CN118924898A - Neuron protection and function recovery method after focal ischemic cerebral apoplexy - Google Patents
Neuron protection and function recovery method after focal ischemic cerebral apoplexy Download PDFInfo
- Publication number
- CN118924898A CN118924898A CN202310536200.8A CN202310536200A CN118924898A CN 118924898 A CN118924898 A CN 118924898A CN 202310536200 A CN202310536200 A CN 202310536200A CN 118924898 A CN118924898 A CN 118924898A
- Authority
- CN
- China
- Prior art keywords
- mcao
- mir
- cerebral apoplexy
- agomir
- group
- Prior art date
- Legal status (The legal status 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 status listed.)
- Pending
Links
- 238000000034 method Methods 0.000 title claims abstract description 34
- 210000002569 neuron Anatomy 0.000 title claims abstract description 20
- 238000011084 recovery Methods 0.000 title claims abstract description 17
- 230000004224 protection Effects 0.000 title claims abstract description 12
- 230000000302 ischemic effect Effects 0.000 title claims description 34
- 208000006011 Stroke Diseases 0.000 title claims description 18
- 230000002490 cerebral effect Effects 0.000 title claims description 17
- 206010008190 Cerebrovascular accident Diseases 0.000 title claims description 13
- 108091053935 miR-212 stem-loop Proteins 0.000 claims abstract description 38
- 238000002474 experimental method Methods 0.000 claims abstract description 21
- 238000012795 verification Methods 0.000 claims abstract description 10
- 101001067187 Homo sapiens Plexin-A2 Proteins 0.000 claims abstract description 8
- 102100034381 Plexin-A2 Human genes 0.000 claims abstract description 7
- 208000028389 Nerve injury Diseases 0.000 claims abstract description 5
- 230000008764 nerve damage Effects 0.000 claims abstract description 5
- 230000001105 regulatory effect Effects 0.000 claims abstract description 5
- 210000005036 nerve Anatomy 0.000 claims abstract description 3
- 210000001808 exosome Anatomy 0.000 claims description 36
- 210000004027 cell Anatomy 0.000 claims description 31
- 230000002025 microglial effect Effects 0.000 claims description 25
- 108090000623 proteins and genes Proteins 0.000 claims description 19
- 230000006870 function Effects 0.000 claims description 17
- 238000001262 western blot Methods 0.000 claims description 16
- 108090000695 Cytokines Proteins 0.000 claims description 13
- 102000004127 Cytokines Human genes 0.000 claims description 13
- 238000011529 RT qPCR Methods 0.000 claims description 13
- 210000005013 brain tissue Anatomy 0.000 claims description 12
- 238000012360 testing method Methods 0.000 claims description 12
- 230000001054 cortical effect Effects 0.000 claims description 11
- 241001465754 Metazoa Species 0.000 claims description 10
- 230000003110 anti-inflammatory effect Effects 0.000 claims description 8
- 230000004112 neuroprotection Effects 0.000 claims description 8
- 230000000770 proinflammatory effect Effects 0.000 claims description 8
- 208000028867 ischemia Diseases 0.000 claims description 6
- 238000010171 animal model Methods 0.000 claims description 5
- 230000008859 change Effects 0.000 claims description 4
- 238000004445 quantitative analysis Methods 0.000 claims description 3
- 201000010875 transient cerebral ischemia Diseases 0.000 claims description 3
- 230000003542 behavioural effect Effects 0.000 claims description 2
- 230000037041 intracellular level Effects 0.000 claims 1
- 208000032382 Ischaemic stroke Diseases 0.000 abstract description 9
- 208000027418 Wounds and injury Diseases 0.000 abstract description 4
- 230000006378 damage Effects 0.000 abstract description 4
- 208000014674 injury Diseases 0.000 abstract description 4
- 230000014509 gene expression Effects 0.000 description 39
- 241000700159 Rattus Species 0.000 description 36
- 108091070501 miRNA Proteins 0.000 description 24
- 210000004556 brain Anatomy 0.000 description 23
- 239000002679 microRNA Substances 0.000 description 23
- 238000001356 surgical procedure Methods 0.000 description 21
- 101000669921 Homo sapiens Rho-associated protein kinase 2 Proteins 0.000 description 17
- 102100039314 Rho-associated protein kinase 2 Human genes 0.000 description 17
- 238000003125 immunofluorescent labeling Methods 0.000 description 17
- 230000002829 reductive effect Effects 0.000 description 17
- 210000001787 dendrite Anatomy 0.000 description 15
- 210000003520 dendritic spine Anatomy 0.000 description 15
- 210000000337 motor cortex Anatomy 0.000 description 12
- 102000009099 rhoA GTP Binding Protein Human genes 0.000 description 12
- 108010087917 rhoA GTP Binding Protein Proteins 0.000 description 12
- 238000010186 staining Methods 0.000 description 12
- 238000004458 analytical method Methods 0.000 description 11
- 108020004999 messenger RNA Proteins 0.000 description 11
- 230000003376 axonal effect Effects 0.000 description 10
- 206010008118 cerebral infarction Diseases 0.000 description 10
- 230000001965 increasing effect Effects 0.000 description 9
- 230000000324 neuroprotective effect Effects 0.000 description 9
- 230000007850 degeneration Effects 0.000 description 8
- 210000001519 tissue Anatomy 0.000 description 8
- 102000010410 Nogo Proteins Human genes 0.000 description 7
- 108010077641 Nogo Proteins Proteins 0.000 description 7
- 230000009223 neuronal apoptosis Effects 0.000 description 7
- 238000012163 sequencing technique Methods 0.000 description 7
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 7
- 239000005089 Luciferase Substances 0.000 description 6
- 230000000694 effects Effects 0.000 description 6
- 239000012528 membrane Substances 0.000 description 6
- 102000004169 proteins and genes Human genes 0.000 description 6
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 5
- 201000006474 Brain Ischemia Diseases 0.000 description 5
- 102100025222 CD63 antigen Human genes 0.000 description 5
- 101100356682 Caenorhabditis elegans rho-1 gene Proteins 0.000 description 5
- 206010008120 Cerebral ischaemia Diseases 0.000 description 5
- 101000934368 Homo sapiens CD63 antigen Proteins 0.000 description 5
- 102100022338 Integrin alpha-M Human genes 0.000 description 5
- 108060001084 Luciferase Proteins 0.000 description 5
- 239000002033 PVDF binder Substances 0.000 description 5
- 101150111584 RHOA gene Proteins 0.000 description 5
- 208000029028 brain injury Diseases 0.000 description 5
- 210000000269 carotid artery external Anatomy 0.000 description 5
- 210000004004 carotid artery internal Anatomy 0.000 description 5
- 238000001514 detection method Methods 0.000 description 5
- 239000012153 distilled water Substances 0.000 description 5
- 230000000763 evoking effect Effects 0.000 description 5
- 238000003384 imaging method Methods 0.000 description 5
- 230000007971 neurological deficit Effects 0.000 description 5
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 5
- 230000000946 synaptic effect Effects 0.000 description 5
- 230000003956 synaptic plasticity Effects 0.000 description 5
- 238000005406 washing Methods 0.000 description 5
- 101150053137 AIF1 gene Proteins 0.000 description 4
- 102000007469 Actins Human genes 0.000 description 4
- 108010085238 Actins Proteins 0.000 description 4
- 102100037904 CD9 antigen Human genes 0.000 description 4
- 101000738354 Homo sapiens CD9 antigen Proteins 0.000 description 4
- 101001046686 Homo sapiens Integrin alpha-M Proteins 0.000 description 4
- 206010061216 Infarction Diseases 0.000 description 4
- 238000010826 Nissl staining Methods 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 230000012010 growth Effects 0.000 description 4
- 210000000274 microglia Anatomy 0.000 description 4
- 230000007659 motor function Effects 0.000 description 4
- 239000000243 solution Substances 0.000 description 4
- 239000006228 supernatant Substances 0.000 description 4
- 238000004627 transmission electron microscopy Methods 0.000 description 4
- 102100027221 CD81 antigen Human genes 0.000 description 3
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 101000914479 Homo sapiens CD81 antigen Proteins 0.000 description 3
- 101001092197 Homo sapiens RNA binding protein fox-1 homolog 3 Proteins 0.000 description 3
- 229930040373 Paraformaldehyde Natural products 0.000 description 3
- 102100035530 RNA binding protein fox-1 homolog 3 Human genes 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 108090000704 Tubulin Proteins 0.000 description 3
- 102000004243 Tubulin Human genes 0.000 description 3
- 230000005540 biological transmission Effects 0.000 description 3
- 230000017531 blood circulation Effects 0.000 description 3
- 229910002091 carbon monoxide Inorganic materials 0.000 description 3
- 210000001168 carotid artery common Anatomy 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 229910052802 copper Inorganic materials 0.000 description 3
- 239000010949 copper Substances 0.000 description 3
- 238000012937 correction Methods 0.000 description 3
- 210000003194 forelimb Anatomy 0.000 description 3
- 238000002599 functional magnetic resonance imaging Methods 0.000 description 3
- 230000005021 gait Effects 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 230000007574 infarction Effects 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 230000003961 neuronal insult Effects 0.000 description 3
- 229910052760 oxygen Inorganic materials 0.000 description 3
- 239000012188 paraffin wax Substances 0.000 description 3
- 229920002866 paraformaldehyde Polymers 0.000 description 3
- 239000008188 pellet Substances 0.000 description 3
- 230000001242 postsynaptic effect Effects 0.000 description 3
- 230000003518 presynaptic effect Effects 0.000 description 3
- 230000001737 promoting effect Effects 0.000 description 3
- 239000000523 sample Substances 0.000 description 3
- 230000001953 sensory effect Effects 0.000 description 3
- 238000007619 statistical method Methods 0.000 description 3
- 210000000225 synapse Anatomy 0.000 description 3
- 230000008685 targeting Effects 0.000 description 3
- 238000001890 transfection Methods 0.000 description 3
- PKDBCJSWQUOKDO-UHFFFAOYSA-M 2,3,5-triphenyltetrazolium chloride Chemical compound [Cl-].C1=CC=CC=C1C(N=[N+]1C=2C=CC=CC=2)=NN1C1=CC=CC=C1 PKDBCJSWQUOKDO-UHFFFAOYSA-M 0.000 description 2
- 108020005345 3' Untranslated Regions Proteins 0.000 description 2
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 108010009307 Forkhead Box Protein O3 Proteins 0.000 description 2
- 102000009562 Forkhead Box Protein O3 Human genes 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- SXRSQZLOMIGNAQ-UHFFFAOYSA-N Glutaraldehyde Chemical compound O=CCCCC=O SXRSQZLOMIGNAQ-UHFFFAOYSA-N 0.000 description 2
- 101000917826 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-a Proteins 0.000 description 2
- 101000917824 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-b Proteins 0.000 description 2
- 108090001005 Interleukin-6 Proteins 0.000 description 2
- PIWKPBJCKXDKJR-UHFFFAOYSA-N Isoflurane Chemical compound FC(F)OC(Cl)C(F)(F)F PIWKPBJCKXDKJR-UHFFFAOYSA-N 0.000 description 2
- 102100029204 Low affinity immunoglobulin gamma Fc region receptor II-a Human genes 0.000 description 2
- 102100023174 Methionine aminopeptidase 2 Human genes 0.000 description 2
- 108090000192 Methionyl aminopeptidases Proteins 0.000 description 2
- 108010011536 PTEN Phosphohydrolase Proteins 0.000 description 2
- 102000014160 PTEN Phosphohydrolase Human genes 0.000 description 2
- 239000013614 RNA sample Substances 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 108091036066 Three prime untranslated region Proteins 0.000 description 2
- 229920004890 Triton X-100 Polymers 0.000 description 2
- 239000013504 Triton X-100 Substances 0.000 description 2
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 210000001159 caudate nucleus Anatomy 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 230000030833 cell death Effects 0.000 description 2
- 208000026106 cerebrovascular disease Diseases 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- 230000009977 dual effect Effects 0.000 description 2
- 238000004043 dyeing Methods 0.000 description 2
- 238000001962 electrophoresis Methods 0.000 description 2
- 235000013305 food Nutrition 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 239000008273 gelatin Substances 0.000 description 2
- 229920000159 gelatin Polymers 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- 230000007946 glucose deprivation Effects 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 239000007928 intraperitoneal injection Substances 0.000 description 2
- 229960002725 isoflurane Drugs 0.000 description 2
- 210000003140 lateral ventricle Anatomy 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 108091047602 miR-126a stem-loop Proteins 0.000 description 2
- 108091059786 miR-128-1 stem-loop Proteins 0.000 description 2
- 108091084874 miR-128-2 stem-loop Proteins 0.000 description 2
- 108091078862 miR-128-3 stem-loop Proteins 0.000 description 2
- 108091073005 miR-128-4 stem-loop Proteins 0.000 description 2
- 108091028513 miR-1949 stem-loop Proteins 0.000 description 2
- 108091055059 miR-30c stem-loop Proteins 0.000 description 2
- 108091072917 miR-30c-1 stem-loop Proteins 0.000 description 2
- 108091066131 miR-30c-2 stem-loop Proteins 0.000 description 2
- 239000000203 mixture Substances 0.000 description 2
- 230000009456 molecular mechanism Effects 0.000 description 2
- 238000000465 moulding Methods 0.000 description 2
- 210000002487 multivesicular body Anatomy 0.000 description 2
- 239000002105 nanoparticle Substances 0.000 description 2
- 230000001537 neural effect Effects 0.000 description 2
- 230000009251 neurologic dysfunction Effects 0.000 description 2
- 208000015015 neurological dysfunction Diseases 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- 239000002245 particle Substances 0.000 description 2
- 229960001412 pentobarbital Drugs 0.000 description 2
- WEXRUCMBJFQVBZ-UHFFFAOYSA-N pentobarbital Chemical compound CCCC(C)C1(CC)C(=O)NC(=O)NC1=O WEXRUCMBJFQVBZ-UHFFFAOYSA-N 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 230000008929 regeneration Effects 0.000 description 2
- 238000011069 regeneration method Methods 0.000 description 2
- 230000000284 resting effect Effects 0.000 description 2
- 230000002441 reversible effect Effects 0.000 description 2
- 210000003625 skull Anatomy 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- LHASLBSEALHFGO-ASZAQJJISA-N 1-[(4s,5r)-4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-5-[[(2r,3r,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxymethyl]pyrimidine-2,4-dione Chemical compound C1[C@H](O)[C@@H](CO)OC1N1C(=O)NC(=O)C(CO[C@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)O)=C1 LHASLBSEALHFGO-ASZAQJJISA-N 0.000 description 1
- 229920000936 Agarose Polymers 0.000 description 1
- VHUUQVKOLVNVRT-UHFFFAOYSA-N Ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 108010017009 CD11b Antigen Proteins 0.000 description 1
- 102000003952 Caspase 3 Human genes 0.000 description 1
- 108090000397 Caspase 3 Proteins 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 108020004635 Complementary DNA Proteins 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 101100348612 Desulfovibrio vulgaris (strain ATCC 29579 / DSM 644 / NCIMB 8303 / VKM B-1760 / Hildenborough) ngr gene Proteins 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 101710088172 HTH-type transcriptional regulator RipA Proteins 0.000 description 1
- 208000016988 Hemorrhagic Stroke Diseases 0.000 description 1
- 101000998969 Homo sapiens Inositol-3-phosphate synthase 1 Proteins 0.000 description 1
- 101000979249 Homo sapiens Neuromodulin Proteins 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 238000012404 In vitro experiment Methods 0.000 description 1
- 102100036881 Inositol-3-phosphate synthase 1 Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 102000004889 Interleukin-6 Human genes 0.000 description 1
- 108700011259 MicroRNAs Proteins 0.000 description 1
- 206010028851 Necrosis Diseases 0.000 description 1
- 208000012902 Nervous system disease Diseases 0.000 description 1
- 208000036110 Neuroinflammatory disease Diseases 0.000 description 1
- 208000025966 Neurological disease Diseases 0.000 description 1
- 102100023206 Neuromodulin Human genes 0.000 description 1
- 102100029438 Nitric oxide synthase, inducible Human genes 0.000 description 1
- 101710089543 Nitric oxide synthase, inducible Proteins 0.000 description 1
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 1
- 206010030113 Oedema Diseases 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 239000004793 Polystyrene Substances 0.000 description 1
- 239000012083 RIPA buffer Substances 0.000 description 1
- 108700008625 Reporter Genes Proteins 0.000 description 1
- 102000004389 Ribonucleoproteins Human genes 0.000 description 1
- 108010081734 Ribonucleoproteins Proteins 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- 102000007562 Serum Albumin Human genes 0.000 description 1
- 108020004682 Single-Stranded DNA Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 239000006180 TBST buffer Substances 0.000 description 1
- 208000007536 Thrombosis Diseases 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 208000030886 Traumatic Brain injury Diseases 0.000 description 1
- 208000003443 Unconsciousness Diseases 0.000 description 1
- MHMCOSLCKPWGQG-UHFFFAOYSA-H [U+6].[O-]C(=O)C([O-])=O.[O-]C(=O)C([O-])=O.[O-]C(=O)C([O-])=O Chemical compound [U+6].[O-]C(=O)C([O-])=O.[O-]C(=O)C([O-])=O.[O-]C(=O)C([O-])=O MHMCOSLCKPWGQG-UHFFFAOYSA-H 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 229960000583 acetic acid Drugs 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 238000007605 air drying Methods 0.000 description 1
- 235000011114 ammonium hydroxide Nutrition 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 238000013528 artificial neural network Methods 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 210000001130 astrocyte Anatomy 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 238000009227 behaviour therapy Methods 0.000 description 1
- 230000002146 bilateral effect Effects 0.000 description 1
- 230000031018 biological processes and functions Effects 0.000 description 1
- 230000008499 blood brain barrier function Effects 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 210000001218 blood-brain barrier Anatomy 0.000 description 1
- 210000004958 brain cell Anatomy 0.000 description 1
- 230000006931 brain damage Effects 0.000 description 1
- 231100000874 brain damage Toxicity 0.000 description 1
- 230000003925 brain function Effects 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 239000002771 cell marker Substances 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 230000006835 compression Effects 0.000 description 1
- 238000007906 compression Methods 0.000 description 1
- 210000000806 cranial fontanelle Anatomy 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 238000001739 density measurement Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- HRLIOXLXPOHXTA-NSHDSACASA-N dexmedetomidine Chemical compound C1([C@@H](C)C=2C(=C(C)C=CC=2)C)=CN=C[N]1 HRLIOXLXPOHXTA-NSHDSACASA-N 0.000 description 1
- 229960004253 dexmedetomidine Drugs 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 238000010864 dual luciferase reporter gene assay Methods 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 230000002888 effect on disease Effects 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 238000002567 electromyography Methods 0.000 description 1
- 238000000635 electron micrograph Methods 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 229940011871 estrogen Drugs 0.000 description 1
- 239000000262 estrogen Substances 0.000 description 1
- 210000001723 extracellular space Anatomy 0.000 description 1
- 210000003414 extremity Anatomy 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 239000000835 fiber Substances 0.000 description 1
- 239000012362 glacial acetic acid Substances 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 210000004884 grey matter Anatomy 0.000 description 1
- 238000009499 grossing Methods 0.000 description 1
- 230000004886 head movement Effects 0.000 description 1
- 210000001320 hippocampus Anatomy 0.000 description 1
- 230000036732 histological change Effects 0.000 description 1
- 230000001146 hypoxic effect Effects 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 230000035992 intercellular communication Effects 0.000 description 1
- 230000010262 intracellular communication Effects 0.000 description 1
- 208000020658 intracerebral hemorrhage Diseases 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 230000003447 ipsilateral effect Effects 0.000 description 1
- 208000037906 ischaemic injury Diseases 0.000 description 1
- 230000007654 ischemic lesion Effects 0.000 description 1
- 230000002530 ischemic preconditioning effect Effects 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- NLYAJNPCOHFWQQ-UHFFFAOYSA-N kaolin Chemical compound O.O.O=[Al]O[Si](=O)O[Si](=O)O[Al]=O NLYAJNPCOHFWQQ-UHFFFAOYSA-N 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 210000003141 lower extremity Anatomy 0.000 description 1
- 238000003670 luciferase enzyme activity assay Methods 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 238000013507 mapping Methods 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 230000017074 necrotic cell death Effects 0.000 description 1
- 210000001577 neostriatum Anatomy 0.000 description 1
- 230000004770 neurodegeneration Effects 0.000 description 1
- 230000004766 neurogenesis Effects 0.000 description 1
- 230000003959 neuroinflammation Effects 0.000 description 1
- 108091027963 non-coding RNA Proteins 0.000 description 1
- 102000042567 non-coding RNA Human genes 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 230000008689 nuclear function Effects 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 229910052762 osmium Inorganic materials 0.000 description 1
- SYQBFIAQOQZEGI-UHFFFAOYSA-N osmium atom Chemical compound [Os] SYQBFIAQOQZEGI-UHFFFAOYSA-N 0.000 description 1
- 210000003254 palate Anatomy 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 231100000915 pathological change Toxicity 0.000 description 1
- 230000036285 pathological change Effects 0.000 description 1
- 230000001991 pathophysiological effect Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 230000010287 polarization Effects 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 238000012257 pre-denaturation Methods 0.000 description 1
- 238000007781 pre-processing Methods 0.000 description 1
- 210000002442 prefrontal cortex Anatomy 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 150000003254 radicals Chemical class 0.000 description 1
- 238000010814 radioimmunoprecipitation assay Methods 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 210000004761 scalp Anatomy 0.000 description 1
- 238000007789 sealing Methods 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 208000020431 spinal cord injury Diseases 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 210000002504 synaptic vesicle Anatomy 0.000 description 1
- 238000012353 t test Methods 0.000 description 1
- 230000000451 tissue damage Effects 0.000 description 1
- 231100000827 tissue damage Toxicity 0.000 description 1
- 208000037816 tissue injury Diseases 0.000 description 1
- 238000012549 training Methods 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 230000009752 translational inhibition Effects 0.000 description 1
- 230000009529 traumatic brain injury Effects 0.000 description 1
- -1 uranyl acetate-lead citrate Chemical compound 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 238000005303 weighing Methods 0.000 description 1
- 239000008096 xylene Substances 0.000 description 1
Abstract
The invention relates to a neuron protection and function recovery method after focal ischemic stroke and a verification experiment method, wherein the neuron protection and function recovery method is to inhibit nerve injury by regulating miR-212-5p/PLXNA2 so as to promote nerve protection and function recovery after ischemic stroke injury.
Description
Technical Field
The invention relates to the field of medical rehabilitation, in particular to a neuron protection and function recovery method after focal ischemic cerebral apoplexy aiming at a targeted gene technology.
Background
Cerebral stroke is a common cerebral vascular emergency that can cause serious brain tissue damage due to abrupt occlusion or rupture of blood vessels, preventing blood flow to the brain. Two main stroke types have been identified: hemorrhagic stroke and ischemic stroke. Ischemic stroke is characterized by high morbidity, mortality, and recurrence rates, accounting for about 80% of all strokes. Brain injury following permanent or transient focal cerebral ischemia can trigger a range of pathophysiological events including free radical release, microglial polarization, blood brain barrier dysfunction, neuronal apoptosis and neuroinflammation. Ischemic penumbra shows increased neuronal apoptosis during the acute phase of cerebral ischemia, a process which is considered an important component of cerebral ischemic injury. Rescue of ischemic penumbra is the heart of the neuroprotection concept of ischemic stroke.
Microglial cells serve as resident immune cells in the central nervous system, and have dual functions in brain tissue injury, regeneration and repair after ischemic stroke. Microglia are classified into pro-inflammatory M1 and anti-inflammatory M2. M2 microglial cells are thought to exert neuroprotective functions by promoting axonal regeneration, remyelination, neurogenesis and angiogenesis. The exosomes are inner vesicles of the multivesicular body with a diameter of 30-150nm, secreted by different types of cells in the brain, including microglia, neurons and astrocytes. Once the multivesicular body fuses with the plasma membrane, the exosomes are released into the extracellular space while protecting their contents from degradation. They are involved in the transport of functional biochemical substances such as messenger RNAs (mrnas), microRNA (miRNAs), cytokines and proteins and thus, in intercellular communication by direct transfer of genetic material to target cells.
Recent studies have shown that exosome miRNAs exhibit regulatory effects on target cells and thus may in fact represent a new way of intracellular communication. Notably, miRNAs are small non-coding RNA molecules that are processed into ribonucleoprotein complexes that bind to target mRNA, resulting in mRNA degradation or translational inhibition. One miRNA can be combined with a plurality of targets, and simultaneously, the miRNAs are inhibited from participating in biological processes functionally, so that the treatment effect on diseases is realized. According to increasing evidence, miRNAs have important functions in a wide range of neurological diseases, such as spinal cord injury, traumatic brain injury and cerebral stroke. They are the essential mediators of the endogenous neuroprotective response of ischemic preconditioning in the brain. Furthermore, miRNAs play a key role in the establishment of functional neurons by modulating neuronal morphology. Some miRNAs are very abundant in the brain and they can act as effector factors required for the development and maintenance of neuronal phenotypes. They are also expressed in dendrites, which are involved in synaptic plasticity by modulating synaptic and dendritic spine structures. Therefore, how to find out some miRNAs with protection and recovery effects on cerebral apoplexy patients through targeting experiments becomes a technical problem which needs to be solved currently.
Disclosure of Invention
The invention aims to solve the technical problems, and the neuron protection and function recovery method is to inhibit nerve injury by regulating miR-212-5p/PLXNA2 so as to promote nerve protection and function recovery after ischemic brain injury. The verification experiment method is used for quantitatively analyzing the phenotype change of microglial cells in a cortical ischemia semi-dark area after transient cerebral ischemia by detecting the levels of intracellular pro-inflammatory cytokines and anti-inflammatory cytokines through qRT-PCR on brain tissues so as to verify the neuroprotection and functional recovery conditions. The verification experiment method is characterized in that on the 3 rd day after operation, microglial exosomes are collected from ischemic penumbra of a cerebral apoplexy animal model, and Western blot analysis of NTA, TEM and exosome surface markers is used for identifying separated exosome samples. The verification experiment method is a behavior test to investigate whether miR-212-5p can improve the exercise function of the MCAO/R animal, and specifically a step walking test is used.
Through the recovery method and the experimental method, whether PLXNA gene in miR-212-5p can inhibit nerve injury better or not can be further judged, so that ischemic brain injury is promoted.
Drawings
FIG. 1 determination of proinflammatory cytokine levels and anti-inflammatory cytokine levels 6h, 1,3, 5, 7 days post-operation;
FIG. 2 miRNA sequencing of microglial exosomes 3 days post MCAO/R surgery;
FIG. 3 treatment with agomir-212-5p may reduce neurological dysfunction 7 days after MCAO/R;
FIG. 4 treatment with agomir-212-5p reduces cerebral infarct volume 7 days after MCAO/R, reduces neuronal apoptosis;
FIG. 5 inhibition of PLXNA and RhoA/ROCK2 expression in ischemic penumbra 3 days after MCAO/R using agomir-212-5p treatment;
FIG. 6 treatment with agomir-212-5p promotes synaptic plasticity at day 7 after MCAO/R and reduces axonal degeneration;
FIG. 7 treatment with agomir-212-5p may promote the formation and growth of dendritic spines on day 7 after MCAO/R surgery;
FIG. 8 inhibition of PLXNA and RhoA/ROCK2 expression in PC12 cells using agomir-212-5p treatment;
FIG. 9 treatment with agomir-212-5p reduces neuronal apoptosis in PC12 cells after OGD/R and reduces axonal degeneration; FIG. 10 schematically illustrates the potential molecular mechanisms that can exert neuroprotective effects to reduce neuronal damage following ischemic stroke using agomir-212-5p therapy.
Detailed Description
FIGS. 1A-E are assays of proinflammatory cytokine levels and anti-inflammatory cytokine levels 6h, 1, 3, 5,7 days post-surgery. Pro-inflammatory cytokines (IL-1β, IL-6, INOS, CD32 and CD 86) released by microglia. Anti-inflammatory cytokines (anti-inflammatory cytokines TGF-beta, IL-10 and CD 206) released by F-H microglia. I the expression of CD86 (green) and Iba1 (red) in the cortical ischemia penumbra at different time points was examined by immunofluorescence staining. J immunofluorescent staining examined CD206 (green) and Iba1 (red) expression at different time points. Scale bar = 50 μm. Data are expressed as mean ± standard error (n=5 per group). *P<0.05,** P <0.01 compared to sham.
FIG. 2 miRNA sequencing of microglial exosomes 3 days post MCAO/R surgery. A, B describe microglial exosomes using nanoparticle tracking analysis and transmission electron microscopy scanning. Scale bar = 200nm. The exosome markers CD9, CD63 and CD81 were detected by C Western blot. D microglial cell detection was performed by Western blot with microglial cell marker CD11 b. E shows a differential heat map of miRNAs expression in microglial exosomes 3 days post MCAO/R surgery. F adopts qRT-PCR to detect the expressions of miR-30c-5p, miR-126a-5p, miR-128-3p, miR-212-5p and miR-1949 in the semi-dark band of the cortical ischemia after MCAO/R operation. 3 days after G MCAO/R surgery, the expression levels of miRNA target genes PLXNA2, PTEN and FOXO3 in the cortical ischemia penumbra. H miR-212-5p is at a target site in the 3 'untranslated region (3' UTR) of PLXNA mRNA. I pmirGLO map of luciferase reporter vector. J double luciferase assay showed binding of miR-212-5p to the 3' UTR of PLXNA 2. Data are expressed as mean ± standard error (n=5 per group). *P<0.05,** P <0.01 compared to sham surgery group; # P <0.05 compared to miR-NC group.
FIG. 3 treatment with agomir-212-5p reduces neurological dysfunction 7 days after MCAO/R and improves motor function. Flow chart of experiment a. Step B, step B walking test scoring standard schematic diagram. And C, testing results of a step walking test. The score for neurological deficit score was Zea Longa on the scale of Zea Longa. Waveforms of the respective sets of Motion Evoked Potentials (MEPs). Amplitude of each set of MEPs. Latency of MEPs of group G. Each group n=6. Schematic representation of H CatWalk gait analysis. I run duration(s). J average running speed (cm/s). K right forelimb stride (cm). L right hind limb stride (cm). Data are expressed as mean ± standard error (n=8 per group). And (5) analyzing the connectivity of the brain functions among the M four groups. *P<0.05,** P <0.01 compared to sham surgery group; #P<0.05,## P < value was 0.01 compared to the MCAO/R group.
FIG. 4 treatment with agomir-212-5p reduced cerebral infarction volume 7 days after MCAO/R, and reduced neuronal apoptosis. At day 7 post AMCAO/R, brain sections were stained with TTC to observe ischemic lesions. The white areas show infarct cores. Quantitative analysis of infarct size percentage. C H & E staining. Scale bar = 1000 μm (low magnification) and 50 μm (high magnification). DNissl staining. Scale bar = 50 μm. Immunofluorescent staining of E, F NeuN showed survival of peri-infarct cortical and striatal neurons 7 days after MCAO/R. Data are expressed as mean ± standard error (n=3 per group). *P<0.05,** P <0.01 compared to sham surgery group; #P<0.05,## P <0.01 compared to the MCAO/R group.
FIG. 5 treatment with agomir-212-5p inhibits PLXNA and RhoA/ROCK2 expression in ischemic penumbra at day 3 after MCAO/R. The A asterisks indicate ischemic penumbra. B, analyzing the expression condition of PLXNA, rhoA and ROCK2 mRNA in the ipsilateral brain by adopting a qRT-PCR method. Representative images of Western blot of groups PLXNA, rhoA and ROCK2, C. Beta-actin was used as an internal reference. Quantitative analysis of DWestern blot results. Immunofluorescent staining of PLXNA, rhoA and ROCK2 proteins in each group E of neurons. Scale bar = 50 μm. Data are expressed as mean ± standard error (n=4-6 per group). *P<0.05,** P <0.01 compared to sham surgery group; # P <0.05 and ## P <0.01 compared to the MCAO/R group.
FIG. 6 treatment with agomir-212-5p promotes synaptic plasticity at day 7 after MCAO/R and reduces axonal degeneration. The ultrastructure of ischemic penumbra synapses was analyzed using transmission electron microscopy. Scale bar = 2 μm. B pre-synaptic (purple) and post-synaptic (green) structural schematic. The number of synapses in the cerebral cortical ischemic penumbra of each group C. Width of D synaptic space (nm). Each group n=3. E immunofluorescent staining showed expression of MAP-2 (green) in the cortical ischemic penumbra. Nuclei were stained with DAPI and blue was seen. Scale bar = 50 μm. Representative images of F Nogo-A, ngR and GAP-43 Western blots. And GWestern blot, quantitatively analyzing the detection result. Each group n=4-6. H-J immunofluorescent staining showed Nogo-A, ngR and βIII tubulin expression in the cortical ischemic penumbra. Nuclei were stained with DAPI and blue was seen. Scale bar = 50 μm. Data are expressed as mean ± standard error. *P<0.05,** P <0.01 compared to sham surgery group; #P<0.05,## P <0.01 compared to the MCAO/R group.
FIG. 7 treatment with agomir-212-5p promotes dendritic spine formation and growth of dendritic spines on day 7 after MCAO/R surgery. Representative golgi stained pictures of the brains of each group of rats. Scale bar = 50 μm and 20 μm respectively. B, calculating the top dendrite intersection point when the distance from the central point is continuously increased. Total number of intersections of the C-roof dendrites. D the basal dendrite intersection point is calculated as the distance from the center point increases. Total number of intersections of E-based dendrites. Total number of branches of the F roof dendrite. Total number of branches of G-base dendrites. Dendritic spine plot on H-roof dendrites. Scale bar = 10 μm. I is the density of dendritic spines along the top dendrites. Dendritic spine map on J-base dendrites. Scale bar = 10 μm. Density of dendritic spine along basal dendrites. Data are expressed as mean ± standard error (n=3 per group). *P<0.05,** P <0.01 compared to sham surgery group; #P<0.05,## P < value was 0.01 compared to the MCAO/R group.
FIG. 8 treatment with agomir-212-5p inhibited PLXNA and RhoA/ROCK2 expression in PC12 cells. Representative Western blot images of a show PLXNA, rhoA and ROCK2 levels for each group. Beta-actin is used as an internal reference. And BWesternblot, quantitatively analyzing the detection result. Immunofluorescent staining results of PLXNA, rhoA and ROCK2 (green) proteins in each group of neurons of C-E. Nuclei were stained with DAPI and blue was seen. Scale bar = 50 μm. F-H mRNA levels of PLXNA, rhoA, and ROCK2 were measured in each group of patients using qRT-PCR. Data are expressed as mean ± standard error (n=4-6 per group). * P <0.05 and ** P <0.01 compared to sham surgery groups; # P <0.05 and ## P < value was 0.01 compared to the MCAO/R group.
FIG. 9 treatment with agomir-212-5p reduced neuronal apoptosis in PC12 cells following OGD/R and reduced axonal degeneration. ACLEAVED CASPASE-3 immunofluorescent staining (green). Nuclei were stained with DAPI and blue was seen. Scale bar = 50 μm. Expression levels of Nogo-A and NgR genes in B-C PC12 cells. The gene expression level, β -actin, was detected by qRT-PCR as an internal control. D-F immunofluorescent staining showed PC12 cell Nogo-A, ngR and βIII tubulin expression levels. Nuclei were stained with DAPI and blue was seen. Scale bar = 50 μm. Data are expressed as mean ± standard error (n=5 per group). *P<0.05,** P <0.01 compared to sham surgery group; #P<0.05,## P <0.01 compared to the MCAO/R group.
FIG. 10 schematically illustrates the potential molecular mechanisms that can exert neuroprotective effects to reduce neuronal damage following ischemic stroke using agomir-212-5p therapy.
The levels of intracellular pro-inflammatory cytokines and anti-inflammatory cytokines were detected by qRT-PCR and the time course of the cortical ischemic penumbra microglial phenotypic changes after transient cerebral ischemia was quantified. mRNA expression of M1-type microglial-related pro-inflammatory mediators IL-6, IL-1. Beta., iNOS increased significantly and gradually after cerebral ischemia, peaking at 24h (FIGS. 1A-C). The expression of CD32 and CD86 increased gradually after ischemia and remained elevated for at least 7 days (fig. 1D and E). For M2 microglial-related markers, including TGF- β, IL-10 and CD206, we found in this study that TGF- β expression peaked 24 hours post-surgery (FIG. 1F). IL-10 and CD206 were induced after MCAO/R and peaked 3-5 days after injury (FIG. 1G, H). Immunofluorescent staining of cerebral cortical ischemic penumbra CD86/Iba1 and CD206/Iba1 showed that expression of CD86 increased gradually over time for at least 7 days (FIG. 1I). Furthermore, CD206 was significantly lower in expression on day 7 than on days 3 and 5 (fig. 1J). Thus, M2 microglial markers were expressed abundantly on day 3 post-surgery. On day 3 after MCAO/R, the expression of miR-212-5p in microglial exosomes from the ischemic penumbra is reduced; Microglial exosomes were collected from ischemic penumbra cortex 3 days after MCAO/R, further investigating the role of microglial exosomes miRNAs in pathological changes after ischemic stroke. Isolated exosome samples were identified using Western blot analysis of NTA, TEM and exosome markers (CD 9, CD63, CD 81). As shown in FIG. 2A, B, the exosomes are in the form of typical cup-shaped membrane vesicles, approximately 30-120nm in diameter. Exosome specific markers CD9, CD63 and CD81 were all at high levels in the pellet (fig. 2C). Western blot analysis also showed high expression of CD11b in the pellet (FIG. 2D). The expression level of miRNA in pseudomodule and MCAO/R mouse microglial exosomes was detected by sequencing. The heat map of miRNA expression is shown in figure 2E. The change in miRNA expression was further verified by qRT-PCR. We screened miR-30c-5p, miR-126a-5p, miR-128-3p, miR-212-5p and miR-1949, which could be the cause of neurodegeneration. Experimental results showed significant downregulation of ischemic penumbra miRNAs (fig. 2F). Based on our miRNA sequencing and qRT-PCR results, we found that miR-212-5p expression was significantly reduced in the ischemic penumbra, which we selected for further study. Searching the predictive database found several predicted mRNA targets of miR-212-5p, which might be involved in the neuroprotective effect of miR-212-5 p. Expression levels of PLXNA, PTEN, and FOXO3 in ischemic penumbra were examined (fig. 2G). Then, a dual luciferase reporter system was performed to verify PLXNA2 as a direct target for miR-212-5 p. transfection of miR-212-5p significantly reduced luciferase activity in PC12 cells transfected with PLXNA2 wild-type 3' -UTR, but did not inhibit luciferase activity in cells containing the mutant structure, indicating that PLXNA is a direct target for miR-212-5p (fig. 2H-J).
MiR-212-5p can improve motor function recovery and relieve brain injury;
FIG. 3A is a flow chart of an experiment. We performed behavioral tests to investigate whether miR-212-5p could improve motor function in MCAO/R rats, including the step walking test (FIG. 3B). The right-hand step rate was significantly higher after MCAO/R than in the sham-operated group, while the MCAO/R+ agomir-212-5p group was significantly lower on day 7 post-operation (FIG. 3C). The MCAO/R rats had higher neurological deficit scores, while the MCAO/r+ agomir-212-5p group had a lower neurological deficit score than the MCAO/R group (fig. 3D). After MCAO/R, MEP latency of animals was prolonged and amplitude peaks were reduced. Animals in the MCAO/R+ agomir-212-5p group were significantly improved 7 days after MCAO/R (FIG. 3E-G). Furthermore, we performed CatWalk gait analysis to assess motor function (fig. 3H). The longer the run time after MCAO/R surgery, the slower the average run speed. Agomir-212-5p shortens the run time to some extent (fig. 3I) and also increases the average run speed (fig. 3J). At all measurement time points after MCAO/R, the stride of the affected limb was significantly reduced. The stride of the rats treated with miR-212-5p was significantly improved compared to the MCAO/R rats (FIG. 3K, L). The FC analysis results showed that MCAO/R rats had reduced functional connectivity of the motor cortex on the affected side with the cortex after healthy side compression, sensory cortex on the affected side, bilateral caudate nucleus, and ventral portion of the healthy side hippocampus compared to sham surgery group. In addition, MCAO/r+ agomir-NC group showed reduced connection of the affected motor cortex to the affected motor cortex, the affected prefrontal cortex, and the healthy caudate nucleus function in rats after molding, compared to sham group. However, compared with the MCAO/R group, the MCAO/R+ agomir-212-5p group of rats had elevated functional connection of the motor cortex on the affected side to the sensory cortex on the healthy side, the motor cortex on the healthy side and the piriform cortex on the affected side. At the same time, the motor cortex of the MCAO/R+ agomir-212-5p group had an elevated gray matter function connection around the motor cortex of the affected side and the healthy side aqueduct compared to the MCAO/R+ agomir- + group (FIG. 3M and Table 3). In summary, treatment with agomir-212-5p promotes MEP recovery, enhances the connectivity of the brain neural network and significantly improves motor behavior by activating functional connections between the affected motor cortex and sensory cortex, the full brain motor cortex.
We collected rat brains on day 7 post MCAO/R and stained with TTC to examine the effect of agomir-212-5p treatment on cerebral infarct volume (FIG. 4A). TTC staining showed a significant increase in the white area of the MCAO/R group, while administration of agomir-212-5p significantly reduced the cerebral infarct volume in the MCAO/R rats (FIG. 4B). This change may indicate that agomir-212-5p has neuroprotective effects. Tissue sections were stained with H & E and Nissl, showing histological changes in neurons, and further confirming neuroprotection by miR-212-5 p. H & E staining showed a disorder of cell alignment, interstitial edema, and massive cell necrosis in MCAO/R rats (FIG. 4C). Nissl staining showed a decrease in the number of Nissl individuals in the ischemic penumbra of the MCAO/R group. Agomir-212-5p treatment was effective in improving brain tissue and neuronal damage (FIG. 4D). NeuN staining further showed reduced neuronal apoptosis after agomir-212-5p treatment of the cortex (FIG. 4E, F) and striatum (FIG. 4G, H) compared to the MCAO/R group. Taken together, these results indicate that miR-212-5p can effectively alleviate brain damage induced by cerebral ischemia. MiR-212-5p promotes neuroprotection in MCAO/R rats by targeting PLXNA;
The mechanism of neuroprotection of miR-212-5p is further studied by detecting the expression of miR-212-5p target gene PLXNA. On day 3 post MCAO/R, qRT-PCR analysis showed significant increases in PLXNA, rhoA and ROCK2 levels, whereas agomir-212-5p treatment significantly down-regulated their expression in ischemic penumbra (fig. 5B). Western blot results were consistent with qRT-PCR results (FIGS. 5C, D). Finally, we detected the expression of PLXNA, rhoA and ROCK2 in neurons by immunofluorescent staining and labeled ischemic penumbral neurons with NeuN antibodies. Immunofluorescent staining showed that agomir-212-5p attenuated PLXNA, rhoA and ROCK2 expression in neurons 3 days after MCAO/R (fig. 5E). The results suggest that PLXNA and RhoA/ROCK2 may be involved in the neuroprotection of miR-212-5 p.
MiR-212-5p promotes synaptic plasticity in MCAO/R rats and reduces axonal degeneration;
The synaptic structure of the sham is complete, including complete pre-synaptic and post-synaptic structures. Fewer synaptic vesicles, blurring or absence of synaptic clefts, and abnormal synaptic structure following MCAO/R surgery (fig. 6A). The presynaptic and postsynaptic structures are represented in light purple and green, respectively (FIG. 6B). After agomir-212-5p treatment, both the number of synapses and the inter-synaptic cleft were improved (FIGS. 6C, D). We observed a significant decrease in MAP-2 staining density, however, agomir-212-5p treatment could reverse this phenomenon (fig. 6E). Next, we studied the effect of agomir-212-5p on the attenuation of axonal degeneration. Western blot results showed that the expression levels of Nogo-A and NgR were significantly lower in the MCAO/R groups after the dry prognosis of the MCAO/R+212-5p group (FIGS. 6F, G). The results of Nogo-A and NgR immunofluorescence staining were consistent with the results of Western blot experiments (FIG. 6H, I). Treatment with agomir-212-5p was effective in reducing MCAO/R-induced axonal injury (figure 6J). Taken together, these findings suggest that increasing miR-212-5p expression may be a potential strategy to promote synaptic plasticity and inhibit axonal degeneration following MCAO/R-induced focal cerebral ischemia.
MiR-212-5p promotes formation and growth of dendritic spines in rats after MCAO/R surgery;
We measured the complexity of the dendritic structures in vivo by performing golgi staining on brain sections to assess the effect of miR-212-5 p. On day 7 post MCAO/R, the complexity of the apical and basal dendrites was significantly reduced, the number of dendrite intersections was reduced in MCAO/R rats (fig. 7A-E), and dendrite branches were reduced (fig. 7f, g). We also compared the density of dendritic spines on the apical and basal dendrites. In calculating the number of dendritic spines, a significant decrease in dendritic spine density was found after MCAO/R. However, the application agomir-212-5p can reduce the decrease in dendritic spine density (FIGS. 7H-K). Taken together, these data suggest that miR-212-5p may play an important role in promoting dendritic spine formation and promoting dendritic growth.
In vitro experiments further prove that miR-212-5p promotes neuroprotection through targeting PLXNA < 2 >;
In an in vitro study, we used PC12 cells of the OGD/R model to assess the neuroprotective effects of miR-212-5 p. We first studied PLXNA, rhoA and ROCK2 protein expression levels using Western blot analysis. PLXNA2, rhoA and ROCK2 were expressed in cells with similar results to rat brain tissue (fig. 8a, b). In addition, after OGD/R injury, the intensity of immunofluorescent staining was increased for PLXNA, rhoA, and ROCK2, but reversed for agomir-212-5p (FIGS. 8C-E). These results were further confirmed by qRT-PCR (FIG. 8F-H). Taken together, PLXNA2 may be involved in the action of miR-212-5 p.
MiR-212-5p can reduce apoptosis and axonal degeneration in vitro;
As shown in FIG. 9A, immunofluorescent staining showed a significant increase in C-caspase3 levels after OGD/R, whereas agomir-212-5p treatment significantly reduced cell death. nogoA and NgR mRNA levels were higher in the OGD/R group than in the agomir-212-5p treated group (FIGS. 9B, C). Also, the results were further confirmed by cellular immunofluorescence experiments. OGD/R increased Nogo-A and NgR mRNA expression and decreased βIII tubulin expression, while agomir-212-5p treatment significantly reversed these changes (FIG. 9D-F). These results indicate that miR-212-5p has neuroprotective effects on cell death in vitro.
Materials and methods;
Animals
We obtained adult male SD rats weighing 260-280 grams from Shanghai laboratory animal research center. Only male rats were used in this study, as previous studies demonstrated the neuroprotective effect of estrogens on brain ischemic animal models. We raised all rats under standard laboratory conditions of 23.+ -. 2 ℃, 40% -50% humidity and 12-12 hours light-dark cycle. These animals can obtain food and water. All protocols were reviewed and approved by the institutional animal ethics committee of the Shanghai university of traditional Chinese medicine, and all animal procedures followed guidelines of national institutional laboratory guidelines for animal care and use.
Establishing an animal model of ischemic cerebral apoplexy and injecting lateral ventricles;
Rats were anesthetized with sodium pentobarbital (30 mg/kg, intraperitoneal injection). The rats were then placed in a supine position with a midline incision made in the neck, exposing the left External Carotid Artery (ECA), internal Carotid Artery (ICA), and Common Carotid Artery (CCA). ECA was ligated with surgical wire to block blood flow, and left ICA and CCA were temporarily clamped with a microcatheter. In ECA, a small incision is made. The thrombus was then gently advanced from the ECA stump to the ICA to occlude blood flow in the MCA. After the fixation of the wire bolt for 2 hours, ICA perfusion was resumed. The sham rats were subjected to the same procedure except for the insertion of the plug.
Animals were randomly divided into 4 groups: sham, MCAO/R, MCAO/R+ agomir-NC, MCAO/R+ agomir-212-5p. According to previous reports, agomir-NC and agomir-212-5p were injected through the lateral ventricle 30min before MCAO/R. The skull is exposed by making an incision along the midline of the scalp. The injection coordinates were 1mm posteriorly and 1.5mm laterally (to the left) from the anterior fontanel and 3.5mm below the skull. Agomir-212-5p or agomir-NC (10. Mu.M in 7. Mu.l) was slowly injected. At the same time, 7. Mu.l of 0.9% sodium chloride was injected into the ventricles of both the sham-operated and MCAO/R groups. The needle was left in place for 5min after injection.
Isolation of microglial exosomes in the injured brain; . The ischemic penumbra of the cortex rapidly separated from the left hemisphere. The tissue was digested with collagenase at 37℃for 15 min. Then, the mixture was centrifuged at 2000 Xg at 4℃for 10min to remove cellular fibers and debris, and the supernatant was collected and centrifuged again at 10000 Xg at 4℃for 30min to remove cellular debris. After collecting the supernatant, it was filtered through a 0.22 μm filter. The sample was centrifuged at 100000 Xg at 4℃for 70min and the supernatant was removed. The pellet was resuspended in 0.35. Mu.l Dulbecco's PBS to obtain an exosome suspension. Mu.g of CD11b antibody was added to 50. Mu.l of 3% BSA and the overlaid samples were incubated at room temperature for 60min to isolate microglial exosomes. Subsequently, 10. Mu.l Pierce TMStreptavidin Plus UltraLinkTM Resin was added to 40. Mu.l 3% BSA and the covered samples incubated for 30min at room temperature. Then, the mixture was centrifuged at 800 Xg for 10min. After centrifugation, the supernatant was discarded, and microglial exosomes were recovered and stored at 4 ℃ for subsequent analysis.
Exosome identification;
Exosomes were identified using nanoparticle tracking analysis, transmission electron microscopy and Western blot analysis. First, exosomes were analyzed by NTA. To measure the size and concentration of exosome particles we used NTAZetaView PMX and zetaview8.04.02. Isolated exosome samples were diluted appropriately with 1 XPBS. NTA detection was performed at 11 positions and the measurement results were recorded. The ZetaView system was calibrated using 110nm polystyrene particles. The temperature is maintained at about 26.26 deg.c to 27.21 deg.c. And analyzing the exosome morphology by a transmission electron microscope. In 50 μl of 2% PFA, the exosomes were resuspended, and 5 μl of exosomes were dropped onto Formvar-carbon-loaded copper mesh and washed 3 times with PBS. The copper mesh was placed on 50 μl 1% glutaraldehyde droplets for 5min, and then washed. After washing with distilled water, the copper mesh was placed on 50. Mu.l uranium oxalate droplets (pH 7) for 5min, and then transferred to 50. Mu.l methylcellulose droplets for 10min. Excess liquid was removed with filter paper and the sample was allowed to air dry for 10min. Electron micrographs were obtained by JEOL 1230 transmission electron microscopy.
Expression of exosome marker proteins CD9, CD63 and CD81 was analyzed by Western blot. We used CD11b to identify microglial cells. Exosome samples were first lysed with RIPA buffer, total proteins separated on a 12% sds-PAGE gel, transferred to PVDF membrane, blocked, and primary incubated overnight. Antibodies include CD9, CD63 and ITGAM/CD11b. PVDF membrane was then incubated with secondary antibody (1:5000) for 1 hour at room temperature. Protein signals were detected using a UVP imaging system.
MiRNA sequencing analysis and target gene prediction;
MiRNA sequencing was performed by Kangshen Biotech company in Shanghai, china. Total RNA samples were extracted with TRIzol reagent, quality and quantification of the extracted total RNA samples were performed by agarose electrophoresis and Nanodrop, library was constructed, library quality was assessed using an Agilent 2100Bioanalyzer, the library was denatured with 0.1M NaOH to generate single stranded DNA, and finally 51 cycle sequencing was performed on an Illumina NextSeq 500 sequencer. . TargetScan (http:// TargetScan. Org /) and miRDB (http:// mirdb. Org/miRDB /) were used to predict the miRNA target genes, and the consensus predicted genes were selected as potential target genes for miRNA.
Double luciferase reporter gene experiments;
wild-type PLXNA 'UTR (WT) or PLXNA' UTR Mutant (MUT) of miR-212-5p binding site was inserted into the dual-luciferase pmirGLO vector. Luminescence was detected using a GloMax instrument (Promega) with a dual luciferase reporter assay.
Cell culture and oxygen glucose deprivation/reoxygenation (Oxygen glucose deprivation/reoxygenation, OGD/R) modeling;
We obtained PC12 cells from Shanghai cell biology institute of China academy of sciences and cultured them in DMEM containing 10% fetal bovine serum and 1% penicillin/streptomycin. Cells were cultured in 5% CO 2 at 37 ℃. For the cell transfection experiments, cells were transfected with either agomir-212-5p or agomir-NC at 100 nM. Cells were used for subsequent experiments 24h after transfection. We then changed the cell culture broth to sugar-free medium. Cells were incubated for 4 hours (1%O 2,94%N2 and 5% CO 2) in a hypoxic incubator at 37 ℃. Subsequently, the sugarless medium was changed to a normal medium and reoxygenation was performed with maintenance under standard conditions (5% CO 2 and 37 ℃).
Behavioral testing;
Neurological deficit was scored using Zea Longa scores: 0 = no signs of neurological deficit; 1 = right front paw cannot be fully straightened; 2 = right turn; 3 = toppling to the right; 4 = inability to walk spontaneously, loss of consciousness.
A junction walking test;
Each group had 8 rats. We tested the ability of rats to walk on irregularly spaced (1-3 cm) ladders and recorded video to assess impairment of right forelimb function after stroke. Scoring was based on the following criteria: 0 = complete miss; 1 = severe slipping; 2 = light slipping; 3 = reset; 4 = correction; 5 = partial placement; 6 = correctly placed. The step rate refers to the number of steps with a step score of 0, 1 or 2. To calculate the step rate, we use the following formula: (number of wrong steps/total steps for right forelimb) ×100%.
Catwalk gait analysis;
Each group included 8 rats. During the experiment, the room was black and quiet. Prior to the formal experiments, rats were subjected to an adaptive training for one week. The rats are placed at one end of a glass runway, food is taken as bait at the other end of the runway, and running conditions of the rats are recorded under the runway through a high-speed camera. The test was performed on day 1, 3, 5, and 7, respectively, before and after the operation. Data were collected and analyzed using catwalk version 10.6 software.
Exercise evoked potential (MEP);
Exercise evoked potential testing was performed using a Keypoint 9033A07 electromyography/evoked potential apparatus manufactured by Denmark at day 7 post-molding. Rats were anesthetized by intraperitoneal injection with sodium pentobarbital (30 mg/kg) for detection, and then fixed in the supine position. The recording electrode was placed on the right bicep muscle and the stimulating electrode was penetrated by the rat palate (here near the left motor cortex of the brain). The stimulation is performed by applying a single square wave electric pulse, the wave width is 100 mu s, the stimulation intensity gradually increases to the latency period and the wave amplitude are not changed obviously any more. The motor evoked potential latency and amplitude changes of the rats were recorded.
Resting state functional magnetic resonance imaging (fMRI);
Rat brain scans were performed using an 11.7T magnetic resonance scanner (Bruker, karlsruhe, germany). Rats were anesthetized using 5% isoflurane inhalation induction, fixed on a magnetic resonance scanning table, and kept anesthetized using 1.5% isoflurane in combination with 0.05mg/kg dexmedetomidine in prone position. Resting fMRI employs a single shot planar Echo imaging sequence (Echo PLANAR IMAGING, EPI) of the barrier scan, the parameters of which are shown below: flip angle=90°, scan layer thickness=0.3 mm, average number of times 1,repetition Time (TR) =3000 ms, echo Time (TE) = 8.142ms,field ofvision (FOV) =27×27mm 2. SPM8 (STASTISTICAL PARAMETTIC MAPPING) statistical analysis software package (http:// www.fil.ion.ucl.ac.uk/SPM /) of Matlab 2014b (MathWorks, USA) platform was used for data preprocessing and data analysis. The data is first converted to Nifti format using Bru < 2 > -nii toolbox, then time-layer correction (SLICE TIMING), head-movement correction (Realign) are performed using SPM8 software, then non-brain tissue is stripped off using MRIcroN software, and the origin of the image is reset to better register the individual brain image with the template brain. A normalization (Normalize) and smoothing (smooths) process is then performed. The motor cortex on the same side (left side) of the focus is selected as a region of interest (Region ofinterest, ROI), the functional connection (Functional Connectivity, FC) of the motor cortex on the affected side and each voxel of the whole brain is calculated, and statistical analysis is performed after Fisher' sZ transformation is performed. Different groups of rats were subjected to the two-sample t-test in SPM8 and the results were presented in a standard rat brain template. P <0.001 considers the difference statistically significant.
Assessing cerebral infarction volume;
The rat brain was rapidly removed and frozen at-20℃for 20min. Subsequently, the frozen brain was coronally sectioned to a thickness of 2mm for a total of 5 slices. . Brain tissue sections were stained with 2%2,3, 5-triphenyltetrazolium chloride (2, 3,5-trihenyl tetrazolium chloride, TTC), incubated in the dark at 37 ℃ for 20min, and then fixed with 4% paraformaldehyde. The infarct size was calculated using Image J software.
Western blot;
Brain tissue and cells were collected and specimens were lysed with RIPA lysate. Protein concentration was determined using BCA kit. After electrophoresis of the tissue samples (80. Mu.g) and the cell samples (30. Mu.g), they were transferred to PVDF membrane and blocked. After the end of the blocking, the PVDF membrane was taken out and placed in TBST for 5min X3 times, and then placed in an antibody incubation box separately, shaking table at 4℃overnight. The next day, PVDF membrane was placed in secondary antibody dilution solution and incubated on a shaking table at room temperature for 1 h. Imaging and picture acquisition was performed using the us UVP gel imaging system. The grey values of the bands were quantitatively analyzed using Image J software.
H & E and Nissl staining;
Nissl staining, hematoxylin-eosin (H & E) staining and immunofluorescence staining of tissue specimens were fixed in 4% paraformaldehyde for 24H, dehydrated with alcohol gradient, transparent xylene, paraffin embedded, and brain sections were performed with coronal plane from front to back, paraffin sections of 5 μm were prepared, guaranteeing five sections each. H & E staining and Nissl staining were performed according to the kit instructions.
Immunofluorescence staining;
Paraffin sections were dewaxed to water, sections were antigen thermally repaired, 10% goat serum blocking solution containing 0.3% triton X-100 was added dropwise, incubated for 1h at room temperature, and incubated overnight at 4 ℃ with the indicated primary antibodies. PC12 cells were washed with PBS, fixed with 4% paraformaldehyde, 0.3% Triton X-100 was infiltrated with PBS for 15min, and incubated with 10% goat serum albumin for 1h at room temperature. Next, the slides were incubated overnight at 4 ℃ against the first antibody. The following day, secondary antibodies were incubated, nuclei counterstained, and blocked. . Images were acquired with an inverted fluorescence microscope at 400 x field of view.
Fluorescent quantitative PCR (qRT-PCR);
Cell and brain tissue RNAs were extracted using TRIzol reagent and reverse transcribed into cdnas using reverse transcription kit. Gene expression levels were detected using SYBR Green kit and LightCycler 480 real-time fluorescent quantitation. The reaction conditions are as follows: pre-denaturation, 5min 1 cycle at 95 ℃; amplification was performed for 10s at 95℃denaturation, 10s at 60℃annealing, and 10s at 72℃extension, for 45 cycles. U6 (for miRNA) and beta-actin (for mRNA) references. Tables 1 and 2 show the primer sequences designed for this study. The relative expression level of the gene was analyzed by the method [73] of 2 -△△CT. The delta CT calculation formula is as follows: ΔΔct= (CT Target gene -CT Reference gene ) Experimental group -(CT Target gene -CT Reference gene ) Control group . Statistical methods were applied to compare the results between groups for statistical differences.
A transmission electron microscope;
Brain tissue was cut into 1mm 3, fixed with 2.5% glutaraldehyde for 2 hours, then samples were fixed with 1% osmium acid for 1 hour, dehydrated, and embedded. Then, ultrathin sections of 50nm are prepared, and dyeing is carried out by adopting a uranyl acetate-lead citrate double dyeing method. And (5) observing by using a transmission electron microscope, and acquiring an image.
Golgi staining, sholl analysis and dendritic spine density measurement;
The FD rapid golgi staining kit performs golgi staining. The extracted brain was soaked in a golgi dye solution for 2 weeks in the dark at room temperature. Immersing and washing 3 times with distilled water, immersing the tissue in 80% glacial acetic acid overnight, washing with distilled water after the tissue becomes soft, and placing in 30% sucrose. . The tissue was cut into slices with a thickness of 100 μm using an oscillating microtome, and the slices were attached to gelatin slides. Treating tissue slide with concentrated ammonia water for 15min, washing with distilled water for 1min, treating with fixing solution for 15min, washing with distilled water for 3min, air drying, and sealing with glycerol gelatin. Analysis of results was performed using Image J, neuronJ, shollAnalysis and NeuronStudio software.
Statistical analysis;
Experimental data were statistically analyzed using SPSS 22.0 software and expressed as mean ± Standard Error (SEM). The comparison between groups was performed by one-wayANOVA, the comparison between the variance individuals was performed by LSD method, and the variance individuals were tested by Dunnet's T3. P <0.05 considered the difference statistically significant.
Claims (4)
1. The neuron protection and function recovery method after focal ischemic cerebral apoplexy and the verification experiment method are characterized in that: the neuron protection and function recovery method is to inhibit nerve injury by regulating miR-212-5p/PLXNA2 so as to promote nerve protection and function recovery after ischemic cerebral apoplexy.
2. The method for protecting neurons and recovering functions after focal ischemic cerebral apoplexy and the verification experiment method according to claim 1, wherein the method is characterized in that: miR-212-5p in brain tissue of cerebral apoplexy animal model targets its downstream PLXNA gene to inhibit nerve injury, and qRT-PCR is used for detecting intracellular levels of pro-inflammatory cytokines and anti-inflammatory cytokines in brain tissue, and quantitative analysis is carried out on the phenotype change of cortical ischemia semi-dark band microglial cells after transient cerebral ischemia so as to verify neuroprotection and functional recovery.
3. The method for protecting neurons and recovering functions after focal ischemic cerebral apoplexy and the verification experiment method according to claim 2, wherein the method is characterized in that: the verification experiment method is that on the 3 rd day after operation, microglial exosomes are collected from ischemic penumbra of a cerebral apoplexy animal model, and separated exosome samples are identified by using Westernblot analysis of NTA, TEM and exosome surface markers.
4. The method for protecting neurons and recovering functions after focal ischemic cerebral apoplexy and the verification experiment method according to claim 2, wherein the method is characterized in that: the verification experiment method is a behavioural test to investigate whether miR-212-5p can improve the exercise function of MCAO/R animals, and specifically a step walking test is used.
Publications (1)
Publication Number | Publication Date |
---|---|
CN118924898A true CN118924898A (en) | 2024-11-12 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Long et al. | Astrocyte-derived exosomes enriched with miR-873a-5p inhibit neuroinflammation via microglia phenotype modulation after traumatic brain injury | |
Zhang et al. | Peripheral macrophage-derived exosomes promote repair after spinal cord injury by inducing local anti-inflammatory type microglial polarization via increasing autophagy | |
Sacheli et al. | Expression patterns of miR-96, miR-182 and miR-183 in the developing inner ear | |
Zhang et al. | Tenascin-C expression by neurons and glial cells in the rat spinal cord: changes during postnatal development and after dorsal root or sciatic nerve injury | |
Wei et al. | Zeb2/Axin2-enriched BMSC-derived exosomes promote post-stroke functional recovery by enhancing neurogenesis and neural plasticity | |
Yew et al. | Nitric oxide synthase neurons in different areas of normal aged and Alzheimer's brains | |
Hu et al. | Knock down of lncRNA H19 promotes axon sprouting and functional recovery after cerebral ischemic stroke | |
Goss et al. | Enhanced glial fibrillary acidic protein RNA response to fornix transection in aged mice | |
Mehta et al. | Beta-catenin (CTNNB1) induces Bmp expression in urogenital sinus epithelium and participates in prostatic bud initiation and patterning | |
Ke et al. | Protocadherin20 promotes excitatory synaptogenesis in dorsal horn and contributes to bone cancer pain | |
CN102686721B (en) | For the method and composition that the amplification of glial restricted progenitor cell in Mammals source, qualification, sign and usefulness strengthen | |
Chen et al. | Expression of WW domain-containing oxidoreductase WOX1 in the developing murine nervous system | |
Dakubo et al. | Control of glial precursor cell development in the mouse optic nerve by sonic hedgehog from retinal ganglion cells | |
Li et al. | Inhibition of miRNA-21 promotes retinal ganglion cell survival and visual function by modulating Müller cell gliosis after optic nerve crush | |
Khoshdel-Sarkarizi et al. | Developmental regulation and lateralization of GABA receptors in the rat hippocampus | |
JPH10500564A (en) | Neuron promoter and utilization | |
Wang et al. | Increased level of exosomal miR-20b-5p derived from hypothermia-treated microglia promotes neurite outgrowth and synapse recovery after traumatic brain injury | |
Zhang et al. | Microencapsulated olfactory ensheathing cell transplantation reduces P2X4 receptor overexpression and inhibits neuropathic pain in rats | |
Li et al. | Focal ischemic stroke modifies microglia-derived exosomal miRNAs: potential role of mir-212-5p in neuronal protection and functional recovery | |
Zhang et al. | Transplantation of microencapsulated neural stem cells inhibits neuropathic pain mediated by P2X7 receptor overexpression | |
Wang et al. | Combinatory effect of mesenchymal stromal cells transplantation and quercetin after spinal cord injury in rat. | |
CN108866179A (en) | LncRNA-SCARNA10 is in preparation liver fibrosis detection kit and the purposes for the treatment of hepatic fibrosis medicines | |
CN118924898A (en) | Neuron protection and function recovery method after focal ischemic cerebral apoplexy | |
Bouzaffour et al. | Implication of type 3 deiodinase induction in zebrafish fin regeneration | |
Attar et al. | Electron microscopic study of the progeny of ependymal stem cells in the normal and injured spinal cord |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication |