Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Neurochem Res ; 49(5): 1254-1267, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38381246

RESUMO

Fibrotic scars play important roles in tissue reconstruction and functional recovery in the late stage of nervous system injury. However, the mechanisms underlying fibrotic scar formation and regulation remain unclear. Casein kinase II (CK2) is a protein kinase that regulates a variety of cellular functions through the phosphorylation of proteins, including bromodomain-containing protein 4 (BRD4). CK2 and BRD4 participate in fibrosis formation in a variety of tissues. However, whether CK2 affects fibrotic scar formation remains unclear, as do the mechanisms of signal regulation after cerebral ischemic injury. In this study, we assessed whether CK2 could modulate fibrotic scar formation after cerebral ischemic injury through BRD4. Primary meningeal fibroblasts were isolated from neonatal rats and treated with transforming growth factor-ß1 (TGF-ß1), SB431542 (a TGF-ß1 receptor kinase inhibitor) or TBB (a highly potent CK2 inhibitor). Adult SD rats were intraperitoneally injected with TBB to inhibit CK2 after MCAO/R. We found that CK2 expression was increased in vitro in the TGF-ß1-induced fibrosis model and in vivo in the MCAO/R injury model. The TGF-ß1 receptor kinase inhibitor SB431542 decreased CK2 expression in fibroblasts. The CK2 inhibitor TBB reduced the increases in proliferation, migration and activation of fibroblasts caused by TGF-ß1 in vitro, and it inhibited fibrotic scar formation, ameliorated histopathological damage, protected Nissl bodies, decreased infarct volume and alleviated neurological deficits after MCAO/R injury in vivo. Furthermore, CK2 inhibition decreased BRD4 phosphorylation both in vitro and in vivo. The findings of the present study suggested that CK2 may control BRD4 phosphorylation to regulate fibrotic scar formation, to affecting outcomes after ischemic stroke.


Assuntos
Benzamidas , Proteínas que Contêm Bromodomínio , Caseína Quinase II , Cicatriz , Dioxóis , AVC Isquêmico , Animais , Ratos , Caseína Quinase II/antagonistas & inibidores , Caseína Quinase II/metabolismo , Cicatriz/metabolismo , Cicatriz/patologia , Fibroblastos/metabolismo , Fibrose , AVC Isquêmico/complicações , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/metabolismo , Proteínas Nucleares , Fosforilação , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Proteínas que Contêm Bromodomínio/efeitos dos fármacos , Proteínas que Contêm Bromodomínio/metabolismo
2.
J Neuroinflammation ; 19(1): 95, 2022 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-35429978

RESUMO

BACKGROUND: Excessively deposited fibrotic scar after spinal cord injury (SCI) inhibits axon regeneration. It has been reported that platelet-derived growth factor receptor beta (PDGFRß), as a marker of fibrotic scar-forming fibroblasts, can only be activated by platelet-derived growth factor (PDGF) B or PDGFD. However, whether the activation of the PDGFRß pathway can mediate fibrotic scar formation after SCI remains unclear. METHODS: A spinal cord compression injury mouse model was used. In situ injection of exogenous PDGFB or PDGFD in the spinal cord was used to specifically activate the PDGFRß pathway in the uninjured spinal cord, while intrathecal injection of SU16f was used to specifically block the PDGFRß pathway in the uninjured or injured spinal cord. Immunofluorescence staining was performed to explore the distributions and cell sources of PDGFB and PDGFD, and to evaluate astrocytic scar, fibrotic scar, inflammatory cells and axon regeneration after SCI. Basso Mouse Scale (BMS) and footprint analysis were performed to evaluate locomotor function recovery after SCI. RESULTS: We found that the expression of PDGFD and PDGFB increased successively after SCI, and PDGFB was mainly secreted by astrocytes, while PDGFD was mainly secreted by macrophages/microglia and fibroblasts. In addition, in situ injection of exogenous PDGFB or PDGFD can lead to fibrosis in the uninjured spinal cord, while this profibrotic effect could be specifically blocked by the PDGFRß inhibitor SU16f. We then treated the mice after SCI with SU16f and found the reduction of fibrotic scar, the interruption of scar boundary and the inhibition of lesion and inflammation, which promoted axon regeneration and locomotor function recovery after SCI. CONCLUSIONS: Our study demonstrates that activation of PDGFRß pathway can directly induce fibrotic scar formation, and specific blocking of this pathway would contribute to the treatment of SCI.


Assuntos
Axônios , Cicatriz , Indóis , Regeneração Nervosa , Pirróis , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Traumatismos da Medula Espinal , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Cicatriz/tratamento farmacológico , Cicatriz/etiologia , Cicatriz/metabolismo , Cicatriz/patologia , Fibrose , Indóis/farmacologia , Locomoção , Camundongos , Regeneração Nervosa/efeitos dos fármacos , Regeneração Nervosa/fisiologia , Proteínas Proto-Oncogênicas c-sis/metabolismo , Pirróis/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Recuperação de Função Fisiológica , Medula Espinal/patologia , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia
3.
Cell Tissue Res ; 387(3): 351-360, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34189605

RESUMO

Formation of a collagenous connective tissue scar after penetrating injuries to the brain or spinal cord has been described and investigated for well over 100 years. However, it was studied almost exclusively in the context of penetrating injuries that resulted in infiltration of meningeal fibroblasts, which raised doubts about translational applicability to most CNS injuries where the meninges remain intact. Recent studies demonstrating the perivascular niche as a source of fibroblasts have debunked the traditional view that a fibrotic scar only forms after penetrating lesions that tear the meninges. These studies have led to a renewed interest in CNS fibrosis not only in the context of axon regeneration after spinal cord injury, but also across a spectrum of CNS disorders. Arising with this renewed interest is some discrepancy about which perivascular cell gives rise to the fibrotic scar, but additional studies are beginning to provide some clarity. Although mechanistic studies on CNS fibrosis are still lacking, the similarities to fibrosis of other organs should provide important insight into how CNS fibrosis can be therapeutically targeted to promote functional recovery.


Assuntos
Axônios , Traumatismos da Medula Espinal , Astrócitos/patologia , Sistema Nervoso Central , Cicatriz/patologia , Fibrose , Humanos , Meninges/patologia , Regeneração Nervosa/fisiologia , Traumatismos da Medula Espinal/patologia
4.
Cell Tissue Res ; 385(3): 539-555, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33864501

RESUMO

Neuron-glia antigen 2 (NG2) proteoglycan and platelet-derived growth factor receptor beta (PDGFR-ß) are widely used markers of pericytes, which are considered cells that form fibrotic scars in response to central nervous system insults. However, the exact phenotypes of NG2- and PDGFR-ß-expressing cells, as well as the origin of the fibrotic scar after central nervous system insults, are still elusive. In the present study, we directly examined the identities and distributions of NG2- and PDGFR-ß-positive cells in the control and lesioned striatum injured by the mitochondrial toxin 3-nitropropionic acid. Immunoelectron microscopy and correlative light and electron microscopy clearly distinguished NG2 and PDGFR-ß expression in the vasculature during the post-injury period. Vascular smooth muscle cells and pericytes expressed NG2, which was prominently increased after the injury. NG2 expression was restricted to these vascular mural cells until 14 days post-lesion. By contrast, PDGFR-ß-positive cells were perivascular fibroblasts located abluminal to smooth muscle cells or pericytes. These PDGFR-ß-expressing cells formed extravascular networks associated with collagen fibrils at 14 days post-lesion. We also found that in the injured striatal parenchyma, PDGFR-ß could be used as a complementary marker of resting and reactive NG2 glia because activated microglia/macrophages shared only the NG2 expression with NG2 glia in the lesioned striatum. These data indicate that NG2 and PDGFR-ß label different vascular mural and parenchymal cells in the healthy and injured brain, suggesting that fibrotic scar-forming cells most likely originate in PDGFR-ß-positive perivascular fibroblasts rather than in NG2-positive pericytes.


Assuntos
Lesões Encefálicas/induzido quimicamente , Encéfalo/fisiopatologia , Fibroblastos/metabolismo , Fibrose/metabolismo , Nitrocompostos/efeitos adversos , Propionatos/efeitos adversos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
5.
Neurobiol Dis ; 134: 104674, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31731043

RESUMO

Remyelination failure is a crucial component of disease progression in the autoimmune demyelinating disease Multiple Sclerosis (MS). The regenerative capacity of oligodendrocyte progenitor cells (OPCs) to replace myelinating oligodendrocytes is likely influenced by many aspects of the lesion environment including inflammatory signaling and extracellular matrix (ECM) deposition. These features of MS lesions are typically attributed to infiltrating leukocytes and reactive astrocytes. Here we demonstrate that fibroblasts also contribute to the inhibitory environment in the animal model of MS, experimental autoimmune encephalomyelitis (EAE). Using Col1α1GFP transgenic mice, we show that perivascular fibroblasts are activated in the spinal cord at EAE onset, and infiltrate the parenchyma by the peak of behavioral deficits where they are closely associated with areas of demyelination, myeloid cell accumulation, and ECM deposition. We further show that both fibroblast conditioned media and fibroblast ECM inhibit the differentiation of OPCs into mature oligodendrocytes. Taken together, our results indicate that the fibrotic scar is a major component of EAE pathology that leads to an inhibitory environment for remyelination, thus raising the possibility that anti-fibrotic mechanisms may serve as novel therapeutic targets for MS.


Assuntos
Diferenciação Celular , Encefalomielite Autoimune Experimental/patologia , Oligodendroglia/patologia , Oligodendroglia/fisiologia , Medula Espinal/patologia , Animais , Fibroblastos/patologia , Fibrose , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/patologia , Substância Branca/patologia
6.
J Neurosci Res ; 98(5): 826-842, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31758600

RESUMO

Scar formation after injury of the brain or spinal cord is a common event. While glial scar formation by astrocytes has been extensively studied, much less is known about the fibrotic scar, in particular after stroke. Platelet-derived growth factor receptor ß-expressing (PDGFRß+ ) pericytes have been suggested as a source of the fibrotic scar depositing fibrous extracellular matrix (ECM) proteins after detaching from the vessel wall. However, to what extent these parenchymal PDGFRß+ cells contribute to the fibrotic scar and whether targeting these cells affects fibrotic scar formation in stroke is still unclear. Here, we utilize male transgenic mice that after a permanent middle cerebral artery occlusion stroke model have a shift from a parenchymal to a perivascular location of PDGFRß+ cells due to the loss of regulator of G-protein signaling 5 in pericytes. We find that only a small fraction of parenchymal PDGFRß+ cells co-label with type I collagen and fibronectin. Consequently, a reduction in parenchymal PDGFRß+ cells by ca. 50% did not affect the overall type I collagen or fibronectin deposition after stroke. The redistribution of PDGFRß+ cells to a perivascular location, however, resulted in a reduced thickening of the vascular basement membrane and changed the temporal dynamics of glial scar maturation after stroke. We demonstrate that parenchymal PDGFRß+ cells are not the main contributor to the fibrotic ECM, and therefore targeting these cells might not impact on fibrotic scar formation after stroke.


Assuntos
Encéfalo/patologia , Matriz Extracelular/patologia , Gliose/patologia , Pericitos/patologia , Acidente Vascular Cerebral/patologia , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Fibrose/metabolismo , Fibrose/patologia , Gliose/metabolismo , Masculino , Camundongos , Pericitos/metabolismo , Acidente Vascular Cerebral/metabolismo
7.
Acta Neuropathol ; 137(5): 785-797, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30929040

RESUMO

Virtually all phases of spinal cord injury pathogenesis, including inflammation, cell proliferation and differentiation, as well as tissue remodeling, are mediated in part by infiltrating monocyte-derived macrophages. It is now clear that these infiltrating macrophages have distinct functions from resident microglia and are capable of mediating both harmful and beneficial effects after injury. These divergent effects have been largely attributed to environmental cues, such as specific cytokines, that influence the macrophage polarization state. In this review, we also consider the possibility that different macrophage origins, including the spleen, bone marrow, and local self-renewal, may also affect macrophage fate, and ultimately their function that contribute to the complex pathobiology of spinal cord injury.


Assuntos
Macrófagos/patologia , Macrófagos/fisiologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Animais , Humanos
8.
J Neurosci ; 37(9): 2362-2376, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28130359

RESUMO

Although infiltrating macrophages influence many pathological processes after spinal cord injury (SCI), the intrinsic molecular mechanisms that regulate their function are poorly understood. A major hurdle has been dissecting macrophage-specific functions from those in other cell types as well as understanding how their functions change over time. Therefore, we used the RiboTag method to obtain macrophage-specific mRNA directly from the injured spinal cord in mice and performed RNA sequencing to investigate their transcriptional profile. Our data show that at 7 d after SCI, macrophages are best described as foam cells, with lipid catabolism representing the main biological process, and canonical nuclear receptor pathways as their potential mediators. Genetic deletion of a lipoprotein receptor, CD36, reduces macrophage lipid content and improves lesion size and locomotor recovery. Therefore, we report the first macrophage-specific transcriptional profile after SCI and highlight the lipid catabolic pathway as an important macrophage function that can be therapeutically targeted after SCI.SIGNIFICANCE STATEMENT The intrinsic molecular mechanisms that regulate macrophage function after spinal cord injury (SCI) are poorly understood. We obtained macrophage-specific mRNA directly from the injured spinal cord and performed RNA sequencing to investigate their transcriptional profile. Our data show that at 7 d after SCI, macrophages are best described as foam cells, with lipid catabolism representing the main biological process and canonical nuclear receptor pathways as their potential mediators. Genetic deletion of a lipoprotein receptor, CD36, reduces macrophage lipid content and improves lesion size and locomotor recovery. Therefore, we report the first macrophage-specific transcriptional profile after SCI and highlight the lipid catabolic pathway as an important macrophage function that can be therapeutically targeted after SCI.


Assuntos
Metabolismo dos Lipídeos/fisiologia , Macrófagos/metabolismo , Traumatismos da Medula Espinal/patologia , Animais , Transplante de Medula Óssea , Antígenos CD36/genética , Antígenos CD36/metabolismo , Movimento Celular/genética , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/genética , Hemaglutininas/metabolismo , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Metabolismo dos Lipídeos/genética , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Ribossômico/administração & dosagem , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Transdução de Sinais/genética , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/cirurgia
9.
Biochem Biophys Res Commun ; 496(4): 1302-1307, 2018 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-29410176

RESUMO

After traumatic spinal cord injury (SCI), a scar may form with a fibrotic core (fibrotic scar) and surrounding reactive astrocytes (glial scar) at the lesion site. The scar tissue is considered a major obstacle preventing regeneration both as a physical barrier and as a source for secretion of inhibitors of axonal regeneration. Understanding the mechanism of scar formation and how to control it may lead to effective SCI therapies. Using a compression-SCI model on adult transgenic mice, we demonstrate that the canonical Wnt/ß-catenin signaling reporter TOPgal (TCF/Lef1-lacZ) positive cells appeared at the lesion site by 5 days, peaked on 7 days, and diminished by 14 days post injury. Using various representative cell lineage markers, we demonstrate that, these transiently TOPgal positive cells are a group of Fibronectin(+);GFAP(-) fibroblast-like cells in the core scar region. Some of them are proliferative. These results indicate that Wnt/ß-catenin signaling may play a key role in fibrotic scar formation after traumatic spinal cord injury.


Assuntos
Cicatriz/metabolismo , Cicatriz/patologia , Compressão da Medula Espinal/metabolismo , Compressão da Medula Espinal/patologia , Medula Espinal/patologia , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Cicatriz/etiologia , Fibrose , Proteína Glial Fibrilar Ácida , Camundongos , Camundongos Transgênicos , Medula Espinal/metabolismo , Compressão da Medula Espinal/complicações
10.
Cell Mol Neurobiol ; 37(5): 771-782, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27517720

RESUMO

Acute injury of central nervous system (CNS) starts a cascade of morphological, molecular, and functional changes including formation of a fibrotic scar, expression of transforming growth factor beta 1 (TGF-ß1), and expression of extracellular matrix proteins leading to arrested neurite outgrowth and failed regeneration. We assessed alteration of electrophysiological properties of cerebellar granule cells (CGCs) in two in vitro models of neuronal injury: (i) model of fibrotic scar created from coculture of meningeal fibroblasts and cerebral astrocytes with addition of TGF-ß1; (ii) a simplified model based on administration of TGF-ß1 to CGCs culture. Both models reproduced suppression of neurite outgrowth caused by neuronal injury, which was equally restored by chondroitinase ABC (ChABC), a key disruptor of fibrotic scar formation. Voltage-dependent calcium current was not affected in either injury model. However, intracellular calcium concentration could be altered as an expression of inositol trisphosphate receptor type 1 was suppressed by TGF-ß1 and restored by ChABC. Voltage-dependent sodium current was significantly suppressed in CGCs cultured on a model of fibrotic scar and was only partly restored by ChABC. Administration of TGF-ß1 significantly shifted current-voltage relation of sodium current toward more positive membrane potential without change to maximal current amplitude. Both transient and sustained potassium currents were significantly suppressed on a fibrotic scar and restored by ChABC to their control amplitudes. In contrast, TGF-ß1 itself significantly upregulated transient and did not change sustained potassium current. Observed changes of voltage-dependent ion currents may contribute to known morphological and functional changes in injured CNS.


Assuntos
Canais de Cálcio/metabolismo , Cerebelo/patologia , Ativação do Canal Iônico , Neurônios/metabolismo , Neurônios/patologia , Canais de Potássio/metabolismo , Canais de Sódio/metabolismo , Animais , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Modelos Biológicos , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurônios/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Fator de Crescimento Transformador beta1/farmacologia
11.
Neurobiol Dis ; 74: 114-25, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25461258

RESUMO

Spinal cord injury (SCI) leads to formation of a fibrotic scar that is inhibitory to axon regeneration. Recent evidence indicates that the fibrotic scar is formed by perivascular fibroblasts, but the mechanism by which they are recruited to the injury site is unknown. Using bone marrow transplantation in mouse model of spinal cord injury, we show that fibroblasts in the fibrotic scar are associated with hematogenous macrophages rather than microglia, which are limited to the surrounding astroglial scar. Depletion of hematogenous macrophages results in reduced fibroblast density and basal lamina formation that is associated with increased axonal growth in the fibrotic scar. Cytokine gene expression analysis after macrophage depletion indicates that decreased Tnfsf8, Tnfsf13 (tumor necrosis factor superfamily members) and increased BMP1-7 (bone morphogenetic proteins) expression may serve as anti-fibrotic mechanisms. Our study demonstrates that hematogenous macrophages are necessary for fibrotic scar formation and macrophage depletion results in changes in multiple cytokines that make the injury site less fibrotic and more conducive to axonal growth.


Assuntos
Axônios/fisiologia , Cicatriz/prevenção & controle , Macrófagos/fisiologia , Regeneração Nervosa/fisiologia , Traumatismos da Medula Espinal/fisiopatologia , Animais , Axônios/patologia , Membrana Basal/patologia , Membrana Basal/fisiopatologia , Transplante de Medula Óssea/métodos , Proteínas Morfogenéticas Ósseas/metabolismo , Ligante CD30/metabolismo , Cicatriz/patologia , Cicatriz/fisiopatologia , Modelos Animais de Doenças , Feminino , Fibroblastos/patologia , Fibroblastos/fisiologia , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Traumatismos da Medula Espinal/patologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
12.
Osteoarthritis Cartilage ; 23(11): 1879-89, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26521733

RESUMO

OBJECTIVE: Articular cartilage defects commonly result from traumatic injury and predispose to degenerative joint diseases. To test the hypothesis that aberrant healing responses and chronic inflammation lead to osteoarthritis (OA), we examined spatiotemporal changes in joint tissues after cartilage injury in murine knees. Since intra-articular injection of hyaluronan (HA) can attenuate injury-induced osteoarthritis in wild-type (WT) mice, we investigated a role for HA in the response to cartilage injury in mice lacking HA synthase 1 (Has1(-/-)). DESIGN: Femoral groove cartilage of WT and Has1(-/-) mice was debrided to generate a non-bleeding wound. Macroscopic imaging, histology, and gene expression were used to evaluate naïve, sham-operated, and injured joints. RESULTS: Acute responses (1-2 weeks) in injured joints from WT mice included synovial hyperplasia with HA deposition and joint-wide increases in expression of genes associated with inflammation, fibrosis, and extracellular matrix (ECM) production. By 4 weeks, some resurfacing of damaged cartilage occurred, and early cell responses were normalized. Cartilage damage in Has1(-/-) mice also induced early responses; however, at 4 weeks, inflammation and fibrosis genes remained elevated with widespread cartilage degeneration and fibrotic scarring in the synovium and joint capsule. CONCLUSIONS: We conclude that the ineffective repair of injured cartilage in Has1(-/-) joints can be at least partly explained by the markedly enhanced expression of particular genes in pathways linked to ECM turnover, IL-17/IL-6 cytokine signaling, and apoptosis. Notably, Has1 ablation does not alter gross HA content in the ECM, suggesting that HAS1 has a unique function in the metabolism of inflammatory HA matrices.


Assuntos
Cartilagem Articular/patologia , Regulação da Expressão Gênica , Glucuronosiltransferase/deficiência , Glucuronosiltransferase/genética , Articulação do Joelho/patologia , Osteoartrite do Joelho/enzimologia , RNA/genética , Animais , Cartilagem Articular/enzimologia , Cartilagem Articular/lesões , Doença Crônica , Modelos Animais de Doenças , Fibrose/enzimologia , Fibrose/patologia , Glucuronosiltransferase/biossíntese , Hialuronan Sintases , Inflamação/enzimologia , Inflamação/genética , Inflamação/patologia , Articulação do Joelho/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite do Joelho/genética , Osteoartrite do Joelho/patologia , Reação em Cadeia da Polimerase
13.
CNS Neurosci Ther ; 30(7): e14826, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38973179

RESUMO

AIM: We aimed to confirm the inhibitory effect of nicotinamide on fibrotic scar formation following spinal cord injury in mice using functional metabolomics. METHODS: We proposed a novel functional metabolomics strategy to establish correlations between gene expression changes and metabolic phenotypes using integrated multi-omics analysis. Through the integration of quantitative metabolites analysis and assessments of differential gene expression, we identified nicotinamide as a functional metabolite capable of inhibiting fibrotic scar formation and confirmed the effect in vivo using a mouse model of spinal cord injury. Furthermore, to mimic fibrosis models in vitro, primary mouse embryonic fibroblasts and spinal cord fibroblasts were stimulated by TGFß, and the influence of nicotinamide on TGFß-induced fibrosis-associated genes and its underlying mechanism were examined. RESULTS: Administration of nicotinamide led to a reduction in fibrotic lesion area and promoted functional rehabilitation following spinal cord injury. Nicotinamide effectively downregulated the expression of fibrosis genes, including Col1α1, Vimentin, Col4α1, Col1α2, Fn1, and Acta2, by repressing the TGFß/SMADs pathway. CONCLUSION: Our functional metabolomics strategy identified nicotinamide as a metabolite with the potential to inhibit fibrotic scar formation following SCI by suppressing the TGFß/SMADs signaling. This finding provides new therapeutic strategies and new ideas for clinical treatment.


Assuntos
Cicatriz , Fibrose , Camundongos Endogâmicos C57BL , Niacinamida , Traumatismos da Medula Espinal , Animais , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/complicações , Cicatriz/tratamento farmacológico , Cicatriz/patologia , Cicatriz/metabolismo , Cicatriz/prevenção & controle , Camundongos , Fibrose/tratamento farmacológico , Fator de Crescimento Transformador beta/metabolismo , Metabolômica , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Feminino
14.
Neurochem Int ; 173: 105674, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38184171

RESUMO

The moderate formation of the fibrotic scar plays an important role in functional recovery after stroke. M2a macrophages have been identified as an important source of early fibrosis after cerebral ischemia. However, the underlying mechanisms by which macrophages interact with fibroblasts in this context remain largely unknown. Therefore, our study aimed to further investigate the potential mechanisms underlying the effects of macrophages on fibroblasts following ischemic stroke. In vitro and in vivo, recombinant rat interleukin 4 (IL4) was used to induce macrophages to polarize into M2a macrophages. In vitro, primary Sprague-Dawley newborn rat meningeal-derived fibroblasts were treated with PU.1 knockdown, the PU.1 inhibitor DB1976 or the mTOR inhibitor rapamycin, which were then co-cultured with M2a macrophage conditioned medium (MCM). In vivo, Sprague-Dawley adult rats were infected with negative control adenoviruses or PU.1-shRNA adenoviruses. Ten days after infection, an injury model of middle cerebral artery occlusion/reperfusion (MCAO/R) was constructed. Subsequently, IL4 was injected intracerebroventricularly to induce M2a macrophages polarization. In vitro, M2a MCM upregulated PU.1 expression and promoted the differentiation, proliferation, migration and extracellular matrix generation of fibroblasts, which could be reversed by treatment with the PU.1 inhibitor DB1976 or PU.1 knockdown. In vivo, PU.1 expression in fibroblasts was increased within ischemic core following MCAO/R, and this upregulation was further enhanced by exposure to IL4. Treatment with IL4 promoted fibrosis, increased angiogenesis, reduced apoptosis and infarct volume, as well as mitigated neurological deficits after MCAO/R, and these effects could be reversed by PU.1 knockdown. Furthermore, both in vivo and in vitro studies showed that IL4 treatment increased the levels of phosphorylated Akt and mTOR proteins, which were markedly decreased by PU.1 knockdown. Additionally, the use of an mTOR inhibitor rapamycin obviously suppressed the migration and differentiation of fibroblasts, and Col1 synthesis. In conclusion, our findings suggest for the first time that M2a macrophages, at least in part, regulate fibrosis and affect the outcome after cerebral ischemic stroke via the PU.1/mTOR signaling pathway in fibroblasts.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Ratos , Animais , Ratos Sprague-Dawley , Interleucina-4/metabolismo , Acidente Vascular Cerebral/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Macrófagos/metabolismo , Traumatismo por Reperfusão/metabolismo , AVC Isquêmico/metabolismo , Fibrose , Fibroblastos/metabolismo , Sirolimo
15.
J Neurotrauma ; 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38534205

RESUMO

In the past decade, signature clinical neuropathology of blast-induced traumatic brain injury has been under intense debate, but interface astroglial scarring (IAS) seems to be convincing. In this study, we examined whether IAS could be replicated in the rat brain exposed to a laser-induced shock wave(s) (LISW[s]), a tool that can produce a pure shock wave (primary mechanism) without dynamic pressure (tertiary mechanism). Under certain conditions, we observed astroglial scarring in the subpial glial plate (SGP), gray-white matter junctions (GM-WM), ventricular wall (VW), and regions surrounding cortical blood vessels, accurately reproducing clinical IAS. We also observed shock wave impulse-dependent meningeal damage (dural microhemorrhage) in vivo by transcranial near-infrared (NIR) reflectance imaging. Importantly, there were significant correlations between the degree of dural microhemorrhage and the extent of astroglial scarring more than 7 days post-exposure, suggesting an association of meningeal damage with astroglial scarring. The results demonstrated that the primary mechanism alone caused the IAS and meningeal damage, both of which are attributable to acoustic impedance mismatching at multi-layered tissue boundaries. The time course of glial fibrillary acidic protein (GFAP) immunoreactivity depended not only on the LISW conditions but also on the regions. In the SGP, significant increases in GFAP immunoreactivity were observed at 3 days post-exposure, whereas in the GM-WM and VW, GFAP immunoreactivity was not significantly increased before 28 days post-exposure, suggesting different pathological mechanisms. With the high-impulse single exposure or the multiple exposure (low impulse), fibrotic reaction or fibrotic scar formation was observed, in addition to astroglial scarring, in the cortical surface region. Although there are some limitations, this seems to be the first report on the shock-wave-induced IAS rodent model. The model may be useful to explore potential therapeutic approaches for IAS.

16.
Brain Res ; 1819: 148533, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37586675

RESUMO

Extracellular matrix (ECM) is a complex and dynamic network of proteoglycans, proteins, and other macromolecules that surrounds cells in tissues. The ECM provides structural support to cells and plays a critical role in regulating various cellular functions. ECM remodeling is a dynamic process involving the breakdown and reconstruction of the ECM. This process occurs naturally during tissue growth, wound healing, and tissue repair. However, in the context of central nervous system (CNS) injuries, dysregulated ECM remodeling can lead to the formation of fibrotic and glial scars. CNS injuries encompass various traumatic events, including concussions and fractures. Following CNS trauma, the formation of glial and fibrotic scars becomes prominent. Glial scars primarily consist of reactive astrocytes, while fibrotic scars are characterized by an abundance of ECM proteins. ECM remodeling plays a pivotal and tightly regulated role in the development of these scars after spinal cord and brain injuries. Various factors like ECM components, ECM remodeling enzymes, cell surface receptors of ECM molecules, and downstream pathways of ECM molecules are responsible for the remodeling of the ECM. The aim of this review article is to explore the changes in ECM during normal physiological conditions and following CNS injuries. Additionally, we discuss various approaches that target various factors responsible for ECM remodeling, with a focus on promoting axon regeneration and functional recovery after CNS injuries. By targeting ECM remodeling, it may be possible to enhance axonal regeneration and facilitate functional recovery after CNS injuries.


Assuntos
Axônios , Traumatismos da Medula Espinal , Humanos , Axônios/metabolismo , Gliose/metabolismo , Cicatriz/metabolismo , Regeneração Nervosa/fisiologia , Sistema Nervoso Central/metabolismo , Matriz Extracelular/metabolismo , Traumatismos da Medula Espinal/metabolismo , Fibrose , Astrócitos/metabolismo
17.
Mol Neurobiol ; 60(4): 2200-2208, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36633805

RESUMO

Astroglial-fibrotic scars resulted from spinal cord injury affect motor and sensory function, leading to paralysis. In particular, the fibrotic scar is a main barrier that disrupts neuronal regeneration after spinal cord injury. However, the association between astrocytes and fibrotic scar formation is not yet understood. We have previously demonstrated that the transcriptional factor Cebpd contributes to astrogliosis, which promotes glial scar formation after spinal cord injury. Herein, we show that fibrotic scar formation was decreased in the epicenter region in Cebpd-/- mice after contusive spinal cord injury and astrocytic Cebpd promoted fibroblast migration through secretion of Ptx3. Furthermore, the expression of Mmp3 was increased under recombinant protein Ptx3 treatment in fibroblasts by observing microarray data, resulting in fibroblast migration. In addition, regulation of Mmp3 occurs through the NFκB signaling pathway by using an irreversible inhibitor of IκBα phosphorylation in pretreated fibroblasts. Of note, we used the synthetic peptide RI37, which blocks fibroblast migration and decreases fibroblast Mmp3 expression in IL-1ß-treated astrocyte conditioned media. Collectively, our data suggest that fibroblast migration can be affected by astrocytic Cebpd through the Ptx3/NFκB/Mmp3 axis pathway and that the RI37 peptide may act as a therapeutic medicine to inhibit fibrotic scar formation after spinal cord injury.


Assuntos
Cicatriz , Traumatismos da Medula Espinal , Camundongos , Animais , Cicatriz/patologia , Astrócitos/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Traumatismos da Medula Espinal/patologia , Fibrose , Gliose/patologia , Medula Espinal/patologia
18.
J Neurotrauma ; 40(23-24): 2580-2595, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-36879472

RESUMO

Following spinal cord injury (SCI), fibrotic scar inhibits axon regeneration and impairs neurological function recovery. It has been reported that T cell-derived interferon (IFN)-γ plays a pivotal role in promoting fibrotic scarring in neurodegenerative disease. However, the role of IFN-γ in fibrotic scar formation after SCI has not been declared. In this study, a spinal cord crush injury mouse was established. Western blot and immunofluorescence showed that IFN-γ was surrounded by fibroblasts at 3, 7, 14, and 28 days post-injury. Moreover, IFN-γ is mainly secreted by T cells after SCI. Further, in situ injection of IFN-γ into the normal spinal cord resulted in fibrotic scar formation and inflammation response at 7 days post-injection. After SCI, the intraperitoneal injection of fingolimod (FTY720), a sphingosine-1-phosphate receptor 1 (S1PR1) modulator and W146, an S1PR1 antagonist, significantly reduced T cell infiltration, attenuating fibrotic scarring via inhibiting IFN-γ/IFN-γR pathway, while in situ injection of IFN-γ diminished the effect of FTY720 on reducing fibrotic scarring. FTY720 treatment inhibited inflammation, decreased lesion size, and promoted neuroprotection and neurological recovery after SCI. These findings demonstrate that the inhibition of T cell-derived IFN-γ by FTY720 suppressed fibrotic scarring and contributed to neurological recovery after SCI.


Assuntos
Doenças Neurodegenerativas , Traumatismos da Medula Espinal , Camundongos , Animais , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Cicatriz/tratamento farmacológico , Cicatriz/etiologia , Cicatriz/metabolismo , Interferon gama , Axônios/patologia , Doenças Neurodegenerativas/patologia , Regeneração Nervosa/fisiologia , Fibrose , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo , Inflamação/patologia , Medula Espinal/metabolismo
19.
Neural Regen Res ; 18(8): 1666-1671, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36751777

RESUMO

Monocytes, including monocyte-derived macrophages and resident microglia, mediate many phases of optic nerve injury pathogenesis. Resident microglia respond first, followed by infiltrating macrophages which regulate neuronal inflammation, cell proliferation and differentiation, scar formation and tissue remodeling following optic nerve injury. However, microglia and macrophages have distinct functions which can be either beneficial or detrimental to the optic nerve depending on the spatial context and temporal sequence of their activity. These divergent effects are attributed to pro- and anti-inflammatory cytokines expressed by monocytes, crosstalk between monocyte and glial cells and even microglia-macrophage communication. In this review, we describe the dynamics and functions of microglia and macrophages in neuronal inflammation and regeneration following optic nerve injury, and their possible role as therapeutic targets for axonal regeneration.

20.
Matrix Biol ; 118: 69-91, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36918086

RESUMO

Fibrotic scars appear after spinal cord injury (SCI) and are mainly composed of fibroblasts and excess extracellular matrix (ECM), including different types of collagen. The temporal and spatial distribution and role of excess collagens and ECM after SCI are not yet fully understood. Here, we identified that the procollagen type I C-terminal propeptide (PICP), a marker of collagen type I deposition, and bone morphogenetic protein 1 (BMP1), a secreted procollagen c-proteinase (PCP) for type I collagen maturation, were significantly elevatedin cerebrospinal fluid of patients with SCI compared with healthy controls, and were associated with spinal cord compression and neurological symptoms. We revealed the deposition of type I collagen in the area damaged by SCI in mice and confirmed that BMP1 was the only expressed PCP and induced collagen deposition. Furthermore, transforming growth factor-ß (TGF-ß), tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) can activate the expression of BMP1. However, inhibition of BMP1 at the acute phase eliminated fibrotic scars in the damaged area and inhibited activation and enrichment of astrocytes, which made the damage difficult to repair and increased hematoma. Unexpectedly, knockdown of Bmp1 by adeno-associated virus or the inhibition of BMP1 biological function by specific inhibitors and monoclonal antibodies at different time points after injury led to distinct therapeutic effects. Only delayed inhibition of BMP1 improved axonal regeneration and myelin repair at the subacute stage post-injury, and led to the recovery of motor function, suggesting that scarring had a dual effect. Early inhibition of the scarring was not conducive to limiting inflammation, while excessive scar formation inhibited the growth of axons. After SCI, the collagen deposition indicators increased in both human cerebrospinal fluid and mouse spinal cord. Therefore, suppression of BMP1 during the subacute phase improves nerve function after SCI and is a potential target for scar reduction.


Assuntos
Colágeno Tipo I , Traumatismos da Medula Espinal , Humanos , Camundongos , Animais , Proteína Morfogenética Óssea 1/genética , Proteína Morfogenética Óssea 1/metabolismo , Colágeno Tipo I/metabolismo , Cicatriz/patologia , Colágeno/genética , Colágeno/metabolismo , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Fibrose
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa