Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Cell Sci ; 134(18)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34471933

RESUMO

Duchenne muscular dystrophy is a genetic muscle disease characterized by chronic inflammation and fibrosis mediated by a pro-fibrotic macrophage population expressing pro-inflammatory markers. Our aim was to characterize cellular events leading to the alteration of macrophage properties and to modulate macrophage inflammatory status using the gaseous mediator hydrogen sulfide (H2S). Using co-culture experiments, we first showed that myofibers derived from mdx mice strongly skewed the polarization of resting macrophages towards a pro-inflammatory phenotype. Treatment of mdx mice with NaHS, an H2S donor, reduced the number of pro-inflammatory macrophages in skeletal muscle, which was associated with a decreased number of nuclei per fiber, as well as reduced myofiber branching and fibrosis. Finally, we established the metabolic sensor AMP-activated protein kinase (AMPK) as a critical NaHS target in muscle macrophages. These results identify an interplay between myofibers and macrophages where dystrophic myofibers contribute to the maintenance of a highly inflammatory environment sustaining a pro-inflammatory macrophage status, which in turn favors myofiber damage, myofiber branching and establishment of fibrosis. Our results also highlight the use of H2S donors as a potential therapeutic strategy to improve the dystrophic muscle phenotype by dampening chronic inflammation. This article has an associated First Person interview with the first author of the paper.


Assuntos
Distrofia Muscular de Duchenne , Animais , Fibrose , Macrófagos , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia
2.
EMBO J ; 36(13): 1946-1962, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28515121

RESUMO

Control of stem cell fate to either enter terminal differentiation versus returning to quiescence (self-renewal) is crucial for tissue repair. Here, we showed that AMP-activated protein kinase (AMPK), the master metabolic regulator of the cell, controls muscle stem cell (MuSC) self-renewal. AMPKα1-/- MuSCs displayed a high self-renewal rate, which impairs muscle regeneration. AMPKα1-/- MuSCs showed a Warburg-like switch of their metabolism to higher glycolysis. We identified lactate dehydrogenase (LDH) as a new functional target of AMPKα1. LDH, which is a non-limiting enzyme of glycolysis in differentiated cells, was tightly regulated in stem cells. In functional experiments, LDH overexpression phenocopied AMPKα1-/- phenotype, that is shifted MuSC metabolism toward glycolysis triggering their return to quiescence, while inhibition of LDH activity rescued AMPKα1-/- MuSC self-renewal. Finally, providing specific nutrients (galactose/glucose) to MuSCs directly controlled their fate through the AMPKα1/LDH pathway, emphasizing the importance of metabolism in stem cell fate.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Diferenciação Celular , Autorrenovação Celular , Homeostase , L-Lactato Desidrogenase/metabolismo , Músculos/citologia , Células-Tronco/metabolismo , Animais , Glicólise , Camundongos , Camundongos Knockout
3.
J Cell Sci ; 126(Pt 3): 756-66, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23264736

RESUMO

ACAPs and ASAPs are Arf-GTPase-activating proteins with BAR, PH, GAP and ankyrin repeat domains and are known to regulate vesicular traffic and actin cytoskeleton dynamics in mammalian cells. The amoeba Dictyostelium has only two proteins with this domain organization, instead of the six in human, enabling a more precise functional analysis. Genetic invalidation of acapA resulted in multinucleated cells with cytokinesis defects. Mutant acapA(-) cells were hardly motile and their multicellular development was significantly delayed. In addition, formation of filopodial protrusions was deficient in these cells. Conversely, re-expression of ACAP-A-GFP resulted in numerous and long filopodia-like protrusions. Mutagenesis studies showed that the ACAP-A actin remodeling function was dependent on its ability to activate its substrate, the small GTPase ArfA. Likewise, the expression of a constitutively active ArfA•GTP mutant in wild-type cells led to a significant reduction in filopodia length. Together, our data support a role for ACAP-A in the control of the actin cytoskeleton organization and dynamics through an ArfA-dependent mechanism.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Dictyostelium/fisiologia , Proteínas Ativadoras de GTPase/metabolismo , Pseudópodes/metabolismo , Animais , Movimento Celular/genética , Citocinese/genética , Dictyostelium/enzimologia , Proteínas Ativadoras de GTPase/genética , Humanos , Mutação/genética , Pseudópodes/patologia , RNA Interferente Pequeno/genética , Transgenes/genética
4.
Int J Cancer ; 133(7): 1589-602, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23553037

RESUMO

Acquisition of resistance to aromatase inhibitors (AIs) remains a major drawback in the treatment of estrogen receptor alpha (ERα)-positive breast cancers. The Res-Ana cells, a new model of acquired resistance to anastrozole, were established by long-term exposure of aromatase-overexpressing MCF-7 cells to this drug. These resistant cells developed ER-independent mechanisms of resistance and decreased sensitivity to the AI letrozole or to ERα antagonists. They also displayed a constitutive activation of the PI3K/Akt/mTOR pathway and a deregulated expression of several ErbB receptors. An observed increase in the phospho-Akt/Akt ratio between primary and matched recurrent breast tumors of patients who relapsed under anastrozole adjuvant therapy also argued for a pivotal role of the Akt pathway in acquired resistance to anastrozole. Ectopic overexpression of constitutively active Akt1 in control cells was sufficient to induce de novo resistance to anastrozole. Strikingly, combining anastrozole with the highly selective and allosteric Akt inhibitor MK-2206 or with the mTOR inhibitor rapamycin increased sensitivity to this AI in the control cells and was sufficient to overcome resistance and restore sensitivity to endocrine therapy in the resistant cells. Our findings lead to us proposing a model of anastrozole-acquired resistance based on the selection of cancer-initiating-like cells possessing self-renewing properties, intrinsic resistance to anastrozole and sensitivity to MK-2206. Altogether, our work demonstrated that the Akt/mTOR pathway plays a key role in resistance to anastrozole and that combining anastrozole with Akt/mTOR pathway inhibitors represents a promising strategy in the clinical management of hormone-dependent breast cancer patients.


Assuntos
Inibidores da Aromatase/farmacologia , Neoplasias da Mama/tratamento farmacológico , Compostos Heterocíclicos com 3 Anéis/farmacologia , Nitrilas/farmacologia , Triazóis/farmacologia , Anastrozol , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/biossíntese , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Células MCF-7 , Recidiva Local de Neoplasia/metabolismo , Nitrilas/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-2/biossíntese , Receptor ErbB-3/biossíntese , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Triazóis/uso terapêutico
5.
iScience ; 26(12): 108343, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38077152

RESUMO

Due to the post-mitotic nature of skeletal muscle fibers, adult muscle maintenance relies on dedicated muscle stem cells (MuSCs). In most physiological contexts, MuSCs support myofiber homeostasis by contributing to myonuclear accretion, which requires a coordination of cell-type specific events between the myofiber and MuSCs. Here, we addressed the role of the kinase AMPKα2 in the coordination of these events supporting myonuclear accretion. We demonstrate that AMPKα2 deletion impairs skeletal muscle regeneration. Through in vitro assessments of MuSC myogenic fate and EdU-based cell tracing, we reveal a MuSC-specific role of AMPKα2 in the regulation of myonuclear accretion, which is mediated by phosphorylation of the non-metabolic substrate BAIAP2. Similar cell tracing in vivo shows that AMPKα2 knockout mice have a lower rate of myonuclear accretion during regeneration, and that MuSC-specific AMPKα2 deletion decreases myonuclear accretion in response to myofiber contraction. Together, this demonstrates that AMPKα2 is a MuSC-intrinsic regulator of myonuclear accretion.

6.
J Neuromuscul Dis ; 8(4): 513-524, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33843691

RESUMO

BACKGROUND: The mdx-C57/B6 mouse model does not show the clinical signs of Duchenne muscular dystrophy (DMD), although muscles exhibit hallmarks of permanent regeneration and alterations in muscle function. The DMDmdx4Cv strain exhibits very few revertant dystrophin positive myofibers, making that model suitable for studies on gene and cell therapies. OBJECTIVE: The study appraises the histological evolution of the Tibialis Anterior muscle of WT and DMD mdx4Cv mutant from 1 to 24 months. METHODS: Histological analysis included a series of immunostainings of muscle sections for assessing tissue features (fibrosis, lipid deposition, necrosis) and cellular characteristics (size of myofibers, number and distribution of myonuclei, number of satellite cells, vessels, macrophages). RESULTS: None of the investigated cell types (satellite cells, endothelial cells, macrophages) showed variations in their density within the tissue in both WT and DMD mdx4Cv muscle. However, analyzing their number per myofiber showed that in DMD mdx4Cv, myofiber capillarization was increased from 1 to 6 months as compared with WT muscle, then dropped from 12 months. Macrophage number did not vary in WT muscle and peaked at 6 months in DMD mdx4Cv muscle. The number of satellite cells per myofiber did not vary in WT muscle while it remained high in DMD mdx4Cv muscle, starting to decrease from 12 months and being significantly lower at 24 months of age. Myofiber size was not different in DMD mdx4Cv from WT except at 24 months, when it strongly decreased in DMD mdx4Cv muscle. Necrosis and lipid deposition were rare in DMD mdx4Cv muscle. Fibrosis did not increase with age in DMD mdx4Cv muscle and was higher than in WT at 6 and 12 months of age. CONCLUSIONS: As a whole, the results show a strong decrease of the myofiber size at 24 months, and an increased capillarization until 6 months of age in DMD mdx4Cv as compared with the WT. Thus, DMD mdx4Cv mice poorly recapitulates histological DMD features, and its use should take into account the age of the animals according to the purpose of the investigation.


Assuntos
Músculo Esquelético/patologia , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/patologia , Animais , Modelos Animais de Doenças , Células Endoteliais/patologia , Fibrose/patologia , Camundongos , Camundongos Endogâmicos mdx
7.
Cell Metab ; 33(2): 283-299.e9, 2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33400911

RESUMO

Platelets are known to enhance the wound-healing activity of mesenchymal stem cells (MSCs). However, the mechanism by which platelets improve the therapeutic potential of MSCs has not been elucidated. Here, we provide evidence that, upon their activation, platelets transfer respiratory-competent mitochondria to MSCs primarily via dynamin-dependent clathrin-mediated endocytosis. We found that this process enhances the therapeutic efficacy of MSCs following their engraftment in several mouse models of tissue injury, including full-thickness cutaneous wound and dystrophic skeletal muscle. By combining in vitro and in vivo experiments, we demonstrate that platelet-derived mitochondria promote the pro-angiogenic activity of MSCs via their metabolic remodeling. Notably, we show that activation of the de novo fatty acid synthesis pathway is required for increased secretion of pro-angiogenic factors by platelet-preconditioned MSCs. These results reveal a new mechanism by which platelets potentiate MSC properties and underline the importance of testing platelet mitochondria quality prior to their clinical use.


Assuntos
Plaquetas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Mitocôndrias/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cicatrização
8.
Mol Cancer ; 9: 291, 2010 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-21059223

RESUMO

BACKGROUND: ZNF217 is a candidate oncogene located at 20q13, a chromosomal region frequently amplified in breast cancers. The precise mechanisms involved in ZNF217 pro-survival function are currently unknown, and utmost importance is given to deciphering the role of ZNF217 in cancer therapy response. RESULTS: We provide evidence that stable overexpression of ZNF217 in MDA-MB-231 breast cancer cells conferred resistance to paclitaxel, stimulated cell proliferation in vitro associated with aberrant expression of several cyclins, and increased tumor growth in mouse xenograft models. Conversely, siRNA-mediated silencing of ZNF217 expression in MCF7 breast cancer cells, which possess high endogenous levels of ZNF217, led to decreased cell proliferation and increased sensitivity to paclitaxel. The paclitaxel resistance developed by ZNF217-overexpressing MDA-MB-231 cells was not mediated by the ABCB1/PgP transporter. However, ZNF217 was able to counteract the apoptotic signals mediated by paclitaxel as a consequence of alterations in the intrinsic apoptotic pathway through constitutive deregulation of the balance of Bcl-2 family proteins. Interestingly, ZNF217 expression levels were correlated with the oncogenic kinase Aurora-A expression levels, as ZNF217 overexpression led to increased expression of the Aurora-A protein, whereas ZNF217 silencing was associated with low Aurora-A expression levels. We showed that a potent Aurora-A kinase inhibitor was able to reverse paclitaxel resistance in the ZNF217-overexpressing cells. CONCLUSION: Altogether, these data suggest that ZNF217 might play an important role in breast neoplastic progression and chemoresistance, and that Aurora-A might be involved in ZNF217-mediated effects.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Paclitaxel/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/metabolismo , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Aurora Quinase A , Aurora Quinases , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Citometria de Fluxo , Humanos , Camundongos , Camundongos Nus , Paclitaxel/uso terapêutico , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética
9.
Int J Cancer ; 126(2): 545-62, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19609946

RESUMO

Endocrine therapy resistance is one of the main challenges in the treatment of estrogen receptor positive (ER+) breast cancer patients. This study showed that two ER+ human breast carcinoma cell lines derived from MCF-7 (MVLN cells) that have acquired under OH-Tamoxifen selection two distinct phenotypes of endocrine resistance both displayed constitutive activation of the PI3K/Akt and MAPK pathways. Aberrant expression and activation of the ErbB system (phospho-EGFR, phospho-ErbB2, phospho-ErbB3, over-expression of ErbB4 and over-expression of several ErbB ligands) were also observed in the two resistant cell lines, suggesting the existence of an autocrine loop leading to constitutive activation of MAPK and PI3K/Akt survival pathways. The recent clinical use of specific signal transduction inhibitors is one of the most promising therapeutic approaches in breast cancers. The MEK inhibitor PD98059 and the PI3K inhibitor LY294002 were both able to enhance the cytostatic effect of OH-Tamoxifen or fulvestrant on MVLN sensitive cells. In the two resistant cell lines, inhibition of the MAPK or the PI3K/Akt pathways associated with endocrine therapy was sufficient to reverse OH-Tamoxifen or fulvestrant resistance. Investigating the effect of a combination of both inhibitors on the reversion of OH-Tamoxifen and fulvestrant resistance in the two resistant cell lines suggested that, in clinical practice, a strategy combining the two inhibitors would be the best approach to target the different endocrine resistance phenotypes possibly present in a tumor. In conclusion, the combination of MAPK and PI3K inhibitors represents a promising strategy to overcome endocrine therapy resistance in ER+ breast cancer patients.


Assuntos
Cromonas/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Flavonoides/farmacologia , Morfolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/farmacologia , Antineoplásicos Hormonais/farmacologia , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Receptor ErbB-4 , Receptores de Estrogênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Tissue Eng Part A ; 25(23-24): 1667-1676, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31062647

RESUMO

Although skeletal muscle is capable of complete recovery after an injury, specific situations require support or acceleration of this process, such as in the elderly and athletes, respectively. Skeletal muscle regeneration is due to muscle stem cells (MuSCs) that undergo adult myogenesis, a process sustained by MuSC environment. Although recognized as important, extracellular matrix (ECM) has been overlooked in this process. Matrix-based therapy aims at improving ECM remodeling to support tissue repair. In this context, we investigated the properties of a single injection of the clinical grade glycosaminoglycan mimetics RGTA® (ReGeneraTing Agents) on skeletal muscle regeneration in a context compatible with a clinical application, that is, 3 days after the injury. Our results show that RGTA-treated muscles showed an increase of the number of myonuclei in regenerating myofibers and an increase of the capillarization of the new myofibers. In vitro experiments showed that RGTA directly acts on MuSCs by stimulating their fusion into myotubes and on endothelial cells by stimulating the formation and maturation of vessels in a 3D culture setup. These results indicate that a single administration of RGTA in regenerating muscle stimulated both myogenesis and angiogenesis, thus accelerating skeletal muscle regeneration. Impact Statement Although highly powerful in normal condition, postinjury skeletal muscle regeneration is less efficient in some situations, such as obese, elderly, or resting people. In other context, such as high-performance sport, skeletal muscle regeneration must be shortened but in a way ensuring a full functional recovery. In this context, our results show that a single injection of the clinical grade glycosaminoglycan mimetics RGTA® (ReGeneraTing Agents), in a context compatible with a clinical application, that is, 3 days after the injury, is beneficial for skeletal muscle regeneration, through the stimulation of both myogenesis and angiogenesis.


Assuntos
Materiais Biomiméticos/farmacologia , Heparitina Sulfato/farmacologia , Músculo Esquelético/lesões , Músculo Esquelético/fisiopatologia , Regeneração/efeitos dos fármacos , Animais , Fusão Celular , Células Endoteliais/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Células Musculares/efeitos dos fármacos , Desenvolvimento Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos
11.
Methods Mol Biol ; 1732: 539-549, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29480498

RESUMO

In this chapter, we describe the methods to isolate and culture muscle stem cells (MuSCs) from murine skeletal muscle in order to decipher the intrinsic effect of AMP-activated kinase activity on MuSC fate. Culture of MuSCs is a powerful model to recapitulate every step of stem cell behavior observed in vivo: activation, proliferation, differentiation, fusion and also self-renewal. We provide the detailed procedures to isolate pure MuSCs by a flow cytometry-based method using the selection of a combination of specific markers and to characterize MuSC fate (quiescence, activation, and differentiation) in response to AMPK activity modulation by assessing of the expression of stem cell (e.g., Pax7) and myogenic marker (e.g., MyoD).


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Ativação Enzimática/fisiologia , Desenvolvimento Muscular/fisiologia , Mioblastos/fisiologia , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Separação Celular/instrumentação , Separação Celular/métodos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Antígeno Ki-67/análise , Antígeno Ki-67/metabolismo , Camundongos , Desenvolvimento Muscular/efeitos dos fármacos , Músculo Esquelético/citologia , Proteína MyoD/análise , Proteína MyoD/metabolismo , Fator de Transcrição PAX7/análise , Fator de Transcrição PAX7/metabolismo , Cultura Primária de Células/instrumentação , Cultura Primária de Células/métodos
12.
Cell Rep ; 25(8): 2163-2176.e6, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30463013

RESUMO

Chronic inflammation and fibrosis characterize Duchenne muscular dystrophy (DMD). We show that pro-inflammatory macrophages are associated with fibrosis in mouse and human DMD muscle. DMD-derived Ly6Cpos macrophages exhibit a profibrotic activity by sustaining fibroblast production of collagen I. This is mediated by the high production of latent-TGF-ß1 due to the higher expression of LTBP4, for which polymorphisms are associated with the progression of fibrosis in DMD patients. Skewing macrophage phenotype via AMPK activation decreases ltbp4 expression by Ly6Cpos macrophages, blunts the production of latent-TGF-ß1, and eventually reduces fibrosis and improves DMD muscle force. Moreover, fibro-adipogenic progenitors are the main providers of TGF-ß-activating enzymes in mouse and human DMD, leading to collagen production by fibroblasts. In vivo pharmacological inhibition of TGF-ß-activating enzymes improves the dystrophic phenotype. Thus, an AMPK-LTBP4 axis in inflammatory macrophages controls the production of TGF-ß1, which is further activated by and acts on fibroblastic cells, leading to fibrosis in DMD.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas de Ligação a TGF-beta Latente/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Biomarcadores/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Fibroblastos/metabolismo , Fibrose , Inflamação/patologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Células NIH 3T3
13.
J Exp Med ; 215(1): 303-318, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29203538

RESUMO

Inflammation and tissue regeneration follow tissue damage, but little is known about how these processes are coordinated. High Mobility Group Box 1 (HMGB1) is a nuclear protein that, when released on injury, triggers inflammation. We previously showed that HMGB1 with reduced cysteines is a chemoattractant, whereas a disulfide bond makes it a proinflammatory cytokine. Here we report that fully reduced HMGB1 orchestrates muscle and liver regeneration via CXCR4, whereas disulfide HMGB1 and its receptors TLR4/MD-2 and RAGE (receptor for advanced glycation end products) are not involved. Injection of HMGB1 accelerates tissue repair by acting on resident muscle stem cells, hepatocytes, and infiltrating cells. The nonoxidizable HMGB1 mutant 3S, in which serines replace cysteines, promotes muscle and liver regeneration more efficiently than the wild-type protein and without exacerbating inflammation by selectively interacting with CXCR4. Overall, our results show that the reduced form of HMGB1 coordinates tissue regeneration and suggest that 3S may be used to safely accelerate healing after injury in diverse clinical contexts.


Assuntos
Proteína HMGB1/metabolismo , Regeneração Hepática/fisiologia , Músculos/metabolismo , Músculos/fisiologia , Receptores CXCR4/metabolismo , Animais , Linhagem Celular , Fatores Quimiotáticos/metabolismo , Citocinas/metabolismo , Células HEK293 , Hepatócitos/metabolismo , Hepatócitos/fisiologia , Humanos , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Cicatrização/fisiologia
14.
Stem Cell Reports ; 9(6): 2018-2033, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29198825

RESUMO

In skeletal muscle, new functions for vessels have recently emerged beyond oxygen and nutrient supply, through the interactions that vascular cells establish with muscle stem cells. Here, we demonstrate in human and mouse that endothelial cells (ECs) and myogenic progenitor cells (MPCs) interacted together to couple myogenesis and angiogenesis in vitro and in vivo during skeletal muscle regeneration. Kinetics of gene expression of ECs and MPCs sorted at different time points of regeneration identified three effectors secreted by both ECs and MPCs. Apelin, Oncostatin M, and Periostin were shown to control myogenesis/angiogenesis coupling in vitro and to be required for myogenesis and vessel formation during muscle regeneration in vivo. Furthermore, restorative macrophages, which have been previously shown to support myogenesis in vivo, were shown in a 3D triculture model to stimulate myogenesis/angiogenesis coupling, notably through Oncostatin M production. Our data demonstrate that restorative macrophages orchestrate muscle regeneration by controlling myogenesis/angiogenesis coupling.


Assuntos
Diferenciação Celular/genética , Desenvolvimento Muscular/genética , Músculo Esquelético/crescimento & desenvolvimento , Neovascularização Fisiológica/genética , Regeneração/genética , Animais , Apelina/genética , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Moléculas de Adesão Celular/genética , Movimento Celular/genética , Células Progenitoras Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Macrófagos/metabolismo , Camundongos , Mioblastos/citologia , Mioblastos/metabolismo , Oncostatina M/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Cicatrização
16.
Int J Antimicrob Agents ; 39(5): 444-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22445492

RESUMO

This study provides insight into the antibacterial activity of the cytotoxic nucleoside analogue gemcitabine against clinical multiresistant Staphylococcus aureus strains. Classical methods were used for determination of the minimum inhibitory concentration (MIC) and synergy in vitro, and polymerase chain reaction (PCR) products were sequenced to search for mutations in nucleoside kinase genes in resistant strains. Gemcitabine and its derivative CP-4126 were effective against meticillin-susceptible S. aureus (MSSA), meticillin-resistant S. aureus (MRSA) and glycopeptide-intermediate S. aureus (GISA) isolates, with MICs ranging between 0.06 mg/L and 4.22 mg/L. Bactericidal activity was shown in time-kill studies as well as synergy with gentamicin. Mutations in the nucleoside kinase gene SadAK were observed in resistant strains, indicating a role for this enzyme in gemcitabine activity. Nucleoside analogues have antimicrobial activity and these results could be used for further identification and development of new antibiotics.


Assuntos
Antibacterianos/farmacologia , Desoxicitidina/análogos & derivados , Farmacorresistência Bacteriana Múltipla , Sinergismo Farmacológico , Gentamicinas/farmacologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Desoxicitidina/farmacologia , Humanos , Testes de Sensibilidade Microbiana , Staphylococcus aureus/isolamento & purificação , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA