Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Cell Metab ; 35(8): 1441-1456.e9, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37494932

RESUMO

This study reveals a previously uncharacterized mechanism to restrict intestinal inflammation via a regulatory RNA transcribed from a noncoding genomic locus. We identified a novel transcript of the lncRNA HOXA11os specifically expressed in the distal colon that is reduced to undetectable levels in colitis. HOXA11os is localized to mitochondria under basal conditions and interacts with a core subunit of complex 1 of the electron transport chain (ETC) to maintain its activity. Deficiency of HOXA11os in colonic myeloid cells results in complex I deficiency, dysfunctional oxidative phosphorylation (OXPHOS), and the production of mitochondrial reactive oxygen species (mtROS). As a result, HOXA11os-deficient mice develop spontaneous intestinal inflammation and are hypersusceptible to colitis. Collectively, these studies identify a new regulatory axis whereby a lncRNA maintains intestinal homeostasis and restricts inflammation in the colon through the regulation of complex I activity.


Assuntos
Colite , RNA Longo não Codificante , Animais , Camundongos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Colite/genética , Colite/metabolismo , Inflamação/metabolismo , Mitocôndrias/genética , Homeostase , Mucosa Intestinal/metabolismo
2.
Cancer Biomark ; 32(3): 327-337, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34151835

RESUMO

BACKGROUND: Recent studies demonstrated the involvement of mesenchymal stem/stromal cells (MSCs) in carcinogenesis, but the molecular mechanism behind this transformation is still obscured. OBJECTIVE: To screen both the expression levels of polycomb and trithorax epigenetic regulators and TrP53 mutations in early and late MSC culture passages in an attempt to decipher the mechanism of spontaneous transformation. METHODS: The study was conducted on early and late passages of MSC culture model from C57BL/6J mice. The expression profile of 84 epigenetic regulators was examined using RT2 profiler PCR array. TrP53 mutations in the DNA binding domain was screened. Codons, amino acids positions and the corresponding human variants were detected in P53 sequences. RESULTS: Sixty-two epigenetic regulators were dysregulated. Abnormalities were detected starting the third passage. Nine regulators were dysregulated in all passages. (C>G) substitution P53 mutation was detected in passage 3 resulting in Ser152Arg substitution. Passages 6, 9, 12 and the last passage showed T>C substitution resulting in Cys235Arg substitution. The last passage had T deletion and A insertion resulting in frame shift mutations changing the p.Phe286Ser and p.Asn103Lys respectively. CONCLUSION: In vitro expanded MSCs undergo transformation through alteration of epigenetic regulators which results in genomic instability and frequent P53 mutations.


Assuntos
Epigênese Genética/genética , Proteína Supressora de Tumor p53/genética , Animais , Proliferação de Células , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Mutação
3.
Immunity ; 54(6): 1137-1153.e8, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34051146

RESUMO

Alterations in the cGAS-STING DNA-sensing pathway affect intestinal homeostasis. We sought to delineate the functional role of STING in intestinal inflammation. Increased STING expression was a feature of intestinal inflammation in mice with colitis and in humans afflicted with inflammatory bowel disease. Mice bearing an allele rendering STING constitutively active exhibited spontaneous colitis and dysbiosis, as well as progressive chronic intestinal inflammation and fibrosis. Bone marrow chimera experiments revealed STING accumulation in intestinal macrophages and monocytes as the initial driver of inflammation. Depletion of Gram-negative bacteria prevented STING accumulation in these cells and alleviated intestinal inflammation. STING accumulation occurred at the protein rather than transcript level, suggesting post-translational stabilization. We found that STING was ubiquitinated in myeloid cells, and this K63-linked ubiquitination could be elicited by bacterial products, including cyclic di-GMP. Our findings suggest a positive feedback loop wherein dysbiosis foments the accumulation of STING in intestinal myeloid cells, driving intestinal inflammation.


Assuntos
Colite/imunologia , Disbiose/imunologia , Imunidade Inata/imunologia , Proteínas de Membrana/imunologia , Células Mieloides/imunologia , Ubiquitinação/imunologia , Animais , Estudos de Casos e Controles , Feminino , Humanos , Inflamação/imunologia , Intestinos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia
4.
Nat Immunol ; 21(3): 274-286, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32066947

RESUMO

Human immunodeficiency virus 1 (HIV-1) infection is associated with heightened inflammation and excess risk of cardiovascular disease, cancer and other complications. These pathologies persist despite antiretroviral therapy. In two independent cohorts, we found that innate lymphoid cells (ILCs) were depleted in the blood and gut of people with HIV-1, even with effective antiretroviral therapy. ILC depletion was associated with neutrophil infiltration of the gut lamina propria, type 1 interferon activation, increased microbial translocation and natural killer (NK) cell skewing towards an inflammatory state, with chromatin structure and phenotype typical of WNT transcription factor TCF7-dependent memory T cells. Cytokines that are elevated during acute HIV-1 infection reproduced the ILC and NK cell abnormalities ex vivo. These results show that inflammatory cytokines associated with HIV-1 infection irreversibly disrupt ILCs. This results in loss of gut epithelial integrity, microbial translocation and memory NK cells with heightened inflammatory potential, and explains the chronic inflammation in people with HIV-1.


Assuntos
Citocinas/sangue , HIV-1/imunologia , HIV-1/patogenicidade , Imunidade Inata , Células Matadoras Naturais/imunologia , Linfócitos/imunologia , Fator 1 de Transcrição de Linfócitos T/imunologia , Regulação da Expressão Gênica , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , Homeostase/imunologia , Humanos , Memória Imunológica , Técnicas In Vitro , Inflamação/genética , Inflamação/imunologia , Inflamação/virologia , Fator 1 de Transcrição de Linfócitos T/genética , Via de Sinalização Wnt/imunologia
5.
J Exp Med ; 215(3): 841-858, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29374027

RESUMO

Genetically predisposed CTLA4 insufficiency in humans is associated with gastric cancer development, which is paradoxical to the prototypical role of CTLA4 in suppressing antitumor immunity. CTLA4 is a critical immune checkpoint against autoimmune disorders. Autoimmunity has been implicated in protumor or antitumor activities. Here, we show that CTLA4 insufficiency initiates de novo tumorigenesis in the mouse stomach through inflammation triggered by host-intrinsic immune dysregulation rather than microbiota, with age-associated progression to malignancy accompanied by epigenetic dysregulation. The inflammatory tumorigenesis required CD4 T cells, but not the TH1 or TH17 subsets. Deficiencies in IL-4 and IL-13 or IL-4 receptor α broke the link between inflammation and initiation of tumorigenesis. This study establishes the causality of CTLA4 insufficiency in gastric cancer and uncovers a role of type 2 inflammation in initiating gastric epithelial transformation. These findings suggest possible improvement of immune therapies by blocking tumorigenic type 2 inflammation while preserving antitumor type 1 immunity.


Assuntos
Antígeno CTLA-4/deficiência , Carcinogênese/patologia , Citocinas/metabolismo , Inflamação/patologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Anticorpos/farmacologia , Autoimunidade , Carcinogênese/metabolismo , Epigênese Genética , Humanos , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Interferon gama/metabolismo , Interleucinas/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microbiota , Peptídeos , Lesões Pré-Cancerosas/patologia , Interferência de RNA , Estômago/patologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Células Th1/imunologia , Células Th17/imunologia
6.
Dig Dis Sci ; 59(6): 1169-79, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24798995

RESUMO

BACKGROUND/AIMS: Stromal cells and the extracellular environment are vital to human tumors, influencing growth and response to therapy. Human tumor cell lines lack stroma and transplantation into immunodeficient mice does not allow meaningful analyses of the effects of stroma on tumor cell growth. Studies of xenografts of primary human tumor fragments in nude mice and in early scid mouse models were constrained by poor tumor growth accompanied by host-versus-graft reactivity, dramatically altering tumor architecture and tumor microenvironment. In contrast, severely immunodeficient NOD-scid and NOD-Rag1 (null) strains carrying the IL2rg (null) mutation (NSG and NRG) support the growth of many types of human primary tumors. METHODS/RESULTS: We compared the take rate, growth and architectural preservation of 10 clinically distinct primary human colon cancers in NOD-scid, NOD-Rag1 (null) , NSG and NRG mice and determined the contribution of mouse and human cells to the stroma during tumor proliferation and expansion in secondary hosts and tumor response to treatment with 5-fluorouracil (5-FU). NSG and NRG mice more readily support growth of human primary colon tumor fragments than do NOD-scid, NOD-Rag1 (null) mice and maintain tumor architectural integrity in the primary recipient and through subsequent transplant generations. The human colon tumors were responsive to treatment with 5-FU. Human stromal cells in the primary graft were replaced by mouse-derived fibroblasts in a dynamic process during subsequent passages. CONCLUSION: Human colon cancer xenografts propagated in NSG and NRG mice maintain structural fidelity while replacing human stromal cells with murine stromal cells.


Assuntos
Neoplasias do Colo/metabolismo , Proteínas de Homeodomínio/metabolismo , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Células Estromais/fisiologia , Idoso , Animais , Antimetabólitos Antineoplásicos/efeitos adversos , Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Fluoruracila/efeitos adversos , Fluoruracila/uso terapêutico , Proteínas de Homeodomínio/genética , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Organismos Livres de Patógenos Específicos
7.
Am J Physiol Gastrointest Liver Physiol ; 306(12): G1075-88, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24789207

RESUMO

Bone marrow-derived mesenchymal stem cells (MSCs) sustain cancer cells by creating a microenvironment favorable for tumor growth. In particular, MSCs have been implicated in gastric cancer development. There is extensive evidence suggesting that Hedgehog signaling regulates tumor growth. However, very little is known regarding the precise roles of Hedgehog signaling and MSCs in tumor development within the stomach. The current study tests that hypothesis that Sonic Hedgehog (Shh), secreted from MSCs, provides a proliferative stimulus for the gastric epithelium in the presence of inflammation. Red fluorescent protein-expressing MSCs transformed in vitro (stMSCs) were transduced with lentiviral constructs containing a vector control (stMSC(vect)) or short hairpin RNA (shRNA) targeting the Shh gene (stMSC(ShhKO)). Gastric submucosal transplantation of wild-type MSCs (wtMSCs), wild-type MSCs overexpressing Shh (wtMSC(Shh)), stMSC(vect), or stMSC(ShhKO) cells in C57BL/6 control (BL/6) or gastrin-deficient (GKO) mice was performed and mice analyzed 30 and 60 days posttransplantation. Compared with BL/6 mice transplanted with wtMSC(Shh) and stMSC(vect) cells, inflamed GKO mice developed aggressive gastric tumors. Tumor development was not observed in mouse stomachs transplanted with wtMSC or stMSC(ShhKO) cells. Compared with stMSC(ShhKO)-transplanted mice, within the inflamed GKO mouse stomach, Shh-expressing stMSC(vect)- and wtMSC(Shh)-induced proliferation of CD44-positive cells. CD44-positive cells clustered in gland-like structures within the tumor stroma and were positive for Patched (Ptch) expression. We conclude that Shh, secreted from MSCs, provides a proliferative stimulus for the gastric epithelium that is associated with tumor development, a response that is sustained by chronic inflammation.


Assuntos
Proliferação de Células , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Gastrite/metabolismo , Proteínas Hedgehog/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Epiteliais/citologia , Mucosa Gástrica/patologia , Gastrinas/deficiência , Gastrite/patologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
8.
PLoS One ; 8(9): e75225, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24069395

RESUMO

Studies using Helicobacter-infected mice show that bone marrow-derived mesenchymal stem cells (MSCs) can repopulate the gastric epithelium and promote gastric cancer progression. Within the tumor microenvironment of the stomach, pro-inflammatory cytokine interferon-gamma (IFNγ) and Sonic hedgehog (Shh) are elevated. IFNγ is implicated in tumor proliferation via activation of the Shh signaling pathway in various tissues but whether a similar mechanism exists in the stomach is unknown. We tested the hypothesis that IFNγ drives MSC proliferation and recruitment, a response mediated by Shh signaling. The current study uses transplantation of an in vitro transformed mesenchymal stem cell line (stMSC(vect)), that over-expresses hedgehog signaling, in comparison to non-transformed wild-type MSCs (wtMSCs), wtMSCs transfected to over-express Shh (wtMSC(Shh)), and stMSCs transduced with lentiviral constructs containing shRNA targeting the Shh gene (stMSC(ShhKO)). The effect of IFNγ on MSC proliferation was assessed by cell cycle analysis in vitro using cells treated with recombinant IFNγ (rmIFNγ) alone, or in combination with anti-Shh 5E1 antibody, and in vivo using mice transplanted with MSCs treated with PBS or rmIFNγ. In vitro, IFNγ significantly increased MSC proliferation, a response mediated by Shh that was blocked by 5E1 antibody. The MSC population collected from bone marrow of PBS- or IFNγ-treated mice showed that IFNγ significantly increased the percentage of all MSC cell lines in S phase, with the exception of the stMSCs(ShhKO) cells. While the MSC cell lines with intact Shh expression were recruited to the gastric mucosa in response to IFNγ, stMSCs(ShhKO) were not. Hedgehog signaling is required for MSC proliferation and recruitment to the stomach in response to IFNγ.


Assuntos
Transformação Celular Neoplásica/metabolismo , Mucosa Gástrica/metabolismo , Proteínas Hedgehog/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Estômago/patologia , Animais , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Quimiocina CXCL12/metabolismo , Adesões Focais/genética , Adesões Focais/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Técnicas de Silenciamento de Genes , Inativação Gênica , Proteínas Hedgehog/genética , Interferon gama/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Transdução de Sinais/efeitos dos fármacos
9.
Dig Dis Sci ; 58(9): 2466-77, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23873382

RESUMO

BACKGROUND: Helicobacter infection is the main risk factor in developing gastric cancer. Mesenchymal stem cells (MSCs) are non-hematopoietic stromal cells, which are able to differentiate into different cell lineages. MSC contribute to cancer development by forming the tumor directly, contributing to the microenvironment, or by promoting angiogenesis and metastasis. CXCR4/SDF-1 axis is used by MSC in trafficking, homing, and engraftment at chronic inflammation sites, and plays an important role in tumorigenesis. AIM: To determine if CXCR4 receptor has a role in MSC contribution to the development of Helicobacter-mediated gastric cancer. METHODS: SDF-1 and CXCR4 expression in mouse gastric mucosa in the setting of acute and chronic inflammation was measured using RT-PCR. Mouse culture-adapted MSC express CXCR4. Wild-type C57BL/6 mice infected with Helicobacter felis for 6 months or controls were given IV injections of CXCR4 knock-down MSC. Animals were followed for another 4 months. Homing of MSC in the stomach was quantified using RT-PCR. MSC differentiation into gastric epithelia lineages was analyzed using immunohistochemistry and fluorescent in situ hybridization. RESULTS: CXCR4 and SDF-1 are both upregulated in the settings of Helicobacter-induced chronic gastric inflammation. CXCR4 is fully required for homing of MSC to the stomach in acute gastric inflammation, but only partially in Helicobacter-induced gastric cancer. MSC lead to gastric intraepithelial neoplasia as early as 10 months of Helicobacter infection. CONCLUSIONS: Our results show that MSC have a tumorigenic effect by promoting an accelerated form of gastric cancer in mice. The engraftment of MSC in chronic inflammation is only partially CXCR4-dependent.


Assuntos
Gastrite/metabolismo , Infecções por Helicobacter/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptores CXCR4/metabolismo , Neoplasias Gástricas/etiologia , Animais , Diferenciação Celular , Células Epiteliais/citologia , Etanol , Gastrite/complicações , Gastrite/microbiologia , Infecções por Helicobacter/complicações , Helicobacter felis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Estômago/patologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
10.
Stem Cells Dev ; 21(10): 1822-30, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21973238

RESUMO

Tumor recurrence after chemotherapy is a major cause of patient morbidity and mortality. Recurrences are thought to be secondary to small subsets of cancer cells that are better able to survive traditional forms of chemotherapy and thus drive tumor regrowth. The ability to isolate and better characterize these therapy-resistant cells is critical for the future development of targeted therapies aimed at achieving more robust and long-lasting responses. Using a novel application for the proliferation marker carboxyfluorescein diacetate, succinimidyl ester (CFSE), we have identified a population of slow-cycling, label-retaining tumor cells in both in vitro sphere cultures and in vivo xenograft models. Strikingly, label-retaining cells exhibit a multifold increase in ability to survive traditional forms of chemotherapy and reenter the cell cycle. Further, we demonstrate the innovative application of CFSE to live sort slow-cycling tumor cells and validate their chemoresistance and tumorigenic potential.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Neoplasias Experimentais/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Fluoresceínas/química , Fluoresceínas/metabolismo , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Fluoruracila/administração & dosagem , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias Experimentais/tratamento farmacológico , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina , Succinimidas/química , Succinimidas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nat Cell Biol ; 13(10): 1214-23, 2011 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-21909099

RESUMO

The midbody is a singular organelle formed between daughter cells during cytokinesis and required for their final separation. Midbodies persist in cells long after division as midbody derivatives (MB(d)s), but their fate is unclear. Here we show that MB(d)s are inherited asymmetrically by the daughter cell with the older centrosome. They selectively accumulate in stem cells, induced pluripotent stem cells and potential cancer 'stem cells' in vivo and in vitro. MB(d) loss accompanies stem-cell differentiation, and involves autophagic degradation mediated by binding of the autophagic receptor NBR1 to the midbody protein CEP55. Differentiating cells and normal dividing cells do not accumulate MB(d)s and possess high autophagic activity. Stem cells and cancer cells accumulate MB(d)s by evading autophagosome encapsulation and exhibit low autophagic activity. MB(d) enrichment enhances reprogramming to induced pluripotent stem cells and increases the in vitro tumorigenicity of cancer cells. These results indicate unexpected roles for MB(d)s in stem cells and cancer 'stem cells'.


Assuntos
Autofagia , Transformação Celular Neoplásica/patologia , Reprogramação Celular , Células-Tronco Embrionárias/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Neoplásicas/patologia , Organelas/patologia , Animais , Autofagia/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Divisão Celular , Linhagem Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Centrossomo/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Técnicas de Cocultura , Células-Tronco Embrionárias/metabolismo , Células HeLa , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Lisossomos/metabolismo , Camundongos , Células-Tronco Neoplásicas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Organelas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Transfecção
12.
Stem Cells Dev ; 20(1): 11-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20677919

RESUMO

This study characterizes the contribution of bone marrow-derived cells (BMDCs) to Barrett's adenocarcinoma of the esophagus using a mouse surgical model of disease and human specimens. Transplantation of bone marrow expressing beta galactosidase into a wild-type mouse, followed by surgical esophagojejunostomy, allowed tracking of BMDCs into the surgical anastomosis and resulting Barrett's metaplasia. Human tissue from a male patient who had been transplanted with female bone marrow and later developed esophageal adenocarcinoma allowed us to tract donor-derived cells into the tumor. Using a combination of antibodies directed against beta-galactosidase (animal studies) and X/Y fluorescent in situ hybridization (FISH) (human studies), combined with specific lineage staining directed against epithelial, fibroblast, endothelial, and leukocyte markers, we show that bone marrow cells contribute to both the epithelial and stromal component of esophageal adenocarcinoma. These findings demonstrate that BMDCs can generate cancer-associated fibroblasts as well as contribute directly to epithelial cells in cancer of the esophagus.


Assuntos
Adenocarcinoma/patologia , Esôfago de Barrett/patologia , Células da Medula Óssea/patologia , Endotélio/patologia , Miofibroblastos/patologia , Transplante de Células-Tronco/efeitos adversos , Adenocarcinoma/cirurgia , Adulto , Animais , Esôfago de Barrett/cirurgia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Masculino , Metaplasia , Camundongos , Pessoa de Meia-Idade , Células Estromais/metabolismo , Células Estromais/patologia , Transplante Homólogo
13.
Dig Dis Sci ; 55(6): 1516-24, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20437100

RESUMO

Sonic Hedgehog (Shh) signaling has been extensively studied for its role in developmental biology and cancer biology. The association between Shh and cancer development in general is well established but the functional role of Shh in the development and progression of gastric cancer specifically is largely unknown. Bone marrow-derived stem cells, specifically mesenchymal stem cells (MSCs) infiltrate and engraft into the gastric mucosa in response to the chronic inflammatory environment of Helicobacter infection. In this review, MSC infiltration and changes in the cytokine and cellular profiles of later-stage chronic environments will be tied into their interactions with the Shh pathway. We will discuss how these changes shape tumorigenesis and tumor progression in the gastric mucosa. The current review focuses on the Shh signaling pathway and its role in the development of gastric cancer, specifically in response to Helicobacter pylori infection. We follow with an in-depth discussion of the regulation of the Hedgehog pathway during acute and chronic gastric inflammation with a focus on signaling within the MSC compartment.


Assuntos
Transformação Celular Neoplásica/metabolismo , Mucosa Gástrica/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Animais , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/patologia , Citocinas/metabolismo , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Gastrite Atrófica/metabolismo , Gastrite Atrófica/microbiologia , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Humanos , Concentração de Íons de Hidrogênio , Mediadores da Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo , Peptídeo Hidrolases/metabolismo , Processamento de Proteína Pós-Traducional , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Regulação para Cima
14.
Stem Cells Dev ; 19(8): 1153-66, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20199238

RESUMO

Neoplastic epithelia may remain dormant and clinically unapparent in human patients for decades. Multiple risk factors including mutations in tumor cells or the stromal cells may affect the switch from dormancy to malignancy. Gene mutations, including p53 mutations, within the stroma of tumors are associated with a worse clinical prognosis; however, it is not known if these stromal mutations can promote tumors in genetically at-risk tissue. To address this question, Apc(Min/+) and Apc(Min/+) Rag2(-/-) mice, which have a predilection to mammary carcinoma (as well as wild-type (wt) mice), received mesenchymal stem cells (MSC) with mutant p53 (p53MSC) transferred via tail vein injection. In the wt mouse, p53MSC circulated in the periphery and homed to the marrow cavity where they could be recovered up to a year later without apparent effect on the health of the mouse. No mammary tumors were found. However, in mice carrying the Apc(Min/+) mutation, p53MSC homed to mammary tissue and significantly increased the incidence of mammary carcinoma. Tumor necrosis factor (TNF)-alpha-dependent factors elaborated from mesenchymal cells converted quiescent epithelia into clinically apparent disease. The increased cancer phenotype was completely preventable with neutralization of TNF-alpha or by transfer of CD4(+) regulatory T cells from immune competent donors, demonstrating that immune competency to regulate inflammation was sufficient to maintain neoplastic dormancy even in the presence of oncogenic epithelial and stromal mutations. The significant synergy between host immunity and mesenchymal cells identified here may restructure treatments to restore an anticancer microenvironment.


Assuntos
Células da Medula Óssea/patologia , Neoplasias Mamárias Animais/etiologia , Células-Tronco Mesenquimais/patologia , Mutação/genética , Proteína Supressora de Tumor p53/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antígenos CD/metabolismo , Medula Óssea , Células da Medula Óssea/metabolismo , Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/etiologia , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Meios de Cultivo Condicionados/metabolismo , Meios de Cultivo Condicionados/farmacologia , Proteínas de Ligação a DNA/genética , Epitélio/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Genes APC , Humanos , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Mutantes , Camundongos SCID , Pessoa de Meia-Idade , Mutação de Sentido Incorreto/genética , Transplante de Neoplasias/patologia , Células Estromais/metabolismo , Células Estromais/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/transplante , Transplante Heterólogo/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
15.
Hematology ; 14(4): 187-97, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19635181

RESUMO

Autologous stem cell transplantation (ASCT) and rituximab based therapy represent effective treatments of indolent B-cell lymphoproliferative disorders (B-LPDs) that often induce molecular remission (MR). We assessed the impact of MR after treatment on prognosis of 57 patients with indolent B-LPDs. We also evaluated the impact of therapy on patients' outcome. Failure to achieve MR was identified as an independent risk factor regardless of treatment modality. PCR positive patients had shorter progression free survival (PFS) in contrast with patients in MR after rituximab (median 0.75 and 2.5 years respectively; p=0.006) or patients in MR after rituximab followed by ASCT (median 3.3 years; p=0.0032). PCR positive patients had a 5-year overall survival (OS) of only 40% compared to a 5-year OS of 76% for PCR negative patients after rituximab (p=0.0186) and 86% PCR negative patients after rituximab with ASCT (p=0.003). All nine patients transplanted with PCR positive graft relapsed (p=0.0023) with shorter PFS (p=0.0008). Rituximab based therapy induced MR in 25 (64%) compared to 18 (100%) patients after rituximab followed by ASCT (p=0.0025). We observed no difference in PFS between the transplant group (3.3 years) and rituximab based treatment (1.9 years), but the 5-year OS of patients with transplant was 85 and 59% respectively (p=0.0271). Patients with indolent B-LPDs who achieve MR have better prognosis. Rituximab based therapy induces MR in high number of patients, which can be further improved by ASCT and patients have an excellent outcome. PCR positive harvest represents a high risk of relapse after ASCT.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Linfoma de Células B/terapia , Transtornos Linfoproliferativos/terapia , Adulto , Idoso , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Murinos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Terapia Combinada , Intervalo Livre de Doença , Humanos , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/genética , Transtornos Linfoproliferativos/tratamento farmacológico , Transtornos Linfoproliferativos/genética , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase/métodos , Análise de Regressão , Rituximab , Taxa de Sobrevida , Resultado do Tratamento , Adulto Jovem
16.
Gastroenterology ; 137(3): 934-44, 944.e1-4, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19524576

RESUMO

BACKGROUND & AIMS: Analysis of clinical colon cancer specimens show alterations in the CD95 (Fas Ag/Fas L) pathway as tumors progress from local to metastatic disease, suggesting that this pathway may play a role in invasive behavior of colon cancer. However, direct causality between these alterations and clinical disease progression has not been shown. METHODS: Surgically resected metastatic colon cancer samples were evaluated for Fas Ag/L and apoptosis. Alterations in the Fas-signaling pathway found in human samples were recreated through a series of staged transfection experiments in the MC38 mouse colon cancer cell line and the effects on growth tested in vitro and in vivo. RESULTS: Expression of FLICE-like inhibitory protein confers apoptosis resistance, increasing the incidence of primary tumors through a survival advantage by avoiding apoptosis and inducing Fas-mediated proliferation. Coexpression of Fas L enables colon cancer cells to metastasize to the liver from local tumors as well as from intravenous injection of cells. MC38-FasL/FLICE-like inhibitory protein colon cancer cells induce apoptosis in hepatocytes via activation of type II Fas Ag signaling, thus creating a niche conducive to tumor growth and fueling their own growth via Fas proliferative signaling. CONCLUSIONS: Alterations in the Fas Ag pathway which inhibit apoptosis and increase Fas-mediated proliferation directly increase local colon cancer growth, and enhance metastasis to the liver. Delineating points in the pathway responsible for growth and metastasis will offer targets that may be exploited for therapy.


Assuntos
Neoplasias do Colo/patologia , Neoplasias Hepáticas/secundário , Transdução de Sinais , Receptor fas/metabolismo , Animais , Apoptose , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Neoplasias do Colo/metabolismo , Proteína Ligante Fas/metabolismo , Feminino , Hepatócitos/metabolismo , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transfecção , Células Tumorais Cultivadas
17.
J Immunol ; 183(1): 642-9, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19535625

RESUMO

Helicobacter infection is the primary risk factor for gastric cancer, with the cytokine environment within the gastric mucosa the strongest predictor of disease risk. Elevated TNF-alpha, IL-1beta, and low IL-10 are associated with the highest risk. In this study, we used C57BL/6 mice to identify T-bet as a central regulator of the cytokine environment during Helicobacter felis infection. We infected male and female C57BL/6 and C57BL/6-T-bet knockout (KO) litter mates with H. felis and examined the bacterial colonization, immune response, and mucosal damage at varying time points. T-bet KO mice maintained infection for 15 mo at similar levels to wild-type mice. Infection and immune response did not differ between male and female mice. Despite sustained infection, T-bet KO mice respond with a blunted Th1 response associated with preservation of parietal and chief cells and protection from the development of gastric cancer. Unexpectedly, T-bet KO mice develop a gastric environment that would not be expected based on the phenotype of T-bet KO CD4 cells alone. T-bet KO mice respond to H. felis infection with a markedly blunted IL-1beta and TNF-alpha and elevated IL-10 levels. Activity of this one master regulator modulates the expression of the key gastric mucosal cytokines associated with gastric cancer and may be a target for therapy to restore immune balance clinically in patients at risk for gastric cancer.


Assuntos
Adenocarcinoma/microbiologia , Adenocarcinoma/prevenção & controle , Infecções por Helicobacter/prevenção & controle , Helicobacter felis/imunologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/prevenção & controle , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Adenocarcinoma/imunologia , Animais , Feminino , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Predisposição Genética para Doença , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/patologia , Interleucina-1beta/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Gástricas/imunologia , Proteínas com Domínio T/fisiologia , Fator de Necrose Tumoral alfa/fisiologia
18.
Methods Mol Biol ; 511: 267-95, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19347301

RESUMO

Animal models are necessary to reproduce the complex host, microbial and environmental influences associated with infectious carcinogenesis of the digestive system. Today, mouse models are preferred by most researchers because of cost efficiencies, rapid reproduction, choice of laboratory reagents, and availability of genetically engineered mutants to study specific gene functions in vivo. Mouse models have validated the once-provocative hypothesis that Helicobacter pylori infection is a major risk factor for gastric carcinoma, dispelling early skepticism over the pathogenic nature of this organism in the human stomach. Enterohepatic Helicobacter spp. induce inflammatory bowel disease and colorectal carcinoma in susceptible mouse strains, permitting study of host immunity and microbial factors at the cellular and molecular level. H. hepaticus is the only proven infectious hepatocarcinogen of mice and has been used to explore mechanisms of inflammation-associated liver cancer as seen in human chronic viral hepatitis. For example, this model was used to identify for the first time a potential mechanism for male-predominant liver cancer risk independent of circulating sex hormones. Helicobacter-based mouse models of digestive system carcino-genesis are used to investigate the basic biology of inflammation-associated human cancers and to evaluate therapeutic interventions at the discovery level. Because of exciting advances in genetic engineering of mice, in vivo imaging, and system-wide genomics and proteomics, these models will provide even more information in the future. This chapter introduces the mouse as a model species; summarizes important models of inflammation-associated cancer incited by murine Helicobacter infection; and describes methods for the collection, sampling, and histologic grading of mouse digestive system tissues.


Assuntos
Modelos Animais de Doenças , Trato Gastrointestinal , Helicobacter/patogenicidade , Neoplasias Gástricas/microbiologia , Animais , Células da Medula Óssea/fisiologia , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/patologia , Trato Gastrointestinal/anatomia & histologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Infecções por Helicobacter/patologia , Infecções por Helicobacter/fisiopatologia , Hepatite/microbiologia , Hepatite/patologia , Humanos , Inflamação/complicações , Inflamação/patologia , Doenças Inflamatórias Intestinais/complicações , Doenças Inflamatórias Intestinais/patologia , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Neoplasias Gástricas/patologia
19.
Curr Opin Oncol ; 21(1): 77-82, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19125022

RESUMO

PURPOSE OF REVIEW: Cancer-associated fibroblasts/myofibroblasts and inflammatory cells produce a vast array of growth factors, chemokines and extracellular matrix (ECM) components that facilitate cancer progression, invasion/metastasis and neovascularization. This review highlights some surprisingly novel mechanisms of this paracrine relationship. RECENT FINDINGS: Mesenchymal stem/stromal cells (MSCs) are known for their tropism towards certain tumours, but now we find that cross-talk between tumours and MSCs leads to greater tumour motility and metastasis. Two closely related populations of immature myeloid cells, so-called 'cap cells' and myeloid-derived suppressor cells (MDSCs) also cross-talk with tumour cells, promoting invasion and metastasis through matrix metalloproteinase (MMP) secretion, as well as contributing to neovascularization and T-cell tolerance. The contribution of bone marrow-derived cells (BMDCs) to tumour neovascularization is controversial, but BMD--endothelial progenitor cells (EPCs)--are strongly implicated in the angiogenic switch in a mouse model. BMDCs are also credited with the creation of premetastatic niches to which metastatic cells adhere via integrins. SUMMARY: There is no doubt that BMDCs are not simply bystanders in the tumour battleground. The mechanisms through which they aid tumour progression are numerous; effective treatments that combat BMDC-tumour cross-talk are surely on the way.


Assuntos
Células da Medula Óssea/imunologia , Neoplasias da Medula Óssea/imunologia , Inflamação , Neoplasias Epiteliais e Glandulares/imunologia , Animais , Humanos
20.
Int J Antimicrob Agents ; 33(5): 473-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19157800

RESUMO

Helicobacter infection, one of the most common bacterial infections in man worldwide, is a type 1 carcinogen and the most important risk factor for gastric cancer. Helicobacter pylori bacterial factors, components of the host genetics and immune response, dietary cofactors and decreased acid secretion resulting in bacterial overgrowth are all considered important factors for induction of gastric cancer. Components found in green tea have been shown to inhibit bacterial growth, including the growth of Helicobacter spp. In this study, we assessed the bactericidal and/or bacteriostatic effect of green tea against Helicobacter felis and H. pylori in vitro and evaluated the effects of green tea on the development of Helicobacter-induced gastritis in an animal model. Our data clearly demonstrate profound growth effects of green tea against Helicobacter and, importantly, demonstrate that green tea consumption can prevent gastric mucosal inflammation if ingested prior to exposure to Helicobacter infection. Research in the area of natural food compounds and their effects on various disease states has gained increased acceptance in the past several years. Components within natural remedies such as green tea could be further used for prevention and treatment of Helicobacter-induced gastritis in humans.


Assuntos
Antibacterianos/uso terapêutico , Infecções por Helicobacter/prevenção & controle , Helicobacter felis/efeitos dos fármacos , Helicobacter pylori/efeitos dos fármacos , Extratos Vegetais/uso terapêutico , Chá/química , Animais , Antibacterianos/farmacologia , Mucosa Gástrica/patologia , Gastrite/prevenção & controle , Humanos , Masculino , Camundongos , Extratos Vegetais/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...