Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 448
Filtrar
1.
Yakugaku Zasshi ; 144(5): 497-501, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-38692923

RESUMO

Signal-transducing adaptor protein-2 (STAP-2) is a unique scaffold protein that regulates several immunological signaling pathways, including LIF/LIF receptor and LPS/TLR4 signals. STAP-2 is required for Fas/FasL-dependent T cell apoptosis and SDF-1α-induced T cell migration. Conversely, STAP-2 modulates integrin-mediated T cell adhesion, suggesting that STAP-2 is essential for several negative and positive T cell functions. However, whether STAP-2 is involved in T cell-antigen receptor (TCR)-mediated T cell activation is unknown. STAP-2 deficiency was recently reported to suppress TCR-mediated T cell activation by inhibiting LCK-mediated CD3ζ and ZAP-70 activation. Using STAP-2 deficient mice, it was demonstrated that STAP-2 is required for the pathogenesis of Propionibacterium acnes-induced granuloma formation and experimental autoimmune encephalomyelitis. Here, detailed functions of STAP-2 in TCR-mediated T cell activation, and how STAP-2 affects the pathogenesis of T cell-mediated inflammation and immune diseases, are reviewed.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Ativação Linfocitária , Receptores de Antígenos de Linfócitos T , Transdução de Sinais , Linfócitos T , Proteína-Tirosina Quinase ZAP-70 , Animais , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Humanos , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Camundongos , Proteína-Tirosina Quinase ZAP-70/metabolismo , Proteína-Tirosina Quinase ZAP-70/fisiologia , Propionibacterium acnes/fisiologia , Propionibacterium acnes/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/etiologia , Inflamação/imunologia , Apoptose , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/fisiologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Movimento Celular , Adesão Celular , Complexo CD3 , Quimiocina CXCL12/fisiologia , Quimiocina CXCL12/metabolismo
2.
Int J Mol Sci ; 23(3)2022 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-35163700

RESUMO

Neurogenesis is a physiological response after cerebral ischemic injury to possibly repair the damaged neural network. Therefore, promoting neurogenesis is very important for functional recovery after cerebral ischemic injury. Our previous research indicated that hyperbaric oxygen therapy (HBOT) exerted neuroprotective effects, such as reducing cerebral infarction volume. The purposes of this study were to further explore the effects of HBOT on the neurogenesis and the expressions of cell migration factors, including the stromal cell-derived factor 1 (SDF1) and its target receptor, the CXC chemokine receptor 4 (CXCR4). Thirty-two Sprague-Dawley rats were divided into the control or HBO group after receiving transient middle cerebral artery occlusion (MCAO). HBOT began to intervene 24 h after MCAO under the pressure of 3 atmospheres for one hour per day for 21 days. Rats in the control group were placed in the same acrylic box without HBOT during the experiment. After the final intervention, half of the rats in each group were cardio-perfused with ice-cold saline followed by 4% paraformaldehyde under anesthesia. The brains were removed, dehydrated and cut into serial 20µm coronal sections for immunofluorescence staining to detect the markers of newborn cell (BrdU+), mature neuron cell (NeuN+), SDF1, and CXCR4. The affected motor cortex of the other half rats in each group was separated under anesthesia and used to detect the expressions of brain-derived neurotrophic factor (BDNF), SDF1, and CXCR4. Motor function was tested by a ladder-climbing test before and after the experiment. HBOT significantly enhanced neurogenesis in the penumbra area and promoted the expressions of SDF1 and CXCR4. The numbers of BrdU+/SDF1+, BrdU+/CXCR4+, and BrdU+/NeuN+ cells and BDNF concentrations in the penumbra were all significantly increased in the HBO group when compared with the control group. The motor functions were improved in both groups, but there was a significant difference between groups in the post-test. Our results indicated that HBOT for 21 days enhanced neurogenesis and promoted cell migration toward the penumbra area in transient brain ischemic rats. HBOT also increased BDNF expression, which might further promote the reconstructions of the impaired neural networks and restore motor function.


Assuntos
Isquemia Encefálica/metabolismo , Movimento Celular , Quimiocina CXCL12/fisiologia , Oxigenoterapia Hiperbárica , Neurônios/metabolismo , Receptores CXCR4/fisiologia , Animais , Isquemia Encefálica/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo , Quimiocina CXCL12/genética , Regulação da Expressão Gênica , Masculino , Neurogênese , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores CXCR4/genética
3.
J Gene Med ; 23(9): e3366, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34032330

RESUMO

BACKGROUND: Stromal cell-derived factor-1 (SDF-1) plays an important role in the osteoblastic differentiation of human bone marrow mesenchymal stem cells (hBMMSCs), but the specific mechanism remains unclear. Our study aimed to clarify the role of the lncRNA-H19/miR-214-5p/BMP2 axis in the osteoblastic differentiation of hBMMSCs induced by SDF-1. METHODS: We used reverse-transcriptase polymerase chain reaction, western blotting, alkaline phosphatase activity test, and Alizarin red staining to evaluate the osteoblastic differentiation of primary hBMMSCs and the luciferase reporter assay to determine if lncRNA-H19 binds with miR-214-5p. RESULTS: Our results indicated that SDF-1 (50 ng/mL) promotes the osteoblastic differentiation of hBMMSCs, significantly upregulates osteoblastogenic genes (OCN, OSX, RUNX2, and ALP), and increases Alizarin red staining, alkaline phosphatase activity, and lncRNA-H19 expression. Luciferase reporter assay verified that lncRNA-H19 binds with and represses miR-214-5p, thereby upregulating BMP2 expression. Use of miR-214-5p inhibitor or overexpression of lncRNA-H19 can promote the osteoblastic differentiation of hBMMSCs, but miR-214-5p or shH19 inhibits the osteoblastic differentiation of hBMMSCs. Treatment with an miR-214-5p inhibitor could rescue the inhibitory effect of shH19 on the osteoblastic differentiation of hBMMSCs. CONCLUSIONS: Taken together, SDF-1 promotes the osteoblastic differentiation of hBMMSCs through the lncRNA-H19/miR-214-5p/BMP2 axis. Increased osteoblastic differentiation by an miR-214-5p inhibitor reveals a new possible strategy for the treatment of bone defect and osteoporosis.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Quimiocina CXCL12/fisiologia , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Osteogênese , RNA Longo não Codificante/metabolismo , Idoso , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Pessoa de Meia-Idade
4.
Med Oncol ; 38(5): 58, 2021 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-33847822

RESUMO

C-X-C chemokine receptor 7 (CXCR7), a novel receptor of C-X-C motif chemokine ligand 12 (CXCL12), is associated with the occurrence and metastasis of various malignant tumours. However, the role, function and underlying mechanisms of CXCR7 expression in cervical cancer remain undefined. The expression level of CXCR7 was evaluated in cervical cancer samples by immunohistochemistry and real-time PCR analyses. Western blot analysis was used to examine the expression level of CXCR7 in cervical cancer cell lines. HeLa cells were genetically silenced or pharmacologically inhibited for CXCR7 or CXCR4. Transwell and CCK-8 assays were used to examine cell migration and proliferation. The expression levels of MMP2, MMP9, TIMP-1 and TIMP-2 in HeLa cells were assessed by western blot or real-time PCR. HeLa cells silenced for CXCR7 were subcutaneously injected into nude mice to form tumours. The expression of CXCR7 in nude mice was investigated by immunohistochemical staining. Tumour volumes and weights were measured. The in vivo expression levels of MMP2, MMP9, TIMP-1 and TIMP-2 were determined by western blot analysis and real-time PCR. CXCR7 was overexpressed in cervical cancer tissues and cell lines. CXCL12 was highly expressed in cervical cancer lines. CXCR7 silencing or CCX733 treatment rather than CXCR4 silencing or AMD3100 treatment suppressed the proliferation, migration and invasion of cervical cancer cells stimulated by CXCL12. In a xenograft tumour model, CXCR7 silencing or CCX733 treatment inhibited the volumes and weights of xenograft tumours. In addition, downregulation of CXCR7 decreased the expression levels of MMP2 and MMP9 but increased the expression levels of TIMP-1 and TIMP-2 in vivo. These data support the finding that the downregulation of CXCR7 suppresses the proliferation and metastasis of cervical cancer cells. Inhibition of CXCR7 may be a potential targeted therapy for cervical cancer.


Assuntos
Carcinoma de Células Escamosas/patologia , Quimiocina CXCL12/fisiologia , Proteínas de Neoplasias/fisiologia , Receptores CXCR/fisiologia , Transdução de Sinais/fisiologia , Neoplasias do Colo do Útero/patologia , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Divisão Celular , Linhagem Celular , Linhagem Celular Tumoral , Colo do Útero/citologia , Células Epiteliais/metabolismo , Feminino , Humanos , Metaloproteinases da Matriz/biossíntese , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores CXCR/antagonistas & inibidores , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Receptores CXCR4/fisiologia , Neoplasias do Colo do Útero/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Endocrinology ; 162(7)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33693593

RESUMO

Vitamin D deficiency is associated with poor cancer outcome in humans, and administration of vitamin D or its analogs decreases tumor progression and metastasis in animal models. Using the mouse mammary tumor virus-polyoma middle T antigen (MMTV-PyMT) model of mammary cancer, we previously demonstrated a significant acceleration of carcinogenesis in animals on a low vitamin D diet and a reduction in spontaneous lung metastases when mice received vitamin D through perfusion. We investigate here the action mechanism for vitamin D in lung metastasis in the same non-immunodeficient model and demonstrate that it involves the control of epithelial to mesenchymal transition as well as interactions between chemokine C-X-C motif chemokine 12 (CXCL12) and its receptor C-X-C chemokine receptor type 4 (CXCR4). In vitro, 10-9M vitamin D treatment modifies the phenotype of MMTV-PyMT primary mammary tumor cells and significantly decreases their invasiveness and mammosphere formation capacity by 40% and 50%, respectively. Vitamin D treatment also inhibits phospho-signal transducer and activator of transcription 3 (p-STAT3), zinc finger E-box-binding homeobox 1 (Zeb1), and vimentin by 52%, 75%, and 77%, respectively, and increases E-cadherin by 87%. In vivo, dietary vitamin D deficiency maintains high levels of Zeb1 and p-STAT3 in cells from primary mammary tumors and increases CXCL12 expression in lung stroma by 64%. In lung metastases, vitamin D deficiency increases CXCL12/CXCR4 co-localization by a factor of 2.5. These findings indicate an involvement of vitamin D in mammary cancer "seed" (primary tumor cell) and "soil" (metastatic site) and link vitamin D deficiency to epithelial-to-mesenchymal transition (EMT), CXCL12/CXCR4 signaling, and accelerated metastasis, suggesting vitamin D repleteness in breast cancer patients could enhance the efficacy of co-administered therapies in preventing spread to distant organs.


Assuntos
Quimiocina CXCL12/análise , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/fisiopatologia , Receptores CXCR4/análise , Deficiência de Vitamina D/patologia , Animais , Linhagem Celular Tumoral , Quimiocina CXCL12/farmacologia , Quimiocina CXCL12/fisiologia , Feminino , Neoplasias Pulmonares/química , Neoplasias Mamárias Experimentais/patologia , Camundongos , Invasividade Neoplásica/fisiopatologia , Receptores CXCR4/fisiologia , Transdução de Sinais , Vitamina D/análogos & derivados , Vitamina D/farmacologia , Vitamina D/fisiologia , Deficiência de Vitamina D/fisiopatologia
6.
Blood ; 137(20): 2770-2784, 2021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-33512478

RESUMO

Dendritic cells (DCs) encompass several cell subsets that collaborate to initiate and regulate immune responses. Proper DC localization determines their function and requires the tightly controlled action of chemokine receptors. All DC subsets express CXCR4, but the genuine contribution of this receptor to their biology has been overlooked. We addressed this question using natural CXCR4 mutants resistant to CXCL12-induced desensitization and harboring a gain of function that cause the warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome (WS), a rare immunodeficiency associated with high susceptibility to the pathogenesis of human papillomavirus (HPV). We report a reduction in the number of circulating plasmacytoid DCs (pDCs) in WHIM patients, whereas that of conventional DCs is preserved. This pattern was reproduced in an original mouse model of WS, enabling us to show that the circulating pDC defect can be corrected upon CXCR4 blockade and that pDC differentiation and function are preserved, despite CXCR4 dysfunction. We further identified proper CXCR4 signaling as a critical checkpoint for Langerhans cell and DC migration from the skin to lymph nodes, with corollary alterations of their activation state and tissue inflammation in a model of HPV-induced dysplasia. Beyond providing new hypotheses to explain the susceptibility of WHIM patients to HPV pathogenesis, this study shows that proper CXCR4 signaling establishes a migration threshold that controls DC egress from CXCL12-containing environments and highlights the critical and subset-specific contribution of CXCR4 signal termination to DC biology.


Assuntos
Células Dendríticas/fisiologia , Inflamação/patologia , Doenças da Imunodeficiência Primária/fisiopatologia , Receptores CXCR4/fisiologia , Verrugas/fisiopatologia , Alphapapillomavirus/genética , Animais , Benzilaminas/farmacologia , Contagem de Células , Diferenciação Celular , Quimiocina CXCL12/fisiologia , Quimiotaxia , Ciclamos/farmacologia , Células Dendríticas/classificação , Epiderme/patologia , Feminino , Técnicas de Introdução de Genes , Genes Virais , Humanos , Inflamação/metabolismo , Células de Langerhans/fisiologia , Tecido Linfoide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Especificidade de Órgãos , Parabiose , Doenças da Imunodeficiência Primária/sangue , Doenças da Imunodeficiência Primária/genética , Doenças da Imunodeficiência Primária/patologia , Proteínas Recombinantes/metabolismo , Verrugas/sangue , Verrugas/genética , Verrugas/patologia
7.
Osteoarthritis Cartilage ; 29(3): 313-322, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33253889

RESUMO

Stromal cell-derived factor 1 (SDF-1), also known as CXC motif chemokine ligand 12 (CXCL12), is recognized as a homeostatic cytokine with strong chemotactic potency. It plays an important role in physiological and pathological processes, such as the development of multiple tissues and organs, the regulation of cell distribution, and tumour metastasis. SDF-1 has two receptors, CXC chemokine receptor type 4 (CXCR4) and CXC chemokine receptor type 7 (CXCR7). SDF-1 affects the proliferation, survival, differentiation and maturation of chondrocytes by binding to CXCR4 on chondrocytes. Therefore, SDF-1 has been used as an exogenous regulatory target in many studies to explore the mechanism of cartilage development. SDF-1 is also a potential therapeutic target for osteoarthritis (OA) and rheumatoid arthritis (RA), because of its role in pathological initiation and regulation. In addition, SDF-1 shows potent capacity in the repair of cartilage defects by recruiting endogenous stem cells in a cartilage tissue engineering context. To summarize the specific role of SDF-1 on cartilage development and disease, all articles had been screened out in PubMed by May 30, 2020. The search was limited to studies published in English. Search terms included SDF-1; CXCL12; CXCR4; chondrocyte; cartilage; OA; RA, and forty-seven papers were studied. Besides, we reviewed references in the articles we searched to get additional relevant backgrounds. The review aims to conclude the current knowledge regarding the physiological and pathological role of SDF-1 on the cartilage and chondrocyte. More investigations are required to determine methods targeted SDF-1 to cartilage development and interventions to cartilage diseases.


Assuntos
Artrite Reumatoide/metabolismo , Quimiocina CXCL12/metabolismo , Condrócitos/fisiologia , Condrogênese/fisiologia , Osteoartrite/metabolismo , Artrite Reumatoide/fisiopatologia , Doenças das Cartilagens/metabolismo , Doenças das Cartilagens/fisiopatologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular , Quimiocina CXCL12/fisiologia , Humanos , Osteoartrite/fisiopatologia , Receptores CXCR/fisiologia , Receptores CXCR4/fisiologia
8.
Mol Med Rep ; 23(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33215217

RESUMO

Stromal cell­derived factor­1 (SDF­1) is a chemokine involved in neuronal differentiation, as well as proliferation and migration. In the present study, the effects of recombinant SDF­1 on neurite outgrowth for nerve regeneration and engineering were evaluated in PC­12 cells. The effects of purified SDF­1 protein on cell toxicity, proliferation and migration were also assessed. SDF­1 significantly augmented cell proliferation in a dose­dependent manner, with low cytotoxicity in PC­12 cells. Cell migration also increased in the presence of SDF­1. SDF­1 significantly increased neurite number and length, compared with the control (untreated cells). Neurofilament mRNA levels, which are involved in neuronal differentiation, were also significantly upregulated in the presence of SDF­1. These results suggested that SDF­1 might prove useful for tissue engineering through induction of neuronal differentiation.


Assuntos
Quimiocina CXCL12/biossíntese , Quimiocina CXCL12/fisiologia , Neuritos/metabolismo , Crescimento Neuronal/fisiologia , Animais , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Quimiocina CXCL12/isolamento & purificação , Filamentos Intermediários/genética , Microscopia Confocal , Microscopia Eletrônica de Varredura , Células PC12 , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Cicatrização/fisiologia
9.
Expert Rev Hematol ; 13(12): 1387-1396, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33170753

RESUMO

INTRODUCTION: A variety of molecular-targeted drugs have been widely used in hematological malignancies and have shown great advances. Nevertheless, as the use of drugs in clinical practice increases, the problem of relapse or of the disease being refractory to treatment is becoming apparent. This problem is closely related to the C-X-C chemokine receptor 4 (CXCR4). AREAS COVERED: This review focuses mainly on the effect of CXCR4 on molecular-targeted drug resistance in hematological malignancies as well as the clinical efficacy of CXCR4 antagonists combined with molecular-targeted drugs. Relevant literatures published between 2006 and 2020 were searched using PubMed/Medline for this review. EXPERT OPINION: Monoclonal antibodies and non-antibody molecular-targeted drugs provide new therapeutic approaches for B-lineage malignancies and leukemia, but the clinical activity of these drugs is affected by CXCR4. In general, high CXCR4 expression or mutation inhibits the effects of molecular-targeted drugs, but there are exceptions, and in studies of proteasome inhibitors bortezomib (Bz) in multiple myeloma (MM), low CXCR4 expression or loss of CXCR4 was associated with Bz resistance (BzR) and poor treatment outcomes. Given that CXCR4 is a critical mediator of molecular-targeted drug resistance, numerous studies have combined molecular-targeted drugs with CXCR4 antagonists, which synergistically enhance the anti-proliferative/pro-apoptotic effect of molecular-targeted drugs.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias Hematológicas/tratamento farmacológico , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Receptores CXCR4/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Bortezomib/uso terapêutico , Linhagem da Célula , Quimiocina CXCL12/fisiologia , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Linfoma não Hodgkin/tratamento farmacológico , Linfoma não Hodgkin/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Inibidores de Proteases/uso terapêutico , Receptores CXCR4/genética , Receptores CXCR4/fisiologia , Transdução de Sinais/efeitos dos fármacos , Macroglobulinemia de Waldenstrom/tratamento farmacológico , Macroglobulinemia de Waldenstrom/metabolismo
10.
J Cell Mol Med ; 24(18): 10589-10603, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32761803

RESUMO

Low-energy shock wave (LESW) has been recognized as a promising non-invasive intervention to prevent the organs or tissues against ischaemia reperfusion injury (IRI), whereas its effect on kidney injury is rarely explored. To investigate the protective role of pretreatment with LESW on renal IRI in rats, animals were randomly divided into Sham, LESW, IRI and LESW + IRI groups. At 4, 12, 24 hours and 3 and 7 days after reperfusion, serum samples and renal tissues were harvested for performing the analysis of renal function, histopathology, immunohistochemistry, flow cytometry and Western blot, as well as enzyme-linked immunosorbent assay. Moreover, circulating endothelial progenitor cells (EPCs) were isolated, labelled with fluorescent dye and injected by tail vein. The fluorescent signals of EPCs were detected using fluorescence microscope and in vivo imaging system to track the distribution of injected circulating EPCs. Results showed that pretreatment with LESW could significantly reduce kidney injury biomarkers, tubular damage, and cell apoptosis, and promote cell proliferation and vascularization in IRI kidneys. The renoprotective role of LESW pretreatment would be attributed to the remarkably increased EPCs in the treated kidneys, part of which were recruited from circulation through SDF-1/CXCR7 pathway. In conclusion, pretreatment with LESW could increase the recruitment of circulating EPCs to attenuate and repair renal IRI.


Assuntos
Células Progenitoras Endoteliais/fisiologia , Tratamento por Ondas de Choque Extracorpóreas , Rim/irrigação sanguínea , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose , Movimento Celular , Quimiocina CXCL12/biossíntese , Quimiocina CXCL12/genética , Quimiocina CXCL12/fisiologia , Tratamento por Ondas de Choque Extracorpóreas/métodos , Corantes Fluorescentes/farmacocinética , Marcação In Situ das Extremidades Cortadas , Rim/patologia , Rim/fisiologia , Masculino , Microscopia de Fluorescência , Microvasos/patologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores CXCR/antagonistas & inibidores , Receptores CXCR/biossíntese , Receptores CXCR/genética , Receptores CXCR/fisiologia , Regeneração , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Fatores de Tempo
11.
Sci Rep ; 10(1): 5049, 2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-32193486

RESUMO

The present study shows that the CXCR4/SDF-1 axis regulates the migration of second branchial arch-derived muscles as well as non-somitic neck muscles. Cxcr4 is expressed by skeletal muscle progenitor cells in the second branchial arch (BA2). Muscles derived from the second branchial arch, but not from the first, fail to form in Cxcr4 mutants at embryonic days E13.5 and E14.5. Cxcr4 is also required for the development of non-somitic neck muscles. In Cxcr4 mutants, non-somitic neck muscle development is severely perturbed. In vivo experiments in chicken by means of loss-of-function approach based on the application of beads loaded with the CXCR4 inhibitor AMD3100 into the cranial paraxial mesoderm resulted in decreased expression of Tbx1 in the BA2. Furthermore, disrupting this chemokine signal at a later stage by implanting these beads into the BA2 caused a reduction in MyoR, Myf5 and MyoD expression. In contrast, gain-of-function experiments based on the implantation of SDF-1 beads into BA2 resulted in an attraction of myogenic progenitor cells, which was reflected in an expansion of the expression domain of these myogenic markers towards the SDF-1 source. Thus, Cxcr4 is required for the formation of the BA2 derived muscles and non-somitic neck muscles.


Assuntos
Quimiocina CXCL12/fisiologia , Face , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Pescoço , Receptores CXCR4/fisiologia , Células-Tronco/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Benzilaminas , Região Branquial/citologia , Região Branquial/embriologia , Região Branquial/metabolismo , Embrião de Galinha , Ciclamos , Expressão Gênica , Compostos Heterocíclicos/farmacologia , Mutação com Perda de Função , Camundongos , Mutação , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
12.
Br J Haematol ; 189(5): 815-825, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32135579

RESUMO

Acute myeloid leukaemia (AML) is the most common adult acute leukaemia with the lowest survival rate. It is characterised by a build-up of immature myeloid cells anchored in the protective niche of the bone marrow (BM) microenvironment. The CXCL12/CXCR4 axis is central to the pathogenesis of AML as it has fundamental control over AML cell adhesion into the protective BM niche, adaptation to the hypoxic environment, cellular migration and survival. High levels of CXCR4 expression are associated with poor relapse-free and overall survival. The CXCR4 ligand, CXCL12 (SDF-1), is expressed by multiple cells types in the BM, facilitating the adhesion and survival of the malignant clone. Blocking the CXCL12/CXCR4 axis is an attractive therapeutic strategy providing a 'multi-hit' therapy that both prevents essential survival signals and releases the AML cells from the BM into the circulation. Once out of the protective niche of the BM they would be more susceptible to destruction by conventional chemotherapeutic drugs. In this review, we disentangle the diverse roles of the CXCL12/CXCR4 axis in AML. We then describe multiple CXCR4 inhibitors, including small molecules, peptides, or monoclonal antibodies, which have been developed to date and their progress in pre-clinical and clinical trials. Finally, the review leads us to the conclusion that there is a need for further investigation into the development of a 'multi-hit' therapy that targets several signalling pathways related to AML cell adhesion and maintenance in the BM.


Assuntos
Quimiocina CXCL12/fisiologia , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/fisiologia , Receptores CXCR4/fisiologia , Transdução de Sinais/fisiologia , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/sangue , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Benzilaminas , Medula Óssea/patologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Hipóxia Celular , Movimento Celular/fisiologia , Micropartículas Derivadas de Células , Ensaios Clínicos como Assunto , Ciclamos , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/fisiologia , Compostos Heterocíclicos/farmacologia , Compostos Heterocíclicos/uso terapêutico , Humanos , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Camundongos , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Peptídeos/uso terapêutico , Peptídeos Cíclicos/uso terapêutico , Piridinas/uso terapêutico , Receptores CXCR4/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Nicho de Células-Tronco , Células Estromais/metabolismo , Células Estromais/patologia , Microambiente Tumoral
13.
Am J Respir Cell Mol Biol ; 62(6): 747-759, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32084325

RESUMO

Pulmonary artery smooth muscle cells (PASMCs) and pericytes are NG2+ mural cells that provide structural support to pulmonary arteries and capillaries. In pulmonary arterial hypertension (PAH), both mural cell types contribute to PA muscularization, but whether similar mechanisms are responsible for their behavior is unknown. RNA-seq was used to compare the gene profile of pericytes and PASMCs from PAH and healthy lungs. NG2-Cre-ER mice were used to generate NG2-selective reporter mice (NG2tdT) for cell lineage identification and tamoxifen-inducible mice for NG2-selective SDF1 knockout (SDF1NG2-KO). Hierarchical clustering of RNA-seq data demonstrated that the genetic profile of PAH pericytes and PASMCs is highly similar. Cellular lineage staining studies on NG2tdT mice in chronic hypoxia showed that, similar to PAH, tdT+ cells accumulate in muscularized microvessels and demonstrate significant upregulation of SDF1, a chemokine involved in chemotaxis and angiogenesis. Compared with control mice, SDF1NG2-KO mice in chronic hypoxia had reduced muscularization and lower abundance of NG2+ cells around microvessels. SDF1 stimulation in healthy pericytes induced greater contractility and impaired their capacity to establish endothelial-pericyte communications. In contrast, SDF1 knockdown reduced PAH pericyte contractility and improved their capacity to associate with vascular tubes in coculture. SDF1 is upregulated in NG2+ mural cells and is associated with PA muscularization. Targeting SDF1 could help prevent and/or reverse muscularization in PAH.


Assuntos
Quimiocina CXCL12/fisiologia , Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Miócitos de Músculo Liso/metabolismo , Pericitos/metabolismo , Animais , Antígenos/análise , Benzilaminas , Divisão Celular , Linhagem da Célula , Quimiocina CXCL12/genética , Doença Crônica , Ciclamos , DNA Nucleotidilexotransferase/análise , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Compostos Heterocíclicos/farmacologia , Compostos Heterocíclicos/uso terapêutico , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/prevenção & controle , Hipóxia/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Proteoglicanas/análise , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores CXCR4/antagonistas & inibidores , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Vasoconstrição
14.
Exp Cell Res ; 389(1): 111893, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32035133

RESUMO

Compared with noninvasive tumor cells, glioma cells overexpress chemokine receptor type 4 (CXCR4), which exhibits significantly greater expression in invasive tumor cells than in noninvasive tumor cells. C-X-C motif chemokine ligand 12 (CXCL12, also known as stromal derived factor-1, SDF-1) and its cell surface receptor CXCR4 activate a signaling axis that induces the expression of membrane type-2 matrix metalloproteinase (MT2-MMP), which plays a pivotal role in the invasion and migration of various cancer cells; however, the specific mechanism involved in this is unclear. Recently, studies have shown that invadopodia can recruit and secrete related enzymes, such as matrix metalloproteinases (MMPs), to degrade the surrounding extracellular matrix (ECM), promoting the invasion and migration of tumor cells. Phosphorylated cortactin (pY421-cortactin) is required for the formation and maturation of invadopodia, but the upstream regulatory factors and kinases involved in phosphorylation have not been elucidated. In this study, we found that CXCL12/CXCR4 was capable of inducing glioma cell invadopodia formation, probably by regulating cortactin phosphorylation. The interaction of cortactin and Arg (also known as Abl-related nonreceptor tyrosine kinase, ABL2) in glioma cells was demonstrated. The silencing of Arg inhibited glioma cell invadopodia formation and invasion by blocking cortactin phosphorylation. Moreover, CXCL12 could not induce glioma cell invasion in Arg-knockdown glioma cells. Based on these results, it can be concluded that Arg mediates CXCL12/CXCR4-induced glioma cell invasion, and CXCL12/CXCR4 regulates invadopodia maturation through the Arg-cortactin pathway, which indicates that Arg could be a candidate therapeutic target to inhibit glioma cell invasion.


Assuntos
Neoplasias Encefálicas/patologia , Quimiocina CXCL12/fisiologia , Glioma/patologia , Podossomos/fisiologia , Proteínas Tirosina Quinases/fisiologia , Receptores CXCR4/fisiologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Quimiocina CXCL12/metabolismo , Glioma/genética , Glioma/metabolismo , Humanos , Invasividade Neoplásica , Fosforilação , Podossomos/metabolismo , Processamento de Proteína Pós-Traducional , Receptores CXCR4/metabolismo , Transdução de Sinais/genética
15.
BMC Pregnancy Childbirth ; 20(1): 87, 2020 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-32041571

RESUMO

BACKGROUND: CXCL12(chemokine ligand 12, CXCL12) and its receptors CXCR4 are widely expressed in maternal-fetal interface and plays an adjust role in materno-fetal dialogue and immune tolerance during early pregnancy. This study aimed to evaluate the role and mechanism of self-derived CXCL12 in modulating the functions of human first-trimester endometrial epithelial cells (EECs) and to identify the potential protein kinase signaling pathways involved in the CXCL12/CXCR4's effect on EECs. METHODS: The expression of CXCL12 and CXCR4 in EECs was measured by using immunohistochemistry, immunofluorescence, real-time polymerase chain reaction and enzyme-linked immunosorbent assay. The effects of EEC-conditioned medium (EEC-CM) and recombinant human CXCL12 (rhCXCL12) on EEC migration and invasion in vitro were evaluated with migration and invasion assays. In-cell western blot analysis was used to examine the phosphorylation of protein kinase B (AKT), extracellular regulated protein kinases (ERKs) and phosphatidylinositol 3-kinase (PI3K) after CXCL12 treatment. RESULTS: CXCL12 and CXCR4 were both expressed in human first-trimester EECs at the mRNA and protein level. Both EEC-CM and rhCXCL12 significantly increased the migration and invasion of EECs (P < 0.05), which could be blocked by neutralizing antibodies against CXCR4 (P < 0.05) or CXCL12 (P < 0.05), respectively. CXCL12 activated both PI3K/AKT and ERK1/2 signaling and CXCR4 neutralizing antibody effectively reduced CXCL12-induced phosphorylation of AKT and ERK1/2. LY294002, a PI3K-AKT inhibitor, was able to reverse the promotive effect of EEC-CM or rhCXCL12 on EEC migration and invasion. CONCLUSIONS: Human first-trimester EECs promoted their own migration and invasion through the autocrine mechanism with CXCL12/CXCR4 axis involvement by activating PI3K/AKT signaling. This study contributes to a better understanding of the epithelium function mediated by chemokine and chemokine receptor during normal pregnancy.


Assuntos
Comunicação Autócrina/genética , Movimento Celular/genética , Quimiocina CXCL12/fisiologia , Endométrio/citologia , Receptores CXCR4/fisiologia , Técnicas de Cultura de Células , Células Epiteliais/fisiologia , Feminino , Humanos , Fosfatidilinositol 3-Quinase/fisiologia , Gravidez , Primeiro Trimestre da Gravidez/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/genética
16.
Ophthalmic Res ; 63(4): 392-403, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31935734

RESUMO

BACKGROUND: CCL2 (also known as monocyte chemoattractant protein 1) and CX3CR1 (also known as Fractalkine receptor)-deficient mice have damaged photoreceptors. OBJECTIVES: We examined the interaction of SDF1 and CXCR4 on the differentiation of retinal progenitors into rhodopsin-positive photoreceptors. METHODS: Cloned retinal progenitors were obtained by Pax6 gene transfection of mouse iPS cells followed by serial dilution. Clones were selected by expression of nestin, Musashi1, Six3, and Chx10 mRNA. Cell surface protein expression was analyzed by flow cytometry. The levels of mRNA and intracellular protein were examined by real-time PCR and immunochemistry, respectively. Transient transfection experiments of retinal progenitors were conducted using a human rhodopsin promoter luciferase plasmid. RESULTS: We selected 10 clones that expressed Six3, Chx10, Crx, Rx1, Nrl, CD73, and rhodopsin mRNA, which, except for rhodopsin, are photoreceptor precursor markers. Clones expressed both CD73 and CXCR4 on the cell surface and differentiated into rhodopsin-positive photoreceptors, which was reinforced by the addition of exogenous SDF1. A CXCR4 inhibitor AMD3100 blocked SDF1-mediated differentiation of progenitors into photoreceptors. SDF1 enhanced human rhodopsin promoter transcription activity, possibly via the NFκB pathway. Addition of SDF1 to the cell culture induced nuclear translocation of NFκB on retinal progenitor cell clones. Neonatal and newborn mouse retinas expressed SDF1 and CXCR4. Cells in the outer nuclear layer where photoreceptors are located expressed CXCR4 at P14 and P56. Cells in the inner nuclear layer expressed SDF1. CONCLUSIONS: These findings suggest that retinal progenitor cell differentiation was at least partly regulated by SDF1 and CXCR4 via upregulation of NFκB activity.


Assuntos
Diferenciação Celular/fisiologia , Quimiocina CXCL12/fisiologia , NF-kappa B/metabolismo , Fator de Transcrição PAX6/genética , Células Fotorreceptoras de Vertebrados/citologia , Receptores CXCR4/fisiologia , Animais , Animais Recém-Nascidos , Benzilaminas/farmacologia , Quimiocina CXCL12/farmacologia , Células Clonais , Ciclamos/farmacologia , Citometria de Fluxo , Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Células Fotorreceptoras de Vertebrados/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores CXCR4/antagonistas & inibidores , Rodopsina/genética , Transdução de Sinais/fisiologia , Transfecção , Regulação para Cima
17.
World Neurosurg ; 133: e165-e172, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31476465

RESUMO

BACKGROUND: Cartilaginous endplate (CEP), a thin layer of hyaline cartilage located between the vertebral endplate and nucleus pulposus, transports the nutrient into the disc. The objective of this study was to evaluate the influence of T140 (polyphemusin II-derived peptide) on the CEP cell growth, apoptosis, and the matrix formation via the stromal cell-derived factor-1 (SDF-1)/cysteine X cysteine (CXC) receptor-4 (CXCR4) signaling pathway. METHODS: Sprague-Dawley rats were euthanized by cervical dislocation and dissected for the isolation and the appraisal of CEP cells that were extracted from the endplate in rat intervertebral discs and were then added with different concentrations of reagents (SDF-1 and T140). The effect of T140 on CEP cell proliferation and apoptosis were analyzed. The messenger RNA (mRNA) and protein expressions of CXCR4, prominin-1, proteoglycans, type II collagen, B-cell lymphoma-2 (Bcl-2), and Bcl-2 associated X protein were analyzed by reverse transcription quantitative polymerase chain reaction and Western blot analysis. RESULTS: T140 promoted the proliferation of CEP cells and inhibited the apoptosis of CEP cells. Additionally, T140 suppressed the mRNA and protein expression of CXCR4, prominin-1, and Bcl-2 associated X protein, and increased the mRNA and protein expression of proteoglycans, type II collagen, and Bcl-2. CONCLUSIONS: T140 promotes the proliferation and matrix formation and inhibits the apoptosis of CEP cells by blocking the SDF-1/CXCR4 signaling pathway in vitro, which provides a certain therapeutic effect on the degeneration of intervertebral discs.


Assuntos
Apoptose/efeitos dos fármacos , Quimiocina CXCL12/fisiologia , Condrócitos/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Cartilagem Hialina/citologia , Disco Intervertebral/citologia , Oligopeptídeos/farmacologia , Receptores CXCR4/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Divisão Celular/efeitos dos fármacos , Proteínas da Matriz Extracelular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Disco Intervertebral/efeitos dos fármacos , Masculino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
18.
Am J Reprod Immunol ; 83(2)2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31650642

RESUMO

PROBLEM: Decidual natural killer (dNK) cells play key roles in maternal-fetal immune regulation, trophoblast invasion, and vascular remodeling, and most dNK cell populations are CD56bright CD16- NK cells. However, the enrichment and redistribution of dNK cells in the local decidua have not been clarified yet. METHOD OF STUDY: A total of 45 women with normal pregnancies and 8 unexplained recurrent spontaneous abortion (RSA) patients were included. We isolated primary human dNK (n = 53) and peripheral blood NK (pNK) cells (n = 5) from specimen and analyzed CD56, CD82, and CD29 by flow cytometry (FCM). We assessed their adhesion ability by cell counts of NK cells adhered to decidual stromal cells (DSCs) in a co-culture system. RESULTS: We found that RSA patients had more CD56dim dNK cells with lower CD82 and higher CD29 than women with normal pregnancies. There were negative correlations of CD82 to CD29 on CD56dim and CD56+ dNK cells. In normal pregnancies, dNK cells had lower CD82 and higher CD29 expression with a stronger adhesion ability than pNK cells. Blocking CD82 on dNK cells increased the adhesive ability and CD29 expression, while blocking CD29 decreased the adhesive ability. Co-culturing dNK cells with trophoblast cells decreased CD82 expression and increased the adhesive ability of dNK cells and the percentage of CD56bright NK cells, while blocking trophoblast-derived CXCL12 increased CD82 expression, decreased CD29 expression, and impaired the adhesive ability of NK cells. CONCLUSION: Trophoblast cells enhance the adhesive ability of NK cells to DSCs via the CXCL12/CD82/CD29 signaling pathway and contribute to CD56bright NK cell enrichment in the uterus.


Assuntos
Quimiocina CXCL12/fisiologia , Decídua/imunologia , Células Matadoras Naturais/citologia , Trofoblastos/metabolismo , Aborto Habitual/imunologia , Adulto , Antígeno CD56/análise , Adesão Celular , Células Cultivadas , Técnicas de Cocultura , Decídua/citologia , Feminino , Idade Gestacional , Humanos , Imunofenotipagem , Integrina beta1/análise , Proteína Kangai-1/análise , Células Matadoras Naturais/química , Células Matadoras Naturais/classificação , Células Matadoras Naturais/imunologia , Contagem de Linfócitos , Gravidez , Células Estromais/citologia
19.
Biomed Mater ; 15(1): 015009, 2019 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-31665702

RESUMO

Because the collagen membrane lacks osteoinductivity, it must be modified with bioactive components to trigger rapid bone regeneration. In this study, we aimed to evaluate the bone regeneration effects of a collagen membrane chemically conjugated with stromal cell-derived factor-1 alpha (SDF-1α) in rat models. To this end, different collagen membranes from four groups including a control group with a Bio-Oss bone substitute + collagen membrane; physical adsorption group with Bio-Oss + SDF-1α physically adsorbed on the collagen membrane; chemical cross-linking group with Bio-Oss + SDF-1α chemically cross-linked to the collagen membrane; and cell-seeding group with Bio-Oss + bone marrow mesenchymal stem cells (BMSCs) seeded onto the collagen membrane were placed in critical-sized defect models using a guided bone regeneration technique. At 4 and 8 weeks, the specimens were analyzed by scanning electron microscopy, energy-dispersive x-ray spectroscopy, micro-computed tomography, and histomorphology analyzes. Furthermore, ectopic osteogenesis was examined by histological analysis with Von Kossa staining, with the samples counterstained by hematoxylin and eosin and immunohistochemical staining. The results showed that in the chemical cross-linking group and cell-seeding group, the bone volume fraction, bone surface area fraction, and trabecular number were significantly increased and showed more new bone formation compared to the control and physical adsorption groups. Von Kossa-stained samples counterstained with hematoxylin and eosin and subjected to immunohistochemical staining of 4-week implanted membranes revealed that the chemical cross-linking group had the largest number of microvessels. The collagen membrane chemically conjugated with SDF-1α to significantly promote new bone and microvessel formation compared to SDF-1α physical adsorption and showed similar effects on new bone formation as a BMSC seeding method. This study provided a cell-free approach for shortening the bone healing time and improving the success rate of guided bone regeneration.


Assuntos
Materiais Biocompatíveis , Regeneração Óssea/fisiologia , Membranas Artificiais , Osteogênese/fisiologia , Animais , Materiais Biocompatíveis/química , Regeneração Óssea/efeitos dos fármacos , Transplante Ósseo , Quimiocina CXCL12/administração & dosagem , Quimiocina CXCL12/fisiologia , Colágeno/química , Masculino , Teste de Materiais , Transplante de Células-Tronco Mesenquimais , Modelos Animais , Osteogênese/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Microtomografia por Raio-X
20.
Skelet Muscle ; 9(1): 25, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31533830

RESUMO

BACKGROUND: During muscle regeneration, the chemokine CXCL12 (SDF-1) and the synthesis of some specific heparan sulfates (HS) have been shown to be critical. CXCL12 activity has been shown to be heavily influenced by its binding to extracellular glycosaminoglycans (GAG) by modulating its presentation to its receptors and by generating haptotactic gradients. Although CXCL12 has been implicated in several phases of tissue repair, the influence of GAG binding under HS influencing conditions such as acute tissue destruction remains understudied. METHODS: To investigate the role of the CXCL12/HS proteoglycan interactions in the pathophysiology of muscle regeneration, we performed two models of muscle injuries (notexin and freeze injury) in mutant CXCL12Gagtm/Gagtm mice, where the CXCL12 gene having been selectively mutated in critical binding sites of CXCL12 to interact with HS. Histological, cytometric, functional transcriptomic, and ultrastructure analysis focusing on the satellite cell behavior and the vessels were conducted on muscles before and after injuries. Unless specified, statistical analysis was performed with the Mann-Whitney test. RESULTS: We showed that despite normal histology of the resting muscle and normal muscle stem cell behavior in the mutant mice, endothelial cells displayed an increase in the angiogenic response in resting muscle despite the downregulated transcriptomic changes induced by the CXCL12 mutation. The regenerative capacity of the CXCL12-mutated mice was only delayed after a notexin injury, but a severe damage by freeze injury revealed a persistent defect in the muscle regeneration of CXCL12 mutant mice associated with vascular defect and fibroadipose deposition with persistent immune cell infiltration. CONCLUSION: The present study shows that CXCL12 is crucial for proper muscle regeneration. We highlight that this homing molecule could play an important role in drastic muscle injuries and that the regeneration defect could be due to an impairment of angiogenesis, associated with a long-lasting fibro-adipogenic scar.


Assuntos
Quimiocina CXCL12/genética , Quimiocina CXCL12/fisiologia , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/fisiopatologia , Neovascularização Fisiológica/genética , Regeneração/genética , Regeneração/fisiologia , Animais , Venenos Elapídicos/toxicidade , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Congelamento/efeitos adversos , Perfilação da Expressão Gênica , Proteoglicanas de Heparan Sulfato/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Músculo Esquelético/lesões , Regeneração/efeitos dos fármacos , Células Satélites de Músculo Esquelético/patologia , Células Satélites de Músculo Esquelético/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA