Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cancer Immunol Res ; 7(6): 874-885, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31018956

RESUMO

Chronic inflammation, including that driven by autoimmunity, is associated with the development of B-cell lymphomas. IL1R8 is a regulatory receptor belonging to the IL1R family, which negatively regulates NF-κB activation following stimulation of IL1R or Toll-like receptor family members. IL1R8 deficiency is associated with the development of severe autoimmune lupus-like disease in lpr mice. We herein investigated whether concomitant exacerbated inflammation and autoimmunity caused by the deficiency of IL1R8 could recapitulate autoimmunity-associated lymphomagenesis. We thus monitored B-cell lymphoma development during the aging of IL1R8-deficient lpr mice, observing an increased lymphoid cell expansion that evolved to diffuse large B-cell lymphoma (DLBCL). Molecular and gene-expression analyses showed that the NF-κB pathway was constitutively activated in Il1r8 -/-/lpr B splenocytes. In human DLBCL, IL1R8 had reduced expression compared with normal B cells, and higher IL1R8 expression was associated with a better outcome. Thus, IL1R8 silencing is associated with increased lymphoproliferation and transformation in the pathogenesis of B-cell lymphomas associated with autoimmunity.


Assuntos
Autoimunidade/genética , Suscetibilidade a Doenças , Linfoma/etiologia , Receptores de Interleucina-1/deficiência , Animais , Biomarcadores , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Predisposição Genética para Doença , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Imuno-Histoquímica , Linfoma/metabolismo , Linfoma/patologia , Linfoma Difuso de Grandes Células B/etiologia , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/patologia , Camundongos , NF-kappa B/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo
2.
Oncotarget ; 7(48): 80059-80076, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27517630

RESUMO

A humoral immune response against aberrant tumor proteins can be elicited in cancer patients, resulting in the production of auto-antibodies (Abs). By serological proteome analysis we identified the surface membrane protein ADAM10, a metalloproteinase that has a role in epithelial-tumor progression and invasion, as a target of the immune response in colorectal cancer (Crc). A screening carried out on the purified protein using testing cohorts of sera (Crc patients n = 57; control subjects n = 39) and validation cohorts of sera (Crc patients n = 49; control subjects n = 52) indicated that anti-ADAM10 auto-Abs were significantly induced in a large group (74%) of colon cancer patients, in particular in patients at stage II and III of the disease. Interestingly, in Crc patients classified as stage III disease, the presence of anti-ADAM10 auto-Abs in the sera was associated with a favourable follow-up with a significant shifting of the recurrence-free survival median time from 23 to 55 months. Even though the ADAM10 protein was expressed in Crc regardless the presence of auto-Abs, the immature/non-functional isoform of ADAM10 was highly expressed in the tumor of anti-ADAM10-positive patients and was the isoform targeted by the auto-Abs. In conclusion, the presence of anti-ADAM10 auto-Abs seems to reflect the increased tumor expression of the immunogenic immature-ADAM10 in a group of Crc patients, and is associated with a favourable prognosis in patients at stage III of the disease.


Assuntos
Proteína ADAM10/imunologia , Secretases da Proteína Precursora do Amiloide/imunologia , Autoanticorpos/sangue , Neoplasias Colorretais/sangue , Neoplasias Colorretais/diagnóstico , Proteínas de Membrana/imunologia , Proteína ADAM10/química , Adulto , Idoso , Idoso de 80 Anos ou mais , Secretases da Proteína Precursora do Amiloide/química , Formação de Anticorpos/fisiologia , Autoanticorpos/metabolismo , Biomarcadores Tumorais/sangue , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Feminino , Humanos , Masculino , Proteínas de Membrana/química , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Domínios Proteicos/imunologia , Precursores de Proteínas/química , Precursores de Proteínas/imunologia
3.
Haematologica ; 99(8): 1356-64, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24859880

RESUMO

The sialic-acid-binding immunoglobulin-like lectin SIGLEC-G is a negative regulator of B-cell receptor-mediated calcium signaling. Its deficiency leads to reduced turnover and increased proliferation and survival of murine B-1a cells. Siglecg(-/-) mice show a premature expansion of polyclonal CD5(+) B cells in the spleen and the peritoneal cavity. Here we studied the fate of B lymphocytes in Siglecg(-/-) mice over time. We demonstrate that in aging animals SIGLEC-G deficiency promotes progressive accumulation of monoclonal B lymphocytes and increases the susceptibility to develop B-cell lymphoproliferative disorders. Lymphoid tumors arising in aged Siglecg(-/-) mice are monoclonal and histologically heterogeneous as they include diffuse large B-cell lymphoma, follicular lymphoma, and medium-to-large B-cell monomorphic lymphoma but surprisingly not chronic lymphocytic leukemia. The tumors express high levels of BCL-2 and are transplantable. In keeping with these findings we have also observed a remarkable down-regulation of the human ortholog SIGLEC10 in human B-cell lymphoma and leukemia cell lines. Taken together, these observations indicate that the down-regulation of negative B-cell receptor regulators such as SIGLEC-G/SIGLEC10 may represent another mechanism relevant to the pathogenesis of B-cell lymphomas.


Assuntos
Linfócitos B/metabolismo , Predisposição Genética para Doença , Lectinas/deficiência , Leucemia de Células B/metabolismo , Linfoma de Células B/metabolismo , Receptores de Antígenos de Linfócitos B/deficiência , Animais , Predisposição Genética para Doença/genética , Humanos , Lectinas/genética , Leucemia de Células B/genética , Leucemia de Células B/patologia , Linfoma de Células B/genética , Linfoma de Células B/patologia , Transtornos Linfoproliferativos/genética , Transtornos Linfoproliferativos/metabolismo , Transtornos Linfoproliferativos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Antígenos de Linfócitos B/genética , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico
4.
Dev Cell ; 24(6): 586-99, 2013 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-23477786

RESUMO

Pericytes are endothelial-associated cells that contribute to vessel wall. Here, we report that pericytes may derive from direct conversion of committed skeletal myoblasts. When exposed to Dll4 and PDGF-BB, but not Dll1, skeletal myoblasts downregulate myogenic genes, except Myf5, and upregulate pericyte markers, whereas inhibition of Notch signaling restores myogenesis. Moreover, when cocultured with endothelial cells, skeletal myoblasts, previously treated with Dll4 and PDGF-BB, adopt a perithelial position stabilizing newly formed vessel-like networks in vitro and in vivo. In a transgenic mouse model in which cells expressing MyoD activate Notch, skeletal myogenesis is abolished and pericyte genes are activated. Even if overexpressed, Myf5 does not trigger myogenesis because Notch induces Id3, partially sequestering Myf5 and inhibiting MEF2 expression. Myf5-expressing cells adopt a perithelial position, as occasionally also observed in wild-type (WT) embryos. These data indicate that endothelium, via Dll4 and PDGF-BB, induces a fate switch in adjacent skeletal myoblasts.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Proteínas de Membrana/farmacologia , Desenvolvimento Muscular , Proteína MyoD/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Fator Regulador Miogênico 5/metabolismo , Pericitos/citologia , Pericitos/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Becaplermina , Proteínas de Ligação ao Cálcio/farmacologia , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais , Regulação da Expressão Gênica no Desenvolvimento , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteínas Inibidoras de Diferenciação/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/biossíntese , Proteínas Serrate-Jagged , Transdução de Sinais , Ativação Transcricional
5.
EMBO Rep ; 12(2): 164-71, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21212806

RESUMO

Despite having distinct expression patterns and phenotypes in mutant mice, the myogenic regulatory factors Myf5 and MyoD have been considered to be functionally equivalent. Here, we report that these factors have a different response to DNA damage, due to the presence in MyoD and absence in Myf5 of a consensus site for Abl-mediated tyrosine phosphorylation that inhibits MyoD activity in response to DNA damage. Genotoxins failed to repress skeletal myogenesis in MyoD-null embryos; reintroduction of wild-type MyoD, but not mutant Abl phosphorylation-resistant MyoD, restored the DNA-damage-dependent inhibition of muscle differentiation. Conversely, introduction of the Abl-responsive phosphorylation motif converts Myf5 into a DNA-damage-sensitive transcription factor. Gene-dosage-dependent reduction of Abl kinase activity in MyoD-expressing cells attenuated the DNA-damage-dependent inhibition of myogenesis. The presence of a DNA-damage-responsive phosphorylation motif in vertebrate, but not in invertebrate MyoD suggests an evolved response to environmental stress, originated from basic helix-loop-helix gene duplication in vertebrate myogenesis.


Assuntos
Desenvolvimento Muscular/efeitos dos fármacos , Mutagênicos/toxicidade , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Evolução Biológica , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Reagentes de Ligações Cruzadas/toxicidade , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Etoposídeo/toxicidade , Feminino , Técnicas de Silenciamento de Genes , Metanossulfonato de Metila/toxicidade , Camundongos/embriologia , Mitomicina/toxicidade , Proteína MyoD/genética , Fator Regulador Miogênico 5/genética , Fosforilação , Gravidez , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-abl/fisiologia , Interferência de RNA , Somitos/efeitos dos fármacos , Somitos/metabolismo , Proteínas Supressoras de Tumor/metabolismo
6.
Nat Cell Biol ; 9(3): 255-67, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17293855

RESUMO

Cells derived from blood vessels of human skeletal muscle can regenerate skeletal muscle, similarly to embryonic mesoangioblasts. However, adult cells do not express endothelial markers, but instead express markers of pericytes, such as NG2 proteoglycan and alkaline phosphatase (ALP), and can be prospectively isolated from freshly dissociated ALP(+) cells. Unlike canonical myogenic precursors (satellite cells), pericyte-derived cells express myogenic markers only in differentiated myotubes, which they form spontaneously with high efficiency. When transplanted into severe combined immune deficient-X-linked, mouse muscular dystrophy (scid-mdx) mice, pericyte-derived cells colonize host muscle and generate numerous fibres expressing human dystrophin. Similar cells isolated from Duchenne patients, and engineered to express human mini-dystrophin, also give rise to many dystrophin-positive fibres in vivo. These data show that myogenic precursors, distinct from satellite cells, are associated with microvascular walls in the human skeletal muscle, may represent a correlate of embryonic 'mesoangioblasts' present after birth and may be a promising candidate for future cell-therapy protocols in patients.


Assuntos
Células-Tronco Adultas/citologia , Músculo Esquelético/citologia , Pericitos/citologia , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/citologia , Adolescente , Adulto , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/transplante , Idoso , Animais , Antígenos CD/análise , Técnicas de Cultura de Células/métodos , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Camundongos Nus , Camundongos SCID , Pessoa de Meia-Idade , Proteínas Musculares/análise , Proteínas Musculares/genética , Músculo Esquelético/química , Músculo Esquelético/fisiologia , Distrofia Muscular de Duchenne/fisiopatologia , Distrofia Muscular de Duchenne/cirurgia , Pericitos/química , Pericitos/transplante , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/transplante , Transplante de Células-Tronco/métodos , Resultado do Tratamento
7.
Proc Natl Acad Sci U S A ; 104(1): 264-9, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17182743

RESUMO

Duchenne muscular dystrophy is a relatively common disease that affects skeletal muscle, leading to progressive paralysis and death. There is currently no resolutive therapy. We have developed a treatment in which we combined the effects of nitric oxide with nonsteroidal antiinflammatory activity by using HCT 1026, a nitric oxide-releasing derivative of flurbiprofen. Here, we report the results of long-term (1-year) oral treatment with HCT 1026 of two murine models for limb girdle and Duchenne muscular dystrophies (alpha-sarcoglycan-null and mdx mice). In both models, HCT 1026 significantly ameliorated the morphological, biochemical, and functional phenotype in the absence of secondary effects, efficiently slowing down disease progression. HCT 1026 acted by reducing inflammation, preventing muscle damage, and preserving the number and function of satellite cells. HCT 1026 was significantly more effective than the corticosteroid prednisolone, which was analyzed in parallel. As an additional beneficial effect, HCT 1026 enhanced the therapeutic efficacy of arterially delivered donor stem cells, by increasing 4-fold their ability to migrate and reconstitute muscle fibers. The therapeutic strategy we propose is not selective for a subset of mutations; it provides ground for immediate clinical experimentation with HCT 1026 alone, which is approved for use in humans; and it sets the stage for combined therapies with donor or autologous, genetically corrected stem cells.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Flurbiprofeno/análogos & derivados , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Doadores de Óxido Nítrico/uso terapêutico , Transplante de Células-Tronco , Animais , Apoptose/efeitos dos fármacos , Terapia Combinada , Creatina Quinase/sangue , Flurbiprofeno/farmacologia , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/patologia , Prednisolona/farmacologia , Regeneração/efeitos dos fármacos , Sarcoglicanas/fisiologia
8.
Nature ; 444(7119): 574-9, 2006 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-17108972

RESUMO

Duchenne muscular dystrophy remains an untreatable genetic disease that severely limits motility and life expectancy in affected children. The only animal model specifically reproducing the alterations in the dystrophin gene and the full spectrum of human pathology is the golden retriever dog model. Affected animals present a single mutation in intron 6, resulting in complete absence of the dystrophin protein, and early and severe muscle degeneration with nearly complete loss of motility and walking ability. Death usually occurs at about 1 year of age as a result of failure of respiratory muscles. Here we report that intra-arterial delivery of wild-type canine mesoangioblasts (vessel-associated stem cells) results in an extensive recovery of dystrophin expression, normal muscle morphology and function (confirmed by measurement of contraction force on single fibres). The outcome is a remarkable clinical amelioration and preservation of active motility. These data qualify mesoangioblasts as candidates for future stem cell therapy for Duchenne patients.


Assuntos
Células-Tronco Adultas/transplante , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Transplante de Células-Tronco , Células-Tronco Adultas/imunologia , Animais , Terapia Combinada , Creatina Quinase/sangue , Cães , Distrofina/biossíntese , Distrofina/genética , Distrofina/imunologia , Terapia Genética , Humanos , Masculino , Células Musculares , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transplante Autólogo , Transplante Heterólogo
9.
Arterioscler Thromb Vasc Biol ; 25(4): 692-7, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15662021

RESUMO

OBJECTIVE: To test the potential of mesoangioblasts (Mabs) in reducing postischemic injury in comparison with bone marrow progenitor cells (BMPCs), fibroblasts (Fbs), and embryonic stem cell-derived endothelial cells (ECs), and to identify putative cellular protective mechanisms. METHODS AND RESULTS: Cells were injected percutaneously in the left ventricular (LV) chamber of C57BL/6 mice, 3 to 6 hours after coronary ligation, and detected in the hearts 2 days and 6 weeks later. Echocardiographic examinations were performed at 6 weeks. LV dilation was reduced and LV shortening fraction was improved with Mabs and BMPCs but not with ECs and Fbs. Donor cell colonization of the host myocardium was modest and predominantly in the smooth muscle layer of vessels. Capillary density was higher in the peripheral infarct area and apoptotic cardiomyocytes were fewer with Mabs and BMPCs. Mabs and BMPCs, but not Fbs or ECs, promoted survival of cultured cardiocytes under low-oxygen in culture. This activity was present in Mab-conditioned medium and could be replaced by a combination of basic fibroblast growth factor (bFGF), insulin-like growth factor (IGF)-1, and hepatocyte growth factor (HGF), all of which are produced by these cells. Conditioned medium from Mabs, but not from Fbs, stimulated proliferation of smooth muscle cells in vitro. CONCLUSIONS: Mabs appear as effective as BMPCs in reducing postinfarction LV dysfunction, likely through production of antiapoptotic and angiogenic factors.


Assuntos
Endotélio Vascular/transplante , Fibroblastos/transplante , Células-Tronco Hematopoéticas/citologia , Células-Tronco Multipotentes/citologia , Infarto do Miocárdio/terapia , Transplante de Células-Tronco , Animais , Apoptose , Vasos Sanguíneos/citologia , Ecocardiografia , Endotélio Vascular/citologia , Fibroblastos/citologia , Sobrevivência de Enxerto , Células-Tronco Hematopoéticas/fisiologia , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/fisiologia , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/fisiopatologia , Miocárdio/citologia , Neovascularização Fisiológica , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia
10.
Science ; 301(5632): 487-92, 2003 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-12855815

RESUMO

Preclinical or clinical trials for muscular dystrophies have met with modest success, mainly because of inefficient delivery of viral vectors or donor cells to dystrophic muscles. We report here that intra-arterial delivery of wild-type mesoangioblasts, a class of vessel-associated stem cells, corrects morphologically and functionally the dystrophic phenotype of virtually all downstream muscles in adult immunocompetent alpha-sarcoglycan (alpha-SG) null mice, a model organism for limb-girdle muscular dystrophy. When mesoangioblasts isolated from juvenile dystrophic mice and transduced with a lentiviral vector expressing alpha-SG were injected into the femoral artery of dystrophic mice, they reconstituted skeletal muscle in a manner similar to that seen in wild-type cells. The success of this protocol was mainly due to widespread distribution of donor stem cells through the capillary network, a distinct advantage of this strategy over previous approaches.


Assuntos
Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Músculo Esquelético/fisiologia , Distrofia Muscular Animal/terapia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/embriologia , Diferenciação Celular , Linhagem Celular , Movimento Celular , Distrofina/metabolismo , Endotélio Vascular/fisiologia , Feminino , Artéria Femoral , Vetores Genéticos , Lentivirus/genética , Locomoção , Masculino , Mesoderma/citologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Contração Muscular , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patologia , Regeneração , Sarcoglicanas , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA