Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 578
Filtrar
1.
Front Immunol ; 15: 1402349, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38938572

RESUMO

Objective: Immunoregulation is a complex and critical process in the pathological process of spinal cord injury (SCI), which is regulated by various factors and plays an important role in the functional repair of SCI. This study aimed to explore the research hotspots and trends of glial cell immunoregulation after SCI from a bibliometric perspective. Methods: Data on publications related to glial cell immunoregulation after SCI, published from 2004 to 2023, were obtained from the Web of Science Core Collection. Countries, institutions, authors, journals, and keywords in the topic were quantitatively analyzed using the R package "bibliometrix", VOSviewer, Citespace, and the Bibliometrics Online Analysis Platform. Results: A total of 613 papers were included, with an average annual growth rate of 9.39%. The papers came from 36 countries, with the United States having the highest output, initiating collaborations with 27 countries. Nantong University was the most influential institution. We identified 3,177 authors, of whom Schwartz, m, of the Weizmann Institute of Science, was ranked first regarding both field-specific H-index (18) and average number of citations per document (151.44). Glia ranked first among journals with 2,574 total citations. The keywords "microglia," "activation," "macrophages," "astrocytes," and "neuroinflammation" represented recent hot topics and are expected to remain a focus of future research. Conclusion: These findings strongly suggest that the immunomodulatory effects of microglia, astrocytes, and glial cell interactions may be critical in promoting nerve regeneration and repair after SCI. Research on the immunoregulation of glial cells after SCI is emerging, and there should be greater cooperation and communication between countries and institutions to promote the development of this field and benefit more SCI patients.


Assuntos
Bibliometria , Neuroglia , Traumatismos da Medula Espinal , Traumatismos da Medula Espinal/imunologia , Humanos , Neuroglia/imunologia , Animais , Astrócitos/imunologia
2.
CNS Neurosci Ther ; 30(6): e14781, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38887195

RESUMO

BACKGROUND: Traumatic brain injury (TBI) and spinal cord injury (SCI) are acquired injuries to the central nervous system (CNS) caused by external forces that cause temporary or permanent sensory and motor impairments and the potential for long-term disability or even death. These conditions currently lack effective treatments and impose substantial physical, social, and economic burdens on millions of people and families worldwide. TBI and SCI involve intricate pathological mechanisms, and the inflammatory response contributes significantly to secondary injury in TBI and SCI. It plays a crucial role in prolonging the post-CNS trauma period and becomes a focal point for a potential therapeutic intervention. Previous research on the inflammatory response has traditionally concentrated on glial cells, such as astrocytes and microglia. However, increasing evidence highlights the crucial involvement of lymphocytes in the inflammatory response to CNS injury, particularly CD8+ T cells and NK cells, along with their downstream XCL1-XCR1 axis. OBJECTIVE: This review aims to provide an overview of the role of the XCL1-XCR1 axis and the T-cell response in inflammation caused by TBI and SCI and identify potential targets for therapy. METHODS: We conducted a comprehensive search of PubMed and Web of Science using relevant keywords related to the XCL1-XCR1 axis, T-cell response, TBI, and SCI. RESULTS: This study examines the upstream and downstream pathways involved in inflammation caused by TBI and SCI, including interleukin-15 (IL-15), interleukin-12 (IL-12), CD8+ T cells, CD4+ T cells, NK cells, XCL1, XCR1+ dendritic cells, interferon-gamma (IFN-γ), helper T0 cells (Th0 cells), helper T1 cells (Th1 cells), and helper T17 cells (Th17 cells). We describe their proinflammatory effect in TBI and SCI. CONCLUSIONS: The findings suggest that the XCL1-XCR1 axis and the T-cell response have great potential for preclinical investigations and treatments for TBI and SCI.


Assuntos
Lesões Encefálicas Traumáticas , Quimiocinas C , Traumatismos da Medula Espinal , Humanos , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/patologia , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Animais , Quimiocinas C/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Doenças Neuroinflamatórias/imunologia
3.
Int Immunopharmacol ; 137: 112505, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-38908081

RESUMO

BACKGROUND: Blood always shows coagulation changes after spinal cord injury (SCI), and identifying these blood changes may be helpful for diagnosis and treatment of SCI. Nevertheless, studies to date on blood coagulation changes after SCI in humans are not comprehensive. Therefore, this study aims to identify blood coagulation diagnostic biomarkers and immune changes related to SCI and its severity levels. METHODS: Human blood sequencing datasets were obtained from public databases. Differentially expressed coagulation-related genes were analyzed (DECRGs). Enrichment analysis and assessment of immune changes were conducted. Weighted gene co-expression network analysis, least absolute shrinkage and selection operator logistic regression were used to identify biomarkers. Validation for these biomarkers was performed. The correlation between biomarkers and immune cells was evaluated. Transcription factors, miRNA, lncRNA, and drugs that can regulate biomarkers were analyzed. RESULTS: DECRGs associated with SCI and its different grades were identified, showing enrichment in altered coagulation and immune-related signaling pathways. ADAM9, CD55, and STAT4 were identified as coagulation diagnostic biomarkers for SCI. IRF4 and PABPC4 were identified as coagulation diagnostic biomarkers for American Spinal Injury Association Impairment Scale (AIS) A grade of SCI. GP9 was designated as a diagnostic biomarker for AIS D grade of SCI. Immune changes in blood of SCI and its different grades were observed. Correlation between diagnostic biomarkers and immune cells were identified. Transcription factors, miRNA, lncRNA, and drugs that can regulate diagnostic biomarker expression were discovered. CONCLUSION: Therefore, detecting the expression of these putative diagnostic biomarkers and related immune changes may be helpful for predicting the severity of SCI. Uncovering potential regulatory mechanisms for biomarkers may be beneficial for further research.


Assuntos
Biomarcadores , Coagulação Sanguínea , Traumatismos da Medula Espinal , Humanos , Traumatismos da Medula Espinal/sangue , Traumatismos da Medula Espinal/diagnóstico , Traumatismos da Medula Espinal/imunologia , Biomarcadores/sangue , Índice de Gravidade de Doença , MicroRNAs/sangue , MicroRNAs/genética , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo
4.
Dev Neurobiol ; 84(3): 169-190, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38812372

RESUMO

Spinal cord injury (SCI) resulting from trauma decreases the quality of human life. Numerous clues indicate that the limited endogenous regenerative potential is a result of the interplay between the inhibitory nature of mature nervous tissue and the inflammatory actions of immune and glial cells. Knowledge gained from comparing regeneration in adult and juvenile animals could draw attention to factors that should be removed or added for effective therapy in adults. Therefore, we generated a minimal SCI (mSCI) model with a comparable impact on the spinal cord of Wistar rats during adulthood, preadolescence, and the neonatal period. The mechanism of injury is based on unilateral incision with a 20 ga needle tip according to stereotaxic coordinates into the dorsal horn of the L4 lumbar spinal segment. The incision should harm a similar amount of gray matter on a coronal section in each group of experimental animals. According to our results, the impact causes mild injury with minimal adverse effects on the neurological functions of animals but still has a remarkable effect on nervous tissue and its cellular and humoral components. Testing the mSCI model in adults, preadolescents, and neonates revealed a rather anti-inflammatory response of immune cells and astrocytes at the lesion site, as well as increased proliferation in the central canal lining in neonates compared with adult animals. Our results indicate that developing nervous tissue could possess superior reparative potential and confirm the importance of comparative studies to advance in the field of neuroregeneration.


Assuntos
Animais Recém-Nascidos , Proliferação de Células , Modelos Animais de Doenças , Ratos Wistar , Traumatismos da Medula Espinal , Animais , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Proliferação de Células/fisiologia , Ratos , Medula Espinal/patologia , Medula Espinal/imunologia , Astrócitos/patologia , Feminino
5.
Nat Immunol ; 25(6): 957-968, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38811815

RESUMO

The adult central nervous system (CNS) possesses a limited capacity for self-repair. Severed CNS axons typically fail to regrow. There is an unmet need for treatments designed to enhance neuronal viability, facilitate axon regeneration and ultimately restore lost neurological functions to individuals affected by traumatic CNS injury, multiple sclerosis, stroke and other neurological disorders. Here we demonstrate that both mouse and human bone marrow neutrophils, when polarized with a combination of recombinant interleukin-4 (IL-4) and granulocyte colony-stimulating factor (G-CSF), upregulate alternative activation markers and produce an array of growth factors, thereby gaining the capacity to promote neurite outgrowth. Moreover, adoptive transfer of IL-4/G-CSF-polarized bone marrow neutrophils into experimental models of CNS injury triggered substantial axon regeneration within the optic nerve and spinal cord. These findings have far-reaching implications for the future development of autologous myeloid cell-based therapies that may bring us closer to effective solutions for reversing CNS damage.


Assuntos
Axônios , Fator Estimulador de Colônias de Granulócitos , Interleucina-4 , Camundongos Endogâmicos C57BL , Regeneração Nervosa , Neutrófilos , Animais , Neutrófilos/imunologia , Regeneração Nervosa/imunologia , Camundongos , Humanos , Axônios/metabolismo , Axônios/fisiologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Interleucina-4/metabolismo , Ativação de Neutrófilo , Traumatismos da Medula Espinal/terapia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/metabolismo , Transferência Adotiva , Citocinas/metabolismo , Células Cultivadas
6.
Neurobiol Dis ; 195: 106500, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38614275

RESUMO

Spinal Cord Injury (SCI) disrupts critical autonomic pathways responsible for the regulation of the immune function. Consequently, individuals with SCI often exhibit a spectrum of immune dysfunctions ranging from the development of damaging pro-inflammatory responses to severe immunosuppression. Thus, it is imperative to gain a more comprehensive understanding of the extent and mechanisms through which SCI-induced autonomic dysfunction influences the immune response. In this review, we provide an overview of the anatomical organization and physiology of the autonomic nervous system (ANS), elucidating how SCI impacts its function, with a particular focus on lymphoid organs and immune activity. We highlight recent advances in understanding how intraspinal plasticity that follows SCI may contribute to aberrant autonomic activity in lymphoid organs. Additionally, we discuss how sympathetic mediators released by these neuron terminals affect immune cell function. Finally, we discuss emerging innovative technologies and potential clinical interventions targeting the ANS as a strategy to restore the normal regulation of the immune response in individuals with SCI.


Assuntos
Vias Autônomas , Traumatismos da Medula Espinal , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia , Humanos , Animais , Vias Autônomas/imunologia , Sistema Nervoso Autônomo/fisiopatologia , Sistema Nervoso Autônomo/imunologia
7.
Int Immunopharmacol ; 132: 111983, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38593504

RESUMO

Developing biomimetic nanoparticles without off-target side-effects remains a major challenge in spinal cord injury (SCI) immunotherapy. In this paper, we have conducted a drug carrier which is biocompatible macrophages-exocytosed exosome-biomimetic manganese (Mn)-iron prussian blue analogues (MPBs) for SCI immunotherapy. Exosome-sheathed MPBs (E-MPBs) exhibit promoted microglia accumulation, alleviation from H2O2-induced microenvironment and inhibition of apoptosis and inflammation in vitro. In addition, E-MPBs possessed significant tissue repair and neuroprotection in vivo. These properties endowed E-MPBs with great improvement in vivo in function recovery, resulting in anti-neuroinflammation activity and excellent biocompatibility in mice SCI model. As a promising treatment for efficient SCI immunotherapy, these results demonstrate the use of exosome-sheathed biomimetic nanoparticles exocytosed by anti-inflammation cells is feasible.


Assuntos
Exossomos , Imunoterapia , Macrófagos , Nanopartículas , Traumatismos da Medula Espinal , Animais , Exossomos/transplante , Exossomos/metabolismo , Traumatismos da Medula Espinal/terapia , Traumatismos da Medula Espinal/imunologia , Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Camundongos , Nanopartículas/química , Imunoterapia/métodos , Ferrocianetos/química , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Humanos , Microglia/imunologia , Células RAW 264.7 , Apoptose/efeitos dos fármacos
8.
Immun Inflamm Dis ; 12(4): e1256, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38652010

RESUMO

BACKGROUND: Spinal cord injury (SCI) is a traumatic neurological disorder with limited therapeutic options. Tumor protein p53-inducible nuclear protein 2 (TP53INP2) is involved in the occurrence and development of various diseases, and it may play a role during SCI via affecting inflammation and neuronal apoptosis. This study investigated the associated roles and mechanisms of TP53INP2 in SCI. METHODS: Mouse and lipopolysaccharide (LPS)-induced SCI BV-2 cell models were constructed to explore the role of TP53INP2 in SCI and the associated mechanisms. Histopathological evaluation of spinal cord tissue was detected by hematoxylin and eosin staining. The Basso, Beattie, and Bresnahan score was used to measure the motor function of the mice, while the spinal cord water content was used to assess spinal cord edema. The expression of TP53INP2 was measured using RT-qPCR. In addition, inflammatory factors in the spinal cord tissue of SCI mice and LPS-treated BV-2 cells were measured using enzyme-linked immunosorbent assay. Apoptosis and related protein expression levels were detected by flow cytometry and western blot analysis, respectively. RESULTS: TP53INP2 levels increased in SCI mice and LPS-treated BV-2 cells. The results of in vivo and in vitro experiments showed that TP53INP2 knockdown inhibited the inflammatory response and neuronal apoptosis in mouse spinal cord tissue or LPS-induced BV-2 cells. CONCLUSIONS: After spinal cord injury, TP53INP2 was upregulated, and TP53INP2 knockdown inhibited the inflammatory response and apoptosis.


Assuntos
Apoptose , Inflamação , Traumatismos da Medula Espinal , Animais , Masculino , Camundongos , Linhagem Celular , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Inflamação/patologia , Inflamação/metabolismo , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Medula Espinal/patologia , Medula Espinal/metabolismo , Medula Espinal/imunologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/genética
9.
Mol Neurobiol ; 61(8): 5525-5540, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38206470

RESUMO

Immune response is pivotal in the secondary injury of spinal cord injury (SCI). Polarization of macrophages (MΦ) influences the immune response in the secondary injury, which is regulated by several immune-related proteins. M2Φ plays the immunomodulatory role in the central nervous system. This study used bioinformatic analysis and machine algorithms to screen hub immune-related proteins after SCI and experimentally investigate the role of the target protein in the M2Φ polarization and immunomodulation in rats and in vitro after SCI. We downloaded GSE151371 and GSE45006, hub immune-related genes were screened using machine learning algorithms, and the expression of S100A9 was verified by datasets. Allen's weight-drop injury SCI model in Sprague-Dawley rat and bone marrow-derived rat MΦ with myelin debris model were used to study the effects of S100A9 on M2Φ polarization and immunomodulation at the lesion site and in vitro. Bioinformatic analysis showed that S100A9 acts as a hub immune-related gene in the SCI patients and rats. S100A9 increased at the lesion site in SCI rats, and its inhibition reduced CD206 and ARG-1 expression. Exogenous S100A9 promoted CD206 and ARG-1 expression in MΦ. S100A9 also increased the expression of PD-L1 and decreased MHC II at the lesion site in SCI rats and MΦ with myelin debris, and enhanced mitochondrial activity in rat MΦ with myelin debris. In conclusion, S100A9 is an indispensable factor in the immune process in secondary injury following SCI.


Assuntos
Calgranulina B , Polaridade Celular , Macrófagos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal , Animais , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/metabolismo , Macrófagos/metabolismo , Macrófagos/imunologia , Calgranulina B/metabolismo , Calgranulina B/genética , Polaridade Celular/efeitos dos fármacos , Ratos , Imunomodulação , Masculino , Humanos
10.
Front Immunol ; 13: 964138, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091018

RESUMO

Macrophages and microglia play important roles in chronic neuroinflammation following spinal cord injury (SCI). Although macrophages and microglia have similar functions, their phagocytic and homeostatic abilities differ. It is difficult to distinguish between these two populations in vivo, but single-cell analysis can improve our understanding of their identity and heterogeneity. We conducted bioinformatics analysis of the single-cell RNA sequencing dataset GSE159638, identifying apolipoprotein E (APOE) as a hub gene in both macrophages and microglia in the subacute and chronic phases of SCI. We then validated these transcriptomic changes in a mouse model of cervical spinal cord hemi-contusion and observed myelin uptake, lipid droplets, and lysosome accumulation in macrophages and microglia following SCI. Finally, we observed that knocking out APOE aggravated neurological dysfunction, increased neuroinflammation, and exacerbated the loss of white matter. Targeting APOE and the related cholesterol efflux represents a promising strategy for reducing neuroinflammation and promoting recovery following SCI.


Assuntos
Apolipoproteínas E , Macrófagos , Microglia , Doenças Neuroinflamatórias , Traumatismos da Medula Espinal , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/imunologia , Biologia Computacional , Macrófagos/imunologia , Camundongos , Microglia/imunologia , Doenças Neuroinflamatórias/genética , Doenças Neuroinflamatórias/imunologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/imunologia
11.
Brain Res Bull ; 180: 59-72, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34995751

RESUMO

After spinal cord injury (SCI), endogenous angiogenesis occurs in the injury core, unexpectedly accompanied by continuous leakage of the blood-spinal cord barrier (BSCB), which may be caused by destruction of the tight junctions (TJs) between vascular endothelial cells-an important structure of the BSCB. Blood-derived macrophages infiltrate into the spinal cord, aggregate to the injury core and then polarize toward M1/M2 phenotypes after SCI. However, the effect of macrophages with different polarizations on the TJs between vascular endothelial cells remains unclear. Here, we demonstrated that from 7 days postinjury (dpi) to 28 dpi, accompanied by the aggregation of macrophages, the expression of claudin-5 (CLN-5) and zonula occludens-1 (ZO-1) in vascular endothelial cells in the injury core was significantly decreased in comparison to that in normal spinal cord tissue and in the penumbra. Moreover, the leakage of the BSCB was severe in the injury core, as demonstrated by FITC-dextran perfusion. Notably, our study demonstrated that depletion of macrophages facilitated the restoration of TJs between vascular endothelial cells and decreased the leakage of BSCB in the injury core after SCI. Furthermore, we confirmed that the endothelial TJs could be impaired by M1 macrophages through secreting IL-6 in vitro, leading to an increased permeability of endothelial cells, but it was not significantly affected by M0 and M2 macrophages. These results indicated that the TJs between vascular endothelial cells were impaired by M1 macrophages in the injury core, potentially resulting in continuous leakage of the BSCB after SCI. Preventing M1 polarization of macrophages or blocking IL-6 in the injury core may promote restoration of the BSCB, thus accelerating functional recovery after SCI.


Assuntos
Células Endoteliais/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Traumatismos da Medula Espinal , Junções Íntimas/fisiologia , Animais , Modelos Animais de Doenças , Ratos , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia
12.
Sci Rep ; 12(1): 1459, 2022 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-35087114

RESUMO

Glycolysis is the metabolic pathway that converts glucose into pyruvate. Central nervous system (CNS) pathologies, such as spinal cord injury (SCI) and ischemia, are accompanied by an increase of the glycolytic pathway in the damaged areas as part of the inflammatory response. Pyruvate kinase is a key glycolytic enzyme that converts phosphoenolpyruvate and ADP to pyruvate and ATP. The protein has two isoforms, PKM1 and PKM2, originated from the same gene. As a homodimer, PKM2 loses the pyruvate kinase activity and acts as a transcription factor that regulates the expression of target genes involved in glycolysis and inflammation. After SCI, resident microglia and hematogenous macrophages are key inducers of the inflammatory response with deleterious effects. Activation of the bile acid receptor TGR5 inhibits the pro-inflammatory NFκB pathway in microglia and macrophages. In the present study we have investigated whether bile acids affect the expression of glycolytic enzymes and their regulation by PKM2. Bacterial lipopolysaccharide (LPS) induced the expression of PKM1, PKM2 and its target genes in primary cultures of microglial and Raw264.7 macrophage cells. SCI caused an increase of PKM2 immunoreactivity in macrophages after SCI. Pretreatment with tauroursodeoxycholic acid (TUDCA) or taurolithocholic acid (TLCA) reduced the expression of PKM2 and its target genes in cell cultures. Similarly, after SCI, TUDCA treatment reduced the expression of PKM2 in the lesion center. These results confirm the importance of PKM2 in the inflammatory response in CNS pathologies and indicate a new mechanism of bile acids as regulators of PKM2 pathway.


Assuntos
Ácidos e Sais Biliares/metabolismo , Microglia/patologia , Doenças Neuroinflamatórias/imunologia , Piruvato Quinase/metabolismo , Traumatismos da Medula Espinal/imunologia , Animais , Modelos Animais de Doenças , Glicólise , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Lipopolissacarídeos/imunologia , Macrófagos , Masculino , Camundongos , Microglia/imunologia , Doenças Neuroinflamatórias/patologia , Cultura Primária de Células , Piruvato Quinase/genética , Células RAW 264.7 , Ratos , Receptores Acoplados a Proteínas G/metabolismo , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/patologia
13.
Neurosci Lett ; 768: 136374, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34852285

RESUMO

Macrophage-dominated inflammation by the activation of Toll-like receptor (TLR) pathway leads to neurological disruption after spinal cord injury (SCI). Regulator of G-protein signaling 1 (RGS1) is reported to be a regulator in inflammation. The present study thus purposes to identify the unknown role of RGS1 mediating TLR on inflammation post SCI. A mouse model of traumatic SCI was established by a mechanical trauma at T10. The mice underwent SCI and a macrophage line activated by lipopolysaccharide (LPS) were treated with shRNA-RGS1 to elucidate the role of RGS1 in inflammatory progression. The inflammatory factors were measured, and the degree of histology and function protection were determined. The expression levels of RGS1, myeloid differentiation primary response protein 88 (Myd88), (TIR-domain-containing adaptor inducing interferon-ß (TRIF), p38, metalloproteinase (MMP)-2, and MMP-9 were determined. RGS1 was robustly increased both in LPS-activated macrophage and SCI mice. The TLR signaling pathway-induced inflammation was suppressed by RGS1 knockdown. shRNA-mediated silence of RGS1 was exhibited a prominent decrease in TNF-α, IL-1ß and IL-6 via TLR/TRIF/ nuclear factor kappa-B (NF-κB) axis. Depletion of RGS1 also inhibited MMP-induced tissue degradation via MAPK-p38 pathway in SCI mice. Moreover, suppression of RGS1 improved spinal cord histology and function recovery. These findings suggest that RGS1 regulates inflammation and tissue disruption via TLR/TRIF/NF-κB signaling pathway in mice with SCI, thereby explaining a novel target that regulates macrophage inflammation post SCI.


Assuntos
Inflamação/imunologia , Macrófagos/imunologia , Proteínas RGS/imunologia , Transdução de Sinais/imunologia , Traumatismos da Medula Espinal/imunologia , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/imunologia , NF-kappa B/metabolismo , Células RAW 264.7 , Proteínas RGS/metabolismo , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo
14.
Int J Neurosci ; 132(1): 1-12, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32672480

RESUMO

MATERIALS AND METHODS: Locomotor outcomes in perforin-deficient (Pfp-/-) mice and wild-type littermate controls were measured after severe compression injury of the lower thoracic spinal cord up to six weeks after injury. RESULTS: According to both the Basso mouse scale score and single frame motion analysis, motor recovery of Pfp-/- mice was similar to wild-type controls. Interestingly, immunohistochemical analysis of cell types at six weeks after injury showed enhanced cholinergic reinnervation of spinal motor neurons caudal to the lesion site and neurofilament-positive structures at the injury site in Pfp-/- mice, whereas numbers of microglia/macrophages and astrocytes were decreased compared with controls. CONCLUSIONS: We conclude that, although, loss of perforin does not change the locomotor outcome after injury, it beneficially affects diverse cellular features, such as number of axons, cholinergic synapses, astrocytes and microglia/macrophages at or caudal to the lesion site. Perforin's ability to contribute to Rag2's influence on locomotion was observed in mice doubly deficient in perforin and Rag2 which recovered better than Rag2-/- or Pfp-/- mice, suggesting that natural killer cells can cooperate with T- and B-cells in spinal cord injury.


Assuntos
Locomoção/fisiologia , Regeneração Nervosa/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Traumatismos da Medula Espinal/enzimologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Citotóxicas Formadoras de Poros/deficiência
15.
Exp Neurol ; 347: 113889, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34624330

RESUMO

Spinal cord injury (SCI) leads to irreversible functional deficits due to the disruption of axons and the death of neurons and glial cells. The inflammatory response that occurs in the injured spinal cord results in tissue degeneration; thus, targeting inflammation after acute SCI is expected to ameliorate histopathological evidence indicative of damage and, consequently, reduce functional disabilities. Interleukin 1 beta (IL-1ß) and interleukin 18 (IL-18) are pro-inflammatory cytokines members of the IL-1 family that initiate and propagate inflammation. Here, we report that protein levels of IL-1ß and IL-18 were increased in spinal cord parenchyma after SCI, but with different expression profiles. Whereas levels of IL-1ß were rapidly increased reaching peak levels at 12 h after the injury, levels of IL-18 did not increase until 7 days after the injury. Since activation of the NLRP3 inflammasome is required for the processing and release of IL-1ß and IL-18, we intraperitoneally administered OLT1177, a selective inhibitor of the NLRP3 inflammasome, to reduce the contribution of these cytokines to SCI. At a dose of 200 mg/kg, OLT1177 protected against neurological deficits and histological evidence of damage. OLT1177 also reduced the levels of IL-1ß in the spinal cord after contusion injury and diminished the accumulation of neutrophils and macrophages at later time points. These data suggest that targeting the NLRP3 inflammasome with OLT1177 could be a novel therapeutic strategy to arrest neuroinflammation and reduce functional impairments after acute SCI in humans.


Assuntos
Anti-Inflamatórios/farmacologia , Inflamassomos/efeitos dos fármacos , Bainha de Mielina/patologia , Nitrilas/farmacologia , Traumatismos da Medula Espinal/imunologia , Sulfonas/farmacologia , Animais , Feminino , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Doenças Neuroinflamatórias/imunologia , Doenças Neuroinflamatórias/patologia , Traumatismos da Medula Espinal/patologia
16.
Cells ; 10(11)2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34831436

RESUMO

Hundreds of thousands of people suffer spinal cord injuries each year. The experimental application of stem cells following spinal cord injury has opened a new era to promote neuroprotection and neuroregeneration of damaged tissue. Currently, there is great interest in the intravenous administration of the secretome produced by mesenchymal stem cells in acute or subacute spinal cord injuries. However, it is important to highlight that undifferentiated neural stem cells and induced pluripotent stem cells are able to adapt to the damaged environment and produce the so-called lesion-induced secretome. This review article focuses on current research related to the secretome and the lesion-induced secretome and their roles in modulating spinal cord injury symptoms and functional recovery, emphasizing different compositions of the lesion-induced secretome in various models of spinal cord injury.


Assuntos
Secretoma/metabolismo , Regeneração da Medula Espinal/fisiologia , Células-Tronco/metabolismo , Animais , Humanos , Imunomodulação , Traumatismos da Medula Espinal/epidemiologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco
17.
Theranostics ; 11(20): 9805-9820, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34815787

RESUMO

Background: Microglia and macrophages adopt a pro-inflammatory phenotype after spinal cord injury (SCI), what is thought to contribute to secondary tissue degeneration. We previously reported that this is due, in part, to the low levels of anti-inflammatory cytokines, such as IL-4. Since IL-13 and IL-4 share receptors and both cytokines drive microglia and macrophages towards an anti-inflammatory phenotype in vitro, here we studied whether administration of IL-13 and IL-4 after SCI leads to beneficial effects. Methods: We injected mice with recombinant IL-13 or IL-4 at 48 h after SCI and assessed their effects on microglia and macrophage phenotype and functional outcomes. We also performed RNA sequencing analysis of macrophages and microglia sorted from the injured spinal cords of mice treated with IL-13 or IL-4 and evaluated the metabolic state of these cells by using Seahorse technology. Results: We observed that IL-13 induced the expression of anti-inflammatory markers in microglia and macrophages after SCI but, in contrast to IL-4, it failed to mediate functional recovery. We found that these two cytokines induced different gene signatures in microglia and macrophages after SCI and that IL-4, in contrast to IL-13, shifted microglia and macrophage metabolism from glycolytic to oxidative phosphorylation. These findings were further confirmed by measuring the metabolic profile of these cells. Importantly, we also revealed that macrophages stimulated with IL-4 or IL-13 are not deleterious to neurons, but they become cytotoxic when oxidative metabolism is blocked. This suggests that the metabolic shift, from glycolysis to oxidative phosphorylation, is required to minimize the cytotoxic responses of microglia and macrophages. Conclusions: These results reveal that the metabolic fitness of microglia and macrophages after SCI contributes to secondary damage and that strategies aimed at boosting oxidative phosphorylation might be a novel approach to minimize the deleterious actions of microglia and macrophages in neurotrauma.


Assuntos
Interleucina-13/metabolismo , Interleucina-4/metabolismo , Traumatismos da Medula Espinal/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Interleucina-13/imunologia , Interleucina-13/farmacologia , Interleucina-4/imunologia , Interleucina-4/farmacologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Recuperação de Função Fisiológica/fisiologia , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia , Resultado do Tratamento
18.
Bioengineered ; 12(1): 8157-8172, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34663169

RESUMO

Bone mesenchymal stem cell-derived extracellular vesicles (BMSC-EVs) are known for recovery of injured tissues. We investigated the possible mechanism of BMSC-EVs in spinal cord injury (SCI). EVs were isolated from BMSCs and injected into SCI rats to evaluate the recovery of hindlimb motor function. The spinal cord tissue was stained after modeling to analyze spinal cord structure and inflammatory cell infiltration and detect microRNA (miR)-23b expression. The activity of lipopolysaccharide (LPS)-induced BV2 inflammatory cells was detected. The protein contents of interleukin (IL)-6, IL-1ß, IL-10 and tumor necrosis factor-α (TNF-α) in spinal cord and BV2 cells were measured. Western blot analysis was used to detect the level of toll-like receptor (TLR)4, p65, p-p65, iNOS, and Arg1 in spinal cord tissue and cells. TLR4 was overexpressed in rats and cells to evaluate the content of inflammatory cytokines. After EV treatment, the motor function of SCI rats was improved, SCI was relieved, and miR-23b expression was increased. After treatment with EV-miR-23b, iNOS, IL-6, IL-1ß, and TNF-α contents were decreased, while Arg1 and IL-10 were increased. The levels of TLR4 and p-p65 in spinal cord and BV2 cells were decreased. The rescue experiments verified that after overexpression of TLR4, the activity of BV2 cells was decreased, the contents of IL-6, IL-1ß, TNF-α, and p-p65 were increased, IL-10 was decreased, and SCI was aggravated. To conclude, The miR-23b delivered by BMSC-EVs targets TLR4 and inhibits the activation of NF-κB pathway, relieves the inflammatory response, so as to improve SCI in rats.


Assuntos
Vesículas Extracelulares/transplante , Lipopolissacarídeos/efeitos adversos , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Traumatismos da Medula Espinal/terapia , Receptor 4 Toll-Like/genética , Animais , Linhagem Celular , Modelos Animais de Doenças , Vesículas Extracelulares/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Traumatismos da Medula Espinal/etiologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/imunologia , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo
19.
Biomed Pharmacother ; 144: 112311, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34653754

RESUMO

Although accumulating evidence indicated that modulating macrophage polarization could ameliorate the immune microenvironment and facilitate the repair of spinal cord injury (SCI), the underlying mechanism of macrophage phenotypic switch is still poorly understood. Exosomes (Exos), a potential tool of cell-to-cell communication, may play important roles in cell reprogramming. Herein, we investigated the roles of macrophages-derived exosomes played for macrophage polarization in the SCI immune microenvironment. In this study, we found the fraction of M2 macrophages was markedly decreased after SCI. Moreover, the M2 macrophages-derived exosomes could increase the percentage of M2 macrophages, decrease that of M1 macrophages while the M1 macrophages-derived exosomes acted oppositely. According to the results of in silico analyses and molecular experiments verification, this phenotypic switch might be mediated by the exosomal miRNA-mRNA network, in which the miR-23a-3p/PTEN/PI3K/AKT axis might play an important role. In conclusion, our study suggests macrophage polarization that regulated by various interventions might be mediated by their own exosomes at last. Moreover, M2 macrophages-derived exosomes could promote M2 macrophage polarization via the potential miRNA-mRNA network. Considering its potential of modulating polarization, M2 macrophages-derived exosomes may be a promising therapeutic agent for SCI repair.


Assuntos
Plasticidade Celular , Microambiente Celular , Exossomos/metabolismo , Macrófagos/metabolismo , Traumatismos da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Animais , Microambiente Celular/imunologia , Modelos Animais de Doenças , Exossomos/imunologia , Exossomos/transplante , Macrófagos/imunologia , Macrófagos/transplante , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células RAW 264.7 , Ratos Wistar , Transdução de Sinais , Medula Espinal/imunologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/cirurgia
20.
Cells ; 10(10)2021 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-34685763

RESUMO

Aldynoglia are growth-promoting cells with a morphology similar to radial glia and share properties and markers with astrocytes and Schwann cells. They are distributed in several locations throughout the adult central nervous system, where the cells of the aldynoglia interact and respond to the signals of the immune cells. After spinal cord injury (SCI), the functions of resident aldynoglia, identified as ependymocytes, tanycytes, and ependymal stem cells (EpSCs) of the spinal cord are crucial for the regeneration of spinal neural tissue. These glial cells facilitate axonal regrowth and remyelination of injured axons. Here, we review the influence of M1 or M2 macrophage/microglia subpopulations on the fate of EpSCs during neuroinflammation and immune responses in the acute, subacute, and chronic phases after SCI.


Assuntos
Inflamação/imunologia , Inflamação/patologia , Neuroglia/patologia , Neurônios/imunologia , Neurônios/patologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/patologia , Animais , Humanos , Imunidade , Regeneração Nervosa , Traumatismos da Medula Espinal/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...