Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Nat Commun ; 14(1): 5529, 2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37684243

RESUMO

Immune checkpoint inhibitors cause side effects ranging from autoimmune endocrine disorders to severe cardiotoxicity. Periodic Fasting mimicking diet (FMD) cycles are emerging as promising enhancers of a wide range of cancer therapies including immunotherapy. Here, either FMD cycles alone or in combination with anti-OX40/anti-PD-L1 are much more effective than immune checkpoint inhibitors alone in delaying melanoma growth in mice. FMD cycles in combination with anti-OX40/anti-PD-L1 also show a trend for increased effects against a lung cancer model. As importantly, the cardiac fibrosis, necrosis and hypertrophy caused by immune checkpoint inhibitors are prevented/reversed by FMD treatment in both cancer models whereas immune infiltration of CD3+ and CD8+ cells in myocardial tissues and systemic and myocardial markers of oxidative stress and inflammation are reduced. These results indicate that FMD cycles in combination with immunotherapy can delay cancer growth while reducing side effects including cardiotoxicity.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Neoplasias Pulmonares , Animais , Camundongos , Cardiotoxicidade , Inibidores de Checkpoint Imunológico/efeitos adversos , Jejum , Dieta , Imunoterapia/efeitos adversos , Neoplasias Pulmonares/terapia , Miocárdio
2.
Neuropathol Appl Neurobiol ; 37(4): 381-94, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20946108

RESUMO

AIMS: Proteins of the Polycomb repressive complex 2 (PRC2) are epigenetic gene silencers and are involved in tumour development. Their oncogenic function might be associated with their role in stem cell maintenance. The histone methyltransferase Enhancer of Zeste 2 (EZH2) is a key member of PRC2 function: we have investigated its expression and function in gliomas. METHODS: EZH2 expression was studied in grade II-IV gliomas and in glioma stem-like cells (GSC) by quantitative PCR and immunohistochemistry. Effects of EZH2 down-regulation were analysed by treating GSC with the histone deacetylase (HDAC) inhibitor suberoylanide hydroxamic acid (SAHA) and by shRNA. RESULTS: DNA microarray analysis showed that EZH2 is highly expressed in murine and human GSC. Real-time PCR on gliomas of different grade (n = 66) indicated that EZH2 is more expressed in glioblastoma multiforme (GBM) than in low-grade gliomas (P = 0.0013). This was confirmed by immunohistochemistry on an independent set of 106 gliomas. Treatment with SAHA caused significant up-regulation of PRC2 predicted target genes, GSC disruption and decreased expression of EZH2 and of the stem cell marker CD133. Inhibition of EZH2 expression by shRNA was associated with a significant decrease of glioma proliferation. CONCLUSION: The data suggest that EZH2 plays a role in glioma progression and encourage the therapeutic targeting of these malignancies by HDAC inhibitors.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Ligação a DNA/genética , Glioma/genética , Fatores de Transcrição/genética , Animais , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Separação Celular , Imunoprecipitação da Cromatina , Progressão da Doença , Proteína Potenciadora do Homólogo 2 de Zeste , Citometria de Fluxo , Glioma/metabolismo , Glioma/patologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Imuno-Histoquímica , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Complexo Repressor Polycomb 2 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/efeitos dos fármacos , Vorinostat
3.
J Cell Biol ; 152(2): 335-48, 2001 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-11266450

RESUMO

Duchenne muscular dystrophy is a lethal recessive disease characterized by widespread muscle damage throughout the body. This increases the difficulty of cell or gene therapy based on direct injections into muscles. One way to circumvent this obstacle would be to use circulating cells capable of homing to the sites of lesions. Here, we showed that stem cell antigen 1 (Sca-1), CD34 double-positive cells purified from the muscle tissues of newborn mice are multipotent in vitro and can undergo both myogenic and multimyeloid differentiation. These muscle-derived stem cells were isolated from newborn mice expressing the LacZ gene under the control of the muscle-specific desmin or troponin I promoter and injected into arterial circulation of the hindlimb of mdx mice. The ability of these cells to interact and firmly adhere to endothelium in mdx muscles microcirculation was demonstrated by intravital microscopy after an intraarterial injection. Donor Sca-1, CD34 muscle-derived stem cells were able to migrate from the circulation into host muscle tissues. Histochemical analysis showed colocalization of LacZ and dystrophin expression in all muscles of the injected hindlimb in all of five out of five 8-wk-old treated mdx mice. Their participation in the formation of muscle fibers was significantly increased by muscle damage done 48 h after their intraarterial injection, as indicated by the presence of 12% beta-galactosidase-positive fibers in muscle cross sections. Normal dystrophin transcripts detected enzymes in the muscles of the hind limb injected intraarterially by the mdx reverse transcription polymerase chain reaction method, which differentiates between normal and mdx message. Our results showed that the muscle-derived stem cells first attach to the capillaries of the muscles and then participate in regeneration after muscle damage.


Assuntos
Transplante de Células/métodos , Distrofina/genética , Células-Tronco Hematopoéticas/fisiologia , Músculo Esquelético/citologia , Distrofia Muscular Animal/terapia , Actinas/análise , Animais , Animais Recém-Nascidos , Antígenos CD34/análise , Antígenos Ly/análise , Adesão Celular , Diferenciação Celular , Linhagem Celular , Distrofina/análise , Endotélio Vascular/fisiologia , Terapia Genética , Células-Tronco Hematopoéticas/citologia , Membro Posterior , Imunofenotipagem , Injeções Intra-Arteriais , Proteínas de Membrana/análise , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Microcirculação/fisiologia , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/fisiologia , Miosinas/análise , Transcrição Gênica , beta-Galactosidase/análise , beta-Galactosidase/genética
4.
Cell Transplant ; 16(6): 563-77, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17912948

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive muscle disease due to defect on the gene encoding dystrophin. The lack of a functional dystrophin in muscles results in the fragility of the muscle fiber membrane with progressive muscle weakness and premature death. There is no cure for DMD and current treatment options focus primarily on respiratory assistance, comfort care, and delaying the loss of ambulation. Recent works support the idea that stem cells can contribute to muscle repair as well as to replenishment of the satellite cell pool. Here we tested the safety of autologous transplantation of muscle-derived CD133+ cells in eight boys with Duchenne muscular dystrophy in a 7-month, double-blind phase I clinical trial. Stem cell safety was tested by measuring muscle strength and evaluating muscle structures with MRI and histological analysis. Timed cardiac and pulmonary function tests were secondary outcome measures. No local or systemic side effects were observed in all treated DMD patients. Treated patients had an increased ratio of capillary per muscle fibers with a switch from slow to fast myosin-positive myofibers.


Assuntos
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Distrofia Muscular de Duchenne/terapia , Mioblastos Esqueléticos/transplante , Peptídeos/metabolismo , Antígeno AC133 , Adolescente , Antígenos CD/classificação , Antígenos CD/isolamento & purificação , Criança , Método Duplo-Cego , Estudos de Viabilidade , Seguimentos , Glicoproteínas/classificação , Glicoproteínas/isolamento & purificação , Humanos , Separação Imunomagnética/classificação , Imunofenotipagem/classificação , Injeções Intramusculares , Masculino , Contração Muscular/fisiologia , Músculo Esquelético/citologia , Distrofia Muscular de Duchenne/patologia , Mioblastos Esqueléticos/citologia , Peptídeos/classificação , Peptídeos/isolamento & purificação , Transplante de Células-Tronco , Células-Tronco/citologia , Transplante Autólogo , Transplante Homólogo/efeitos adversos , Resultado do Tratamento
5.
Neuroscience ; 125(1): 179-89, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15051157

RESUMO

Rat dermis is a source of cells capable of growing in vitro and, in appropriate conditions, forming floating spheres constituted by nestin-positive cells. We have clonally grown these spheres up to the 15th generation. These spheres can be dissociated into cells that differentiate in vitro under appropriate conditions, these cells are labeled by antibodies to immature neuron markers such as nestin and beta-tubulin III and, later, to mature neuron markers such as microtubule-associated protein 2 and neurofilaments. However, most cells are positive to the astroglial marker glia fibrillary acidic protein (GFAP). When sphere-derived cells are transplanted into the spinal cord after traumatic injury, their migration into the lesion cavity is optimal but their differentiation is dependent upon the time interval between lesioning and cell transplantation. Injection of skin-derived stem cell within 30 min from injury yields mainly membrane activated complex-1 (MAC-1), cluster of differentiation-4 (CD-4) and CD-8 positive cells, that 60-90 days later undergo apoptosis. However, when transplantation is performed 7 days after injury, most cells (65% of total) are positive to staining with antibodies to GFAP, others (16%) to neurofilaments, and a smaller amount (2%) to the endothelial marker, platelet endothelial cell adhesion molecule. Thus our study shows that delayed transplantations of dermis-derived stem cells yield healthy cells that do not die, migrate to the lesion site, and there differentiate mainly in cells expressing glia and neuronal markers. On the other hand there is the possibility of dye transfer from labeled cells to endogenous cells, and this might influence the data.


Assuntos
Diferenciação Celular/fisiologia , Derme/citologia , Neurônios/fisiologia , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco , Animais , Western Blotting , Movimento Celular/fisiologia , Derme/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Tempo
6.
Cell Transplant ; 12(1): 91-100, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12693669

RESUMO

Migration of transplanted myogenic cells occurs during both embryogenesis and regeneration of skeletal muscles and is important for successful myoblast transplantation, but little is known about factors that promote chemotaxis of these cells. Tumor necrosis factor-alpha (TNF-alpha) is known to induce chemotactic effect on several cell types. In this study, we investigated its influence on the in vitro and in vivo motility of C2C12 and primary myoblasts. In the in vitro test performed in the blind-well Boyden chambers, we showed that TNF-alpha (50-400 U/ml) significantly enhanced the ability of myogenic cells to migrate. The dose-response curve for this factor was bell shaped, with maximum activity in the 200 U/ml range. In the in vivo test, intramuscular administration of TNF-alpha was performed by an Alzet pump connected to a perforated polyethylene microtube inserted in the tibialis anterior (TA) of CD1 mice. In these experiments, myoblasts were injected under the muscle epimysium. The recipient mice were immunosuppressed with FK506. Our results showed that, 5 days after myoblast transplantation, cells migrated further in the muscles infused with TNF-alpha than in the muscles not exposed to TNF-alpha. TNF-alpha not only has a chemotactic activity but may also modify cell migration via its action on matrix metalloproteinase (MMP) expression. The proteolytic activities of the MMPs secreted in the muscles were thus also assessed by gelatin zymography. The results showed an increased of MMP-2 and MMP-9 transcripts in the TNF-alpha-infused muscles injected with myogenic cells. Myoblast migration during transplantation may be enhanced by overlapping gradients of several effector molecules such as TNF-alpha, interferon-gamma (INF-gamma), and interleukins, released at the site of muscle injury. We propose that TNF-alpha may promote myoblast migration directly through chemotactic activity and indirectly by enhancing MMP activity at the site of muscle injury.


Assuntos
Diferenciação Celular/fisiologia , Quimiotaxia/fisiologia , Músculo Esquelético/metabolismo , Doenças Musculares/terapia , Mioblastos/transplante , Transplante de Tecidos/métodos , Fator de Necrose Tumoral alfa/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Citocinas/imunologia , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Imunossupressores/farmacologia , Antígeno-1 Associado à Função Linfocitária/efeitos dos fármacos , Antígeno-1 Associado à Função Linfocitária/metabolismo , Antígeno de Macrófago 1/efeitos dos fármacos , Antígeno de Macrófago 1/metabolismo , Metaloproteinases da Matriz/efeitos dos fármacos , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/farmacologia
7.
Cell Transplant ; 10(2): 209-21, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11332636

RESUMO

Ex vivo gene therapy of Duchenne muscular dystrophy based on autologous transplantation of genetically modified myoblasts is limited by their premature senescence. MyoD-converted fibroblasts represent an alternative source of myogenic cells. In this study the forced MyoD-dependent conversion of murine NIH-3T3 fibroblasts into myoblasts under the control of an inducible promoter silent in the presence of tetracycline was evaluated. After tetracycline withdrawal this promoter drives the transcription of MyoD in the engineered fibroblasts, inducing their myogenesis and giving rise to beta-galactosidase-positive cells. MyoD-expressing fibroblasts withdrew from the cell cycle, but were unable to fuse in vitro into multinucleated myotubes. Five days following implantation of engineered fibroblasts in muscles of C57BL/10J mice we observed a sevenfold increase of beta-galactosidase-positive regenerating myofibers in animals not treated with antibiotic compared with treated animals. After 1 week the number of positive fibers decreased and several apoptotic myonuclei were detected. Three weeks following implantation of MyoD-converted fibroblasts in recipient mice, no positive "blue" fiber was observed. Our results suggest that transactivation by tetracycline of MyoD may drive an in vivo myogenic conversion of NIH-3T3 fibroblasts and that, in this experimental setting, apoptosis plays a relevant role in limiting the efficacy of engineered fibroblast transplantation. This work opens the question whether apoptotic phenomena also play a general role as limiting factors of cell-mediated gene therapy of inherited muscle disorders.


Assuntos
Apoptose/fisiologia , Diferenciação Celular/fisiologia , Transplante de Células , Músculo Esquelético/citologia , Proteína MyoD/genética , Tetraciclina/farmacologia , Células 3T3 , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Regulação da Expressão Gênica , Terapia Genética/métodos , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/citologia , Músculo Esquelético/efeitos dos fármacos , Distrofias Musculares/terapia , Proteína MyoD/fisiologia , Regiões Promotoras Genéticas , Transcrição Gênica , Transfecção , beta-Galactosidase/análise , beta-Galactosidase/biossíntese
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa