence may be sufficient for decreasing the blood-brain barrier function; however, it is also within the scope of the invention to combine multiple nucleic acids for decreasing BBB function according to the invention such that an improved decrease of the BBB function can be obtained.
Furthermore, it is also within the scope of the invention to provide a nucleic acid that encodes a nucleic acid according to the invention, for instance DNA constructs that drive expression of an siRNA duplex or of a nucleic acid (partially) complementary to a nucleic acid sequence selected from SEQ ID No 1-16. Thus, the nucleic acid may also be synthesised de novo, for example by the cell itself.
Different modes or strategies for delivery of the nucleic acid to the brain endothelial cells also fall within the scope of the invention. For instance, in one embodiment, a nucleic acid according to the invention may be coupled to a ligand for delivery into the brain endothelial cells. By coupling a ligand to a nucleic acid according to the invention, the ligand can mediate the uptake of the nucleic acid by the brain endothelial cells, upon which the nucleic acids can execute its function. For instance, a cholesterol conjugate may be used as a ligand and can be used as follows: a nucleic acid is provided, chemically modified and labelled with a cholesterol moiety (Soutschek et al., J Nature. 2004 Nov l l;432(7014): 173-8).The nucleic acids can then for instance be injected intravenously, where the cholesterol group enables the siRNAs to be taken up by cells. Alternatively, peptide or nucleotide aptamers may also be used as ligands, and the nucleic acid according to the invention may be coupled thereto (as reviewed e.g. in Cerchia et al. Methods Mol Biol. 2009;535:59-78 and Borghouts et al.,Comb Chem High Throughput Screen. 2008 Feb;l 1(2): 135-45). Aptamers are molecules that are capable of binding to a specific target molecule, for instance a receptor on a cell, resulting in cellular uptake. Aptamers are usually created by selecting them from a large random sequence pool, but natural aptamers also exist in riboswitches. Aptamers are used for both basic research and clinical purposes as macromolecular drugs. These compound molecules have additional research, industrial and clinical applications. Nucleotide aptamers normally contain (usually short) strands of oligonucleotides. Peptide aptamers normally contain a short variable peptide domain, which are often attached at both ends to a protein scaffold. Aptamers can be part of a nucleic acid .(Dassie JP, et al, Nat Biotechnol. 2009 Sep;27(9):839-49, McNamara JO 2nd, et al., Nat Biotechnol. 2006 Aug;24(8): 1005-15). Thus, according to the invention, the nucleic acid may be coupled to a ligand which enables the delivery of the nucleic acid to brain endothelial cells, such that the nucleic acid sequences can have its modulating effect, i.e. increasing or decreasing the blood brain barrier function. In an alternative embodiment, delivery of a nucleic acid according to the invention may be achieved through a delivery vehicle comprising the nucleic acid. All kinds of delivery vehicle are suitable, as long as the delivery vehicles are capable of introducing the nucleic acid according to the invention in a cell. For instance, a delivery vehicle may be a gene therapy vector. Suitable vectors for instance comprise adenoviral vectors, adeno-associated viral (AAV) vectors, gamma retroviral vectors and lentiviral vectors. Furthermore, a delivery vehicle according to the present invention may also comprise a plasmid delivery system, a virus like particle (VLP), a stable nucleic acid lipid particle (SNALP), a cationic delivery system, a cationic liposomal delivery system, a cationic polymer, a lipoplex, or a liposome. Virus like particles contain viral protein(s) derived from the structural proteins of a virus. In some cases these proteins are embedded within a lipid bilayer. These particles resemble the virus from which they were derived but lack viral nucleic acid, meaning that they are not infectious. Also VLPs are suitable for packaging plasmids or any other nucleic acid, such as for instance pre-miRNAs, pri-miRNAs, shRNAs, miRNA duplexes, siRNA duplexes. By modifications of the viral proteins and linkage of other molecules to the surface of the particle their tropism can be modulated. Cationic macromolecules can also be used for delivery vehicles, the two major classes being lipids and polymers. Examples of cationic vectors are liposomes (lipoplexes) and polyethelyenimine (PEI) (polyplexes).
The invention also includes the use of a nucleic acid according to the invention, the use of a nucleic acid coupled to a ligand according to the invention, or the use of a delivery vehicle according to the invention, for modulating the blood-brain barrier function.
The nucleic acids according to the invention, i.e. for increasing or decreasing the blood-brain barrier function, which may also comprise a nucleic acid coupled to a ligand or a delivery vehicle according to the invention, may be incorporated into a pharmaceutical composition suitable for administration. Such a pharmaceutical composition may comprise a pharmaceutically acceptable carrier, which is intended to include any and all solvents, dispersion media, coatings, isotonic and adsorption delaying agents and the like compatible with pharmaceutical administration. Furthermore, the pharmaceutical composition may also comprise medicaments that are suitable for use in combination with a nucleic acid according to the invention in a treatment for increasing and/or decreasing the blood-brain barrier. Routes of administration of pharmaceutical compositions according to the invention comprise oral administration, application on the skin, or injection, but are not limited thereto. In another embodiment, a method is provided for modulating the blood brain barrier by providing the nucleic acids as described above. The blood brain barrier can be modulated in vitro or in vivo. When a method involving modulation of the blood brain barrier is in a human and in vivo, such a method does not involve a method for treatment of the human or animal body practised on the human or animal body. When in any of the methods embodiments, the nucleic acid, or delivery vehicle is provided in vitro, the method may comprise providing a vessel or a dish which contains or comprises the nucleic acid, or delivery vehicle. For example, there may be provided the nucleic acid, or delivery vehicle that is freeze dried in a well (e.g. Reinisalo et al., J Control Release. 2006 Jan 10;110(2):437-43 and del Pozo- Rodriguez et al., Eur J Pharm Biopharm. 2009 Feb;71(2): 181-9). In a further embodiment, the nucleic acid or delivery vehicle is introduced in a cell, preferably a brain endothelial cell. Said brain endothelial cell can be a primary cell or a cell line, preferably HCMEC/D3 (Weksler et al., the FASEB Journal. 2005;19: 1872-1874). The current invention now provides for the above methods, and now makes it possible to modulate, e.g. in vitro, the blood-brain barrier by the provision of said nucleic acid, protein or delivery vehicle. These methods may in particular be useful in the study of the blood-brain barrier, in particular in multiple sclerosis.
Alternatively, the methods of the invention also provides for the modulation of the blood brain barrier in animal models that would be of particular interest for studying the effect of modulating BBB function. For instance, animal models exist for multiple sclerosis. Studying the effect of the nucleic acids, and/or delivery vehicles according to the invention in these models may be of interest for the development of novel therapies, but is not limited thereto. As long as in an animal model for a disease wherein the blood brain barrier is impaired, it would be of interest to study the effect of increasing or decreasing the blood brain barrier permeability, the nucleic acids and proteins according to the invention may be used.
The invention further also provides for an assay (method) for screening for compounds that may modulate the blood brain barrier function, preferably that increases blood brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis, comprising the steps of incubating a cell in the presence of a compound to be tested (i.e. screened), and determining the expression level of a nucleic acid sequence selected from the group of SEQ ID No. 1-16, wherein the expression level of said nucleic acid sequence is compared with the expression level of said nucleic acid sequence determined in a cell incubated without the presence of said compound to be tested (i.e. screened).
Cells may for instance be contained in multi-well plates either in suspension or attached. The cells may be incubated in the presence of a compound, for instance by adding the compound to the cell culturing medium. Cell culturing medium is defined as a solution which provides the cell with nutrients such that the cells may be sustained. Alternatively, the compound may also be provided in a suitable standard buffer, such as for instance phosphate buffered saline (PBS), for a period of time such that the cells are contacted with the compound. Any method incubating the cells with the compounds are allowed, as long as the compound may have its effect on the cells. When the cells have been incubated with the said compound, the nucleic acid sequence expression level is determined. The nucleic acid sequences from the group of SEQ ID No. 1-16 are suitable in a screening assay for compounds that modulate the BBB. The term modulation means either increasing or decreasing. When the expression level of a said nucleic acid sequence is upregulated in a cell incubated in the presence of a compound as compared to a cell incubated in the absence of said compound, such a compound is capable of increasing the BBB function, and when the expression level of a said nucleic acid sequence is downregulated in the presence of a compound as compared to a cell incubated without the presence of said compound to be tested, such a compound is capable of decreasing the BBB function. In general, compounds that result in either an up- or downregulation of a nucleic acid sequence selected from the group of SEQ ID No. 1-16 of at least 2-fold may be considered as compounds that are identified to have the potential to modulate the BBB. In general, the higher the up- or downregulation, the lowest number of potential compounds may be identified but these compounds also may have a stronger effect on BBB modulation. Not only single compounds may be tested, but it is also envisaged that combinations of compounds may be tested for their potential effect on modulation of the BBB function. Furthermore, it is also within the scope of the invention to determine, in an assay for screening for compounds that modulate the BBB function, the expression levels of more than one nucleic acid sequence selected from the group consisting of SEQ ID No. 1-16. In view of the disclosure herein, compounds able to increase the expression of at least one of the miRNA's with a sequence according to SEQ ID NO 1- 16 are useful in improving blood brain barrier function, in particular in patients diagnose with or suffering from multiple sclerosis.
In a different embodiment, the assay as disclosed above comprises in the step of incubating a cell in the presence of a compound to be tested, astrocyte conditioned medium in addition and/or wherein the assay further comprises the step of: incubating the cell in the presence of astrocyte conditioned medium, before and/or after the step of incubating a cell in the presence of a compound to be tested. Astrocyte conditioned medium (ACM) is defined as cell-free medium in which primary human astrocytes have been cultured (see Example 1). The astrocyte conditioned medium, which may preferably be fetal astrocyte medium, has an effect on the level of expression of SEQ ID No 1-16, i.e. the level of expression is increased, and thus the BBB function is increased. Therefore, ACM increases the expression of a nucleic acid sequence selected from the group of SEQ ID No. 1-16, and the said medium thus has an effect on the expression said nucleic acid sequences. Accordingly, compounds may interact with the ACM, or influence the interaction between ACM and the endothelial brain cells. Thus, when compounds interact with ACM or influence the interaction between ACM and the endothelial brain cells, the effect of ACM with regard to increasing the expression of a nucleic acid from SEQ ID No. 1-16 may be modulated. Thus, the expression of a nucleic acid sequence selected from the group of SEQ ID No. 1-16 may be further increased, or is decreased (or even completely blocked), in a cell incubated in the presence of astrocyte conditioned medium and a compound, as compared to a cell incubated in the presence of ACM and the absence of the said compound. For example, compounds may have an effect on the expression of a nucleic acid from SEQ ID No. 1-16 induced by the astrocyte conditioned medium, for instance compounds may interfere, inhibit, enhance pathways/receptors and the like involved in the regulation of the expression of a nucleic acid sequence from the group of SEQ ID No. 1-16 by the astrocyte conditioned medium and thereby increase or decrease the effect of the ACM on the expression level of a nucleic acid sequence from SEQ ID No. 1-16. Such an increasing or decreasing effect may not be observed as effectively without the step of incubating the cell in the presence of astrocyte conditioned medium, although it is to be understood that the use of the astrocyte medium is optional. According to this embodiment, when the expression level of said nucleic acid sequence is upregulated in a cell incubated in the presence of a compound and in the presence of ACM (which may be present before, during and/or after the incubation in the presence of said compound) as compared to a cell incubated without the presence of a compound, such a compound is capable of increasing the BBB function. When the expression level of said nucleic acid sequence is downregulated in a cell incubated in the presence of a compound and in the presence of ACM (which may be present before, during and/or after the incubation in the presence of said compound) as compared to a cell incubated without the presence of a compound, such a compound is capable of decreasing the BBB function. In general, compounds that result in either an up- or downregulation of (a) nucleic acid(s) sequence(s) selected from the group of SEQ ID No. 1-16 of at least 2-fold may be considered as compounds that are identified to have the potential to modulate the BBB function, i.e. increase or decrease the BBB function.
It is preferred to use brain endothelial cells in the above assays, as the BBB largely comprises brain endothelial cells, and thus, these cells are preferred. Therefore, in a further embodiment, the cell in the assays as described is a brain endothelial cell. The brain endothelial cell can be a primary cell, but a cell line is preferred. Human primary brain endothelial cells may be obtained from subjects from biopsy material, and thus supply might be limited and restricted by regulations as consent is required, furthermore, primary cells have a limited life span. Therefore, it is preferred to use an immortalized human brain endothelial cell line, such as for instance HCMEC/D3.
In the above assays, the expression level of a nucleic acid sequence from SEQ ID No 1-16 is determined. However, it is to be noted that it is also possible to determine the expression level of several nucleic acid sequences from SEQ ID No 1-16, for example simultaneously. In one embodiment, the expression level of the nucleic acid sequences in the above assays can be determined with a microarray (Li and Ruan, Anal Bioanal Chem. 2009 Jun;394(4): 1117-24). A microarray is a multiplex technology used in molecular biology and in medicine which consists of an arrayed series of thousands of microscopic spots of oligonucleotides, each containing picomoles of a specific oligonucleotide sequence. This sequence is complementary to a sequence from SEQ ID No 1-16 that are used as probes to hybridize to a target nucleic acid sequence under suitable, preferably high-stringency conditions. The target nucleic acid sequences can be labelled for example with a fluorescent label, and probe-target hybridization can thus be detected and quantified by measuring the fluorescence intensity. For instance, in example 2 it is described in close detail how a microarray may be performed. In an alternative embodiment, the expression level is determined with a PCR reaction, preferably a quantitative PCR reaction. In example 6 it is described in close detail how such a PCR can be performed; in addition and based on the sequences disclosed herein any skilled person understands how to perform such PCR, preferably quantitative PCR reaction (as described in detail in Mestdagh et al., Nucleic Acids Res. 2008 Dec;36(21) and Chen et al., Nucleic Acids Res. 2005 Nov 27;33(20)).
In another embodiment, the expression level of a nucleic acid sequence selected from the group consisting of SEQ ID No 1-16 may be determined with the use of a reporter construct. A reporter gene construct expresses a reporter gene, for instance Green Fluorescent Protein (GFP) or luciferase. The reporter gene construct thus expresses a mRNA transcript that encodes GFP. Within the 3' untranslated region of the GFP transcript, a complementary target sequence, or multiple complementary target sequences can be introduced, such that a nucleic acid sequence from the group consisting of SEQ D No. 1-16 can bind to the complementary target sequence (Brown BD, et al. Nat Med. 2006 May;12(5):585-91). The reporter gene construct can be introduced in a cell transiently, for instance via transfection, or may be stably introduced, for instance via recombination or transduction. As the nucleic acid sequence from the group consisting of SEQ ID No. 1-16 targets the reporter gene construct via complementary base paring and/or RNA interference, the level of expression of the reporter gene is controlled by level of expression of the said nucleic acid sequence, because binding and/or cleavage of the nucleic acid sequence to the reporter transcript reduces mRNA translation into protein. Thus, in case a GFP construct with a complementary target sequence, or multiple target sequences to a sequence selected from the group consisting of SEQ ID. No. 1-16 is used in the said assay, the GFP expression level is inversely proportional to the level of expression of a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID No. 1-16. Thus, a reduction in the GFP expression corresponds to an increase in the said nucleic acid sequence expression and thus an increase in the BBB function, while an increase in GFP expression corresponds to a decrease in the said nucleic acid sequence expression and thus a decrease in the BBB function. In a further embodiment, in an assay for screening for compounds that modulate the blood brain barrier function as disclosed above, the expression level of a nucleic acid sequence selected from the group of SEQ ID No. 1-16 is determined with a microarray, a PCR reaction, a quantitative PCR or a reporter gene construct.
EXAMPLES
Example 1
Astrocyte factors increase the blood brain barrier function
The effect of soluble astrocyte factors on the blood brain barrier function was evaluated in vitro with brain endothelial cells. For this, 200.000 HCMEC/D3 cells, a brain endothelial cell line, were seeded at confluency onto each well of an 8W10+ ECIS array coated with collagen cells. Cells were incubated in in EGM-2 medium (Clonetics) containing 2.5% foetal calf serum, or in a 1 : 1 mixture of astrocyte conditioned medium (cell free medium from human astrocytes cultured for 7 days in standard DMEM (Gibco) medium) and EGM-2 medium containing 2.5% foetal calf serum. The ECISTM Model 1600R (Applied BioPhysics, Troy, NY, USA) was used to measure the transendothelial electrical resistance (TEER) for 60-80 hours using ECIS according to the manufacturer's instructions and impedance was measured at 6000 Hz in real time. After about 70 hours post-seeding, the TEER values of a control well, in which the endothelial brain cells were incubated without astrocyte conditioned medium (ACM) was about 1250 ohm. The cells incubated with ACM had a value of about 1700 ohm. Thus, the endothelial brain cells incubated in ACM had an increased TEER value, representing an increased BBB function. It is concluded that ACM improves BBB function. Example 2
Astrocyte conditioned medium regulates miRNA expression
miRNAs were studied for their involvement in the blood-brain barrier function. Large-scale expression analysis was performed of miRNAs in HCMEC/D3s either or not treated with astrocyte conditioned medium. 13 - 18 μg total RNA was isolated using Trizol from 1 x 106 brain endothelial cells which were cultured in 50% astrocytes-conditioned medium / 50% complete EGM2 for 48 h. Before mRNA and miRNA profiling the RNA samples underwent quality control by Agilent Bioanalyzer Picochip analysis (RIN>9).
For miRNA expression analyses (Exiqon, Denmark), 1 μg total RNA from sample and reference was labeled with Hy3™ and Hy5™ fluorescent label, respectively, using the miRCURY™ LNA Array power labeling kit (Exiqon, Denmark) following the procedure described by the manufacturer. The Hy3™-labeled samples and a Hy5™-labeled reference RNA sample were mixed pair-wise and hybridized to the miRCURY™ LNA array version 11.0 (Exiqon, Denmark), which contains capture probes targeting all miRNAs for human, mouse or rat registered in the miRBASE version 12.0 at the Sanger Institute (Sanger Institute details: http://www.mirbase.org/ ). The hybridization was performed according to the miRCURY™ LNA array manual using a Tecan HS4800 hybridization station (Tecan, Austria). After hybridization the microarray slides were scanned and stored in an ozone free environment (ozone level below 2.0 ppb) in order to prevent potential bleaching of the fluorescent dyes. The miRCURY™ LNA array microarray slides were scanned using the Agilent G2565BA Microarray Scanner System (Agilent Technologies, Inc., USA) and the image analysis was carried out using the ImaGene 8.0 software (BioDiscovery, Inc., USA). Generation and statistical analysis of miRNA expression profiles were performed similarly using R/Bioconductor. Quantile normalization of intensities of individual spots was followed by reference colour adjustment and ratios of probes representing the same miRNA were averaged. Differential expression of ACM-treated versus control samples was determined by Baysian statistics t-test (Baldi and Long 2001) and multiple testing adjustment using Benjamini and Hochberg's method (Benjamini and Hochberg 1995). Analyses of array hybridizations revealed the putative miRNA expression of in HCMEC/D3s and confirmed the expression of various miRNAs previously described to be expressed in endothelial cells (Kuehbacher et al. 2007;Poliseno et al. 2006;Suarez et al. 2007;Tuccoli et al. 2006;Wurdinger et al. 2008). Exposure of HCMEC/D3s to astrocyte conditioned medium changed the expression level of miRNAs (Table I; SEQ ID No. 1-16) at least 2-fold as compared to HCMEC/D3s not exposed to astrocyte conditioned medium. It was concluded that an increase of the nucleic acid sequences of SEQ ID No. 1-16 increases the BBB function. Furthermore, it was also concluded that these (putative) miRNA sequences can be used as markers for BBB function, and thus are suitable for use in assays in which compounds are screened for that modulate, i.e. increase or decrease, the BBB function. Finally, it was also concluded that these nucleic acid sequences, or nucleic acids comprising nucleic acid sequences with some degree of identity therewith, are suitable for increasing the BBB function, and that nucleic acids with some degree of complementarity with the nucleic acid sequences from SEQ ID No. 1-16 (See Table 1) would be suitable for downregulating the BBB function. Table 1
Figure imgf000028_0001
Should any discrepancy exist between the information depicted in Table 1 and the accompanying sequence listing, Table 1 should be considered as the true original with the information as intended.
Example 3
Blocking the RNAi pathway via knock-down of DICER results in a decrease in BBB function To confirm that the nucleic acids according to the invention, in particular miRNAs having a sequence according to any of seq ID no 1-16 improve BBB function, the effect of Dicer knock-down with an siRNA against Dicer was studied. Knocking down Dicer should result in blocking the processing of pri-miRNAs and hence prevent incorporation of endogenous miRNAs into the RISC complex. Endothelial brain cells were transfected with an siRNA against DICER (sc-40489, Santa Cruz) or a silencing control siRNA (also obtained from Santa Cruz) and trans fections were performed with DharmaFECT-4 according to the manufacturer's instructions (Dharmacon). 48 hours post-transfection, the cells were seeded in an 8W10+ ECIS array as described above and TEER was measure as described above. Results show knocking down the DICER enzyme reduced the resistance of the in endothelial brain cells, confirming that indeed the RNA interference pathway and/or miRNA biogenesis pathway is involved and required for BBB function. These data therefore confirm the concept that the nucleic acid sequences from SEQ ID No. 1-16 are involved in regulating the BBB function, in particular in increasing the BBB function.
Example 4
Lentiviral delivery of microRNAs increases BBB function
MiRNAs were cloned from human genomic DNA and ligated between the Smal and EcoRI restriction sites of pRRL-cPPT-CMV-X2-PRE-SIN-IRES-eGFP vector (kindly provided by Dr. J. Seppen, Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands). Recombinant lentiviruses were produced by co-transfecting subconfluent HEK 293T cells with the specific expression plasmids and packaging plasmids (pMDLg/pRRE, pRSV-Rev and pMD2G) using calcium phosphate as a transfection reagent. HEK 293T cells were cultured in DMEM supplemented with 10% FCS, 1% penicillin/streptomycin, in a 37°C incubator with 5% C02. Infectious lentiviruses were collected 48 h after transfection. The supernatant was centrifuged to remove cell debris and stored at -80°C. For barrier resistance analyses hCMEC/D3 human brain endothelial cells were transduced with the lentiviruses containing the microRNAs. The ECIS™ Model 1600R (Applied BioPhysics, Troy, NY, USA) was used to measure the transendothehal electrical resistance (TEER) for 60-80 hours using ECIS according to the manufacturer's instructions and impedance was measured at 6000 Hz in real time. The results of real-time TEER analysis show that increasing the amount of a microRNA selected from the group consisting of SEQ ID No. 1-16 enhanced the resistance barrier properties. Thus, this experiment confirms that nucleic acids from SEQ ID No. 1-16 are suitable for increasing the BBB function, in particular in multiple sclerosis.
Example 5
Transendothelial migration of primary monocytes Another functional aspect of the BBB is the prevention of monocyte transmigration. Human blood monocytes were isolated from buffy coats of healthy donors by Ficoll gradient and CD14-positive beads (Elkord et al. 2005, Immunology 114, 204-212). Primary monocyte transmigration through a monolayer of hCMEC/D3 was assayed as described previously (Reijerkerk et al. 2008 J. Immunol. 181, 3567-3574). Briefly, hCMEC/D3 cells were seeded onto 96 well plates and infected with lentivirus expressing miR-30a,b,c,d or e for two days. hCMEC/D3 cells were immune activated with TNF alpha (5 ng/ml) for 24 h. Subsequently, 7.5 x 104 monocytes were added to 96-well plates containing hCMEC/D3 monolayers. Monocytes were allowed to settle and migrate over a 4-h period. To monitor monocyte migration, cocultures were placed in an inverted phase-contrast microscope (Nikon Eclipse TE300) housed in a temperature-controlled (37°C), 5% C02 gassed chamber. A field (220 x 220 μπι) was randomly selected and recorded for 10 min at 50 times by using a digital video camera using Cell F imaging software (Olympus, Heidelberg, Germany). Monocyte diapedesis was assessed by enumerating the number of monocytes within the field that had either adhered or migrated through the monolayer. Transmigrated monocytes could be readily distinguished from those remaining on the cell surface by their highly refractive morphology. The results confirm the data presented above.
Example 6
miRNA in capillaries from MS tissue.
Tissues were obtained from The Netherlands Brain Bank (NBB), Netherlands Institute for Neuroscience, Amsterdam. All material has been collected from donors from whom a written informed consent for brain autopsy and the use of the material and clinical information for research purposes had been obtained by the NBB. Brain capillaries were isolated from periventricular MS lesions from post-mortem MS patients or non-neurological controls. After resection, the tissues were homogenized using pestles and capillaries were separated from myelin and single cells using DEAE-dextran gradient (15%; Sigma Life Science) centrifuged 25 minutes 2500 rpm without brake. Isolated capillaries were washed on a 40 μιη meshed filter (BD Falcon) to remove red blood cells. Finally, RNA was extracted with Trizol and stored in -80°C. qPCR analysis of miRs
For cDNA synthesis, the Universal cDNA synthesis kit (cat # 203300) from Exiqon was used. Extracted RNA was diluted to 5 μg/μl using RNAse free water. For each reaction 2 μΐ diluted RNA was added to a mixture of 2 μΐ 5x buffer, 4.5 μΐ RNAse free water, 1 μΐ enzyme mix and 0.5 μΐ RNA spike-in. Total volume was 10 ul per reaction. The samples were incubated at 42oC for one hour followed by heat-inactivation at 95oC for 5 minutes. Samples were cooled to 4oC and thereafter stored at -20oC. The qPCR was performed using the Exiqon "pick and mix" plates (cat# 203801). Three independent samples per plate were tested. The cDNA was diluted 1 : 100 in RNAse free water. Per well, 5 μΐ of sample and 5 μΐ of 2x SYBRgreen mastermix were added. Plates were sealed and centrifuged. qPCR was performed on an ABI VILA 7 machine which performed 40 cycles of 95 °C 10 sec, 60 °C 1 minute followed by a melt curve.
We assessed the expression level of a set of miRNAs in freshly isolated brain capillaries from MS lesion areas and non-neurological controls. Isolated brain capillaries were predominantly comprised of brain endothelial cells as indicated by qPCR analysis of ZO-1, claudin-5, P- glycoprotein and VE-cadherin expression at contained minor amounts of astrocytes (glial fibrillary acidic protein a and β) and pericytes (platelet-derived growth factor receptor-β). Of these, 16 barrier-enhancing micro RN As were diminished in brain capillaries of MS patients (see Figure 1). We conclude that therapeutic application of miRNAs, could re-establish normal functioning of the brain vasculature in endothelial cell based neurological diseases, in particular MS.

Claims

Use of a compound, preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis.
A compound, preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%, preferably at least 75%, more preferably 80%,even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for use in increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis.
Use of a compound, preferably a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 - 16, or a nucleic acid sequence which has at least 70%», preferably at least 75%», more preferably 80%,even more preferably 85%», even more preferably 90%», even more preferably 95%, most preferably 100%» sequence identity with a nucleic acid sequence selected from the group consisting of SEQ ID: 1 - 16, for the manufacture of a medicament for increasing blood-brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis.
Use according to claim 1 or 3, or a compound according to claim 2, wherein the compound is is or comprises miR-15a, miR-18b, miR-26a, miR-27a, miR-27b, miR- 30a, miR-30b, miR-30c, miR-30d, miR-30e, miR-31, miR-106b, miR-125a-5p, miR- 155, miR-195, miR-487b.
5. Use of compound, preferably a nucleic acid, comprising a nucleic acid sequence of which at least 8 adjacent nucleotides are at least 70%», preferably 75%», more preferably 80%, even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100%) complementary to 8 adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16, for decreasing blood -brain barrier function.
6. A compound, preferably a nucleic acid, comprising a nucleic acid sequence of which at least 8 adjacent nucleotides are at least 70%, preferably 75%, more preferably 80%, even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% complementary to 8 adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16, for use in decreasing blood- brain barrier function.
7. Use of compound, preferably a nucleic acid, comprising a nucleic acid sequence of which at least 8 adjacent nucleotides are at least 70%, preferably 75%, more preferably 80%, even more preferably 85%, even more preferably 90%, even more preferably 95%, most preferably 100% complementary to 8 adjacent nucleotides of a nucleic acid sequence selected from the group of SEQ ID No 1-16, for the manufacture of a medicament for decreasing blood-brain barrier function.
8. A compound, preferably a nucleic acid, comprising a nucleic acid sequence which has at least 70%», preferably 75%», more preferably 80%,even more preferably 85%, even more preferably 90%», even more preferably 95%», most preferably 99%» sequence identity with a nucleic acid sequence selected f om the group consisting of SEQ ID No 1-16, with the provision that the compound is not mi -1 a, miR-18b, miR-26a, miR- 27a, miR-27b, miR-30a, miR-30b, miR-30c, miR-30d, miR-30e, miR-31, miR-106b, miR-125a-5p, miR-155, miR-195, or miR-487b.
9. An assay for screening for compounds that modulate blood brain barrier function, preferably that increases blood brain barrier function in patients diagnosed with or suffering from Multiple Sclerosis comprising the steps of:
incubating a cell in the presence of a compound to be tested,
determining the expression level of a nucleic acid sequence selected from the group of SEQ ID No 1-16, wherein the expression level of said nucleic acid sequence is compared with the expression level of said nucleic acid sequence as determined in a cell incubated without the presence of said compound to be tested.
10. Assay according to claim 9 wherein in the step of incubating a cell in the presence of a compound to be tested, astrocyte conditioned medium is present and/or wherein the assay further comprises the step of:
incubating the cell in the presence of astrocyte conditioned medium, before and/or or after the step of incubating a cell in the presence of a compound to be tested.
11. Assay according to claim 9 - 10, wherein the cell is a brain endothelial cell, preferably a brain endothelial cell line, preferably the brain endothelial cell line HCMEC/D3.
12. Use of miR-15a, miR-18b, miR-26a, miR-27a, miR-27b, miR-30a, miR-30b, miR-30c, miR-30d, miR-30e, miR-31, miR-106b, miR-125a-5p, miR-155, miR-195,or miR-487, the use preferably comprising determining expression level of at least one of miR-15a, miR-18b, miR-26a, miR-27a, miR-27b, miR-30a, miR-30b, miR-30c, miR-30d, miR- 30e, miR-31, miR-106b, miR-125a-5p, miR-155, miR-195, or miR-487b, in the diagnosis of blood brain barrier function, preferably in the diagnosis of blood brain barrier function in patients suffering from or diagnosed with Multiple Sclerosis.
PCT/NL2012/050793 2011-11-10 2012-11-09 miRNA IN BLOOD BRAIN BARRIER OF MS PATIENTS WO2013070079A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NL2007767 2011-11-10
NL2007767 2011-11-10

Publications (3)

Publication Number Publication Date
WO2013070079A2 true WO2013070079A2 (en) 2013-05-16
WO2013070079A3 WO2013070079A3 (en) 2013-07-11
WO2013070079A8 WO2013070079A8 (en) 2013-10-24

Family

ID=47278967

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2012/050793 WO2013070079A2 (en) 2011-11-10 2012-11-09 miRNA IN BLOOD BRAIN BARRIER OF MS PATIENTS

Country Status (1)

Country Link
WO (1) WO2013070079A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018228875A1 (en) * 2017-06-15 2018-12-20 Nestec S.A. Micro-rna biomarkers of blood-brain barrier dysfunction
JP2020534349A (en) * 2017-08-18 2020-11-26 クラミス シーオー., エルティーディー.Curamys Co., Ltd. Uses of miR-18b for the prevention, treatment or diagnosis of muscular and neuromuscular disorders

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008147528A1 (en) 2007-05-24 2008-12-04 The Board Of Trustees Of The Leland Stanford Junior University Methods of modulating angiogenesis
WO2009047362A2 (en) 2007-10-12 2009-04-16 The Provost, Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth, Near Dublin Method for opening tight junctions

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2152865B1 (en) * 2007-05-11 2015-04-15 Thomas Jefferson University Methods of treatment and prevention of neurodegenerative diseases and disorders
AU2008261644B2 (en) * 2007-06-12 2014-04-03 Case Western Reserve University Targeted cell death
EP2611918A1 (en) * 2010-05-28 2013-07-10 Vereniging Voor Christelijk Hoger Onderwijs, Weten Mirnas involved in the blood brain barrier function

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008147528A1 (en) 2007-05-24 2008-12-04 The Board Of Trustees Of The Leland Stanford Junior University Methods of modulating angiogenesis
WO2009047362A2 (en) 2007-10-12 2009-04-16 The Provost, Fellows And Scholars Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth, Near Dublin Method for opening tight junctions

Non-Patent Citations (40)

* Cited by examiner, † Cited by third party
Title
AAGAARD ET AL., GENE THER., vol. 15, no. 23, December 2008 (2008-12-01), pages 1536 - 49
ABBOTT ET AL., NAT REV NEUROSC, vol. 7, 2006, pages 41 - 53
ABBOTT ET AL., NEUROBIOL DIS., vol. 37, no. 1, January 2010 (2010-01-01), pages 13 - 25
ALBERT L. LEHNINGER: "Principles of Biochemistry", 1982, pages: 793 - 800
BARTEL D. P., CELL, vol. 116, 2004, pages 281 - 297
BARTEL, CELL, vol. 116, 2004, pages 281 - 297
BEGLEY; BRIGHTMAN, PROG DRUG RES., vol. 61, 2003, pages 39 - 78
BORGHOUTS ET AL., COMB CHEM HIGH THROUGHPUT SCREEN, vol. 11, no. 2, February 2008 (2008-02-01), pages 135 - 45
BROWN BD ET AL., NAT MED., vol. 12, no. 5, May 2006 (2006-05-01), pages 585 - 91
BRUMMELKAMP ET AL., RNA, vol. 296, no. 5567, 19 April 2002 (2002-04-19), pages 550 - 3
CARTHEW; SONTHEIMER, CELL, vol. 136, 2009, pages 642 - 655
CARVEY ET AL., NEUROCHEM., vol. 111, no. 2, October 2009 (2009-10-01), pages 291 - 314
CERCHIA ET AL., METHODS MOL BIOL., vol. 535, 2009, pages 59 - 78
CHAN ET AL., CLIN EXP PHARMACOL PHYSIOL., vol. 33, no. 5-6, May 2006 (2006-05-01), pages 533 - 40
CHEN ET AL., NUCLEIC ACIDS RES., vol. 33, no. 20, 27 November 2005 (2005-11-27)
CHUA ET AL., CURR OPIN MOL THER., vol. 11, no. 2, April 2009 (2009-04-01), pages 189 - 99
DAFFERTSHOFER; FATAR, EUR J ULTRASOUND., vol. 16, no. 1-2, November 2002 (2002-11-01), pages 121 - 30
DASSIE JP ET AL., NAT BIOTECHNOL., vol. 27, no. 9, September 2009 (2009-09-01), pages 839 - 49
DELI ET AL., CELL MOL NEUROBIOL., vol. 25, no. 1, February 2005 (2005-02-01), pages 59 - 127
DOOLITTLE ET AL., J NEUROSCI NURS., vol. 30, no. 2, April 1998 (1998-04-01), pages 81 - 90
ELBASHIR ET AL., NATURE, vol. 411, no. 6836, 24 May 2001 (2001-05-24), pages 494 - 8
ELKORD ET AL., IMMUNOLOGY, vol. 114, 2005, pages 204 - 212
KIM, NATURE REV. MOL. CELL. BIOL., vol. 6, 2005, pages 376 - 385
KRÜTZFELDT ET AL., NATURE, vol. 438, no. 7068, 1 December 2005 (2005-12-01), pages 685 - 9
LI; RUAN, ANAL BIOANAL CHEM., vol. 394, no. 4, June 2009 (2009-06-01), pages 1117 - 24
LIU ET AL., NUCLEIC ACIDS RES., vol. 36, no. 9, May 2008 (2008-05-01), pages 2811 - 24
LOSCHER; POTSCHKA, NEURORX, vol. 2, no. 1, January 2005 (2005-01-01), pages 86 - 98
MCNAMARA JO 2ND ET AL., NAT BIOTECHNOL., vol. 24, no. 8, August 2006 (2006-08-01), pages 1005 - 15
MESTDAGH ET AL., NUCLEIC ACIDS RES., vol. 36, no. 21, December 2008 (2008-12-01)
MINAGAR ET AL., MULTIPLE SCLEROSIS, vol. 9, 2003, pages 540
MORRIS, BIOTECHNIQUES, April 2006 (2006-04-01), pages 7 - 13
PACHTER ET AL., J NEUROPATHOL EXP NEUROL., vol. 62, no. 6, June 2003 (2003-06-01), pages 593 - 604
POZO-RODRIGUEZ ET AL., EUR J PHARM BIOPHARM., vol. 71, no. 2, February 2009 (2009-02-01), pages 181 - 9
REIJERKERK ET AL., J. IMMUNOL., vol. 181, 2008, pages 3567 - 3574
REINISALO ET AL., J CONTROL RELEASE, vol. 110, no. 2, 10 January 2006 (2006-01-10), pages 437 - 43
SOUTSCHEK ET AL., J NATURE, vol. 432, no. 7014, 11 November 2004 (2004-11-11), pages 173 - 8
SUMMERTON AND WELLER ANTISENSE NUCLEIC ACID DRUG DEV., vol. 7, no. 3, June 1997 (1997-06-01), pages 187 - 95
WAHL ET AL., ACTA PHYSIOL HUNG., vol. 86, no. 2, 1999, pages 155 - 60
WEKSLER ET AL., FASEB JOURNAL, vol. 19, 2005, pages 1872 - 1874
WINTER ET AL., NATURE CELL BIOL, vol. 11, no. 3, 2009, pages 228 - 234

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018228875A1 (en) * 2017-06-15 2018-12-20 Nestec S.A. Micro-rna biomarkers of blood-brain barrier dysfunction
JP2020534349A (en) * 2017-08-18 2020-11-26 クラミス シーオー., エルティーディー.Curamys Co., Ltd. Uses of miR-18b for the prevention, treatment or diagnosis of muscular and neuromuscular disorders
US11286484B2 (en) 2017-08-18 2022-03-29 Curamys Co., Ltd. Use of miR-18b for prevention, treatment, or diagnosis of muscle disease and neuromuscular disease
US11891604B2 (en) 2017-08-18 2024-02-06 Curamys Co., Ltd. Use of miR-18b for prevention, treatment, or diagnosis of muscle disease and neuromuscular disease

Also Published As

Publication number Publication date
WO2013070079A8 (en) 2013-10-24
WO2013070079A3 (en) 2013-07-11

Similar Documents

Publication Publication Date Title
US9297007B2 (en) miRNAs involved in the blood brain barrier function
Maciotta et al. The involvement of microRNAs in neurodegenerative diseases
Ghosh et al. Molecular mechanism of diabetic cardiomyopathy and modulation of microRNA function by synthetic oligonucleotides
Meloni et al. Local inhibition of microRNA-24 improves reparative angiogenesis and left ventricle remodeling and function in mice with myocardial infarction
US9862949B2 (en) Method for the inhibition of angiogenesis
Christensen et al. Recombinant adeno-associated virus-mediated microRNA delivery into the postnatal mouse brain reveals a role for miR-134 in dendritogenesis in vivo
US10519443B2 (en) Microrna inhibitor system and methods of use thereof
US20100008981A1 (en) Methods and sequences to suppress primate huntington gene expression
US20130331436A1 (en) Micrornas as new therapeutic targets for the prevention and/or treatment of retinopathy
Ruberti et al. Targeting microRNAs in neurons: tools and perspectives
WO2012105826A1 (en) Use of micrornas in diagnosis and therapy of aging
WO2013070079A2 (en) miRNA IN BLOOD BRAIN BARRIER OF MS PATIENTS
EP3704250A1 (en) Mirna molecule, equivalent, antagomir, or source thereof for treating and/or diagnosing a condition and/or a disease associated with neuronal deficiency or for neuronal (re)generation
US20150275210A1 (en) Treatment of metastatic breast cancer
WO2015044890A2 (en) Tools and methods using mirna 182, 96 and/or 183 for treating pathologies
EP3344768B1 (en) Micrornas for the treatment of heart diseases
WO2016081542A1 (en) Use of microrna 375 in augmenting stem cell based and endogenous ischemic tissue repair
Pinnarò Role of noncoding RNAs in muscle differentiation

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: 12795102

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12795102

Country of ref document: EP

Kind code of ref document: A2