Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 541(7636): 228-232, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28024296

RESUMO

Although long non-coding RNAs (lncRNAs) are non-protein-coding transcripts by definition, recent studies have shown that a fraction of putative small open reading frames within lncRNAs are translated. However, the biological significance of these hidden polypeptides is still unclear. Here we identify and functionally characterize a novel polypeptide encoded by the lncRNA LINC00961. This polypeptide is conserved between human and mouse, is localized to the late endosome/lysosome and interacts with the lysosomal v-ATPase to negatively regulate mTORC1 activation. This regulation of mTORC1 is specific to activation of mTORC1 by amino acid stimulation, rather than by growth factors. Hence, we termed this polypeptide 'small regulatory polypeptide of amino acid response' (SPAR). We show that the SPAR-encoding lncRNA is highly expressed in a subset of tissues and use CRISPR/Cas9 engineering to develop a SPAR-polypeptide-specific knockout mouse while maintaining expression of the host lncRNA. We find that the SPAR-encoding lncRNA is downregulated in skeletal muscle upon acute injury, and using this in vivo model we establish that SPAR downregulation enables efficient activation of mTORC1 and promotes muscle regeneration. Our data provide a mechanism by which mTORC1 activation may be finely regulated in a tissue-specific manner in response to injury, and a paradigm by which lncRNAs encoding small polypeptides can modulate general biological pathways and processes to facilitate tissue-specific requirements, consistent with their restricted and highly regulated expression profile.


Assuntos
Complexos Multiproteicos/metabolismo , Músculos/fisiologia , Peptídeos/metabolismo , RNA Longo não Codificante/genética , Regeneração/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Adenosina Trifosfatases/metabolismo , Aminoácidos/metabolismo , Aminoácidos/farmacologia , Animais , Sistemas CRISPR-Cas/genética , Endossomos/metabolismo , Edição de Genes , Células HEK293 , Humanos , Lisossomos/enzimologia , Lisossomos/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/agonistas , Músculos/lesões , Especificidade de Órgãos , Peptídeos/deficiência , Peptídeos/genética , Transdução de Sinais/efeitos dos fármacos
2.
Biochem Biophys Res Commun ; 515(4): 538-543, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31176486

RESUMO

Chronic inflammatory responses have profound effects on the differentiation and activity of both the bone-forming osteoblasts and bone-resorbing osteoclasts. Importantly, inflammatory bone diseases characterized by clinical osteolysis promote bone resorption and decrease bone formation by uncoupling the process in favor of excess resorption. Notch signaling regulates osteoclast development and thus its manipulation has the potential to suppress resorptive potential. Here, we have utilized a genetic model of Notch inhibition in osteoclasts by expression of dnMAML to prevent formation of transcriptional complex essential for downstream Notch signaling. Using this model and LPS as a tool for experimental inflammatory osteolysis, we have demonstrated that dnMAML-expressing osteoclasts exhibited significantly lower maturation and resorption/functional potential ex vivo using TRAP staining and calcium phosphate coated surfaces. Moreover, we observed that while LPS stimulated the formation of wildtype osteoclasts pre-treated with RANKL, dnMAML expression produced resistance to osteoclast maturation after LPS stimulation. Genetically, Notch-inhibited animals showed a significantly lower TRAP and CTX-1 levels in serum after LPS treatment compared to the control groups in addition to a marked reduction in osteoclast surfaces in calvaria sections. This report provides evidence for modulation of Notch signaling activity to protect against inflammatory osteolysis. Taken together, the findings of this study will help guide the development of Notch signaling-based therapeutic approaches to prevent bone loss.


Assuntos
Lipopolissacarídeos/farmacologia , Osteoclastos/citologia , Osteólise/prevenção & controle , Receptores Notch/deficiência , Transdução de Sinais , Animais , Colágeno Tipo I/sangue , Colágeno Tipo I/deficiência , Feminino , Camundongos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Peptídeos/sangue , Peptídeos/deficiência , Ligante RANK/farmacologia , Receptores Notch/biossíntese , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fosfatase Ácida Resistente a Tartarato/sangue , Fosfatase Ácida Resistente a Tartarato/deficiência , Fosfatase Ácida Resistente a Tartarato/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
3.
J Am Chem Soc ; 139(42): 14817-14824, 2017 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-28949546

RESUMO

A significant challenge toward studies of the human microbiota involves establishing causal links between bacterial metabolites and human health and disease states. Certain strains of commensal Escherichia coli harbor the 54-kb clb gene cluster which codes for small molecules named precolibactins and colibactins. Several studies suggest colibactins are genotoxins and support a role for clb metabolites in colorectal cancer formation. Significant advances toward elucidating the structures and biosynthesis of the precolibactins and colibactins have been made using genetic approaches, but their full structures remain unknown. In this Perspective we describe recent synthetic efforts that have leveraged biosynthetic advances and shed light on the mechanism of action of clb metabolites. These studies indicate that deletion of the colibactin peptidase ClbP, a modification introduced to promote accumulation of precolibactins, leads to the production of non-genotoxic pyridone-based isolates derived from the diversion of linear biosynthetic intermediates toward alternative cyclization pathways. Furthermore, these studies suggest the active genotoxins (colibactins) are unsaturated imines that are potent DNA damaging agents, thereby confirming an earlier mechanism of action hypothesis. Although these imines have very recently been detected in bacterial extracts, they have to date confounded isolation. As the power of "meta-omics" approaches to natural products discovery further advance, we anticipate that chemical synthetic and biosynthetic studies will become increasingly interdependent.


Assuntos
Neoplasias Colorretais/microbiologia , Microbioma Gastrointestinal , Mutagênicos/metabolismo , Peptídeos/metabolismo , Policetídeos/metabolismo , Animais , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Dano ao DNA , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Humanos , Iminas/química , Iminas/metabolismo , Peptídeo Hidrolases/metabolismo , Peptídeos/deficiência , Peptídeos/genética
4.
J Biol Chem ; 289(49): 34408-21, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25342752

RESUMO

The Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) family proteins mediate the adherence of infected erythrocytes to microvascular endothelia of various organs, including the placenta, thereby contributing to cerebral, placental, and other severe malaria pathogenesis. Several parasite proteins, including KAHRP and PfEMP3, play important roles in the cytoadherence by mediating the clustering of PfEMP1 in rigid knoblike structures on the infected erythrocyte surface. The lack of a subtelomeric region of chromosome 2 that contains kahrp and pfemp3 causes reduced cytoadherence. In this study, microarray transcriptome analysis showed that the absence of a gene cluster, comprising kahrp, pfemp3, and four other genes, results in the loss of parasitized erythrocytes adhering to chondroitin 4-sulfate (C4S). The role of one of these genes, PF3D7_0201600/PFB0080c, which encodes PHISTb (Plasmodium helical interspersed subtelomeric b) domain-containing RESA-like protein 1 expressed on the infected erythrocyte surface, was investigated. Disruption of PFB0080c resulted in increased var2csa transcription and VAR2CSA surface expression, leading to higher C4S-binding capacity of infected erythrocytes. Further, PFB0080c-knock-out parasites stably maintained the C4S adherence through many generations of growth. Although the majority of PFB0080c-knock-out parasites bound to C4S even after culturing for 6 months, a minor population bound to both C4S and CD36. These results strongly suggest that the loss of PFB0080c markedly compromises the var gene switching process, leading to a marked reduction in the switching rate and additional PfEMP1 expression by a minor population of parasites. PFB0080c interacts with VAR2CSA and modulates knob-associated Hsp40 expression. Thus, PFB0080c may regulate VAR2CSA expression through these processes. Overall, we conclude that PFB0080c regulates PfEMP1 expression and the parasite's cytoadherence.


Assuntos
Antígenos de Protozoários/genética , Sulfatos de Condroitina/química , Eritrócitos/parasitologia , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Antígenos de Protozoários/metabolismo , Antígenos CD36/genética , Antígenos CD36/metabolismo , Adesão Celular , Sulfatos de Condroitina/metabolismo , Cromossomos , Eritrócitos/química , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Proteínas de Choque Térmico HSP40/genética , Proteínas de Choque Térmico HSP40/metabolismo , Humanos , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Família Multigênica , Peptídeos/deficiência , Peptídeos/genética , Plasmodium falciparum/metabolismo , Ligação Proteica , Proteínas de Protozoários/metabolismo
5.
Am J Physiol Gastrointest Liver Physiol ; 308(1): G12-24, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25324506

RESUMO

The trefoil factor TFF2 is a member of a tripartite family of small proteins that is produced by the stomach and the colon. Recombinant TFF2, when applied intrarectally in a rodent model of hapten colitis, hastens mucosal healing and reduces inflammatory indexes. Additionally, TFF2 is expressed in immune organs, supporting a potential immunomodulatory and reparative role in the bowel. In this study we confirm that TFF2 is expressed in the colon and is specifically enriched in epithelial cells relative to colonic leukocytes. TFF2-deficient, but not TFF1-deficient, mice exhibit a more severe response to acute or chronic dextran sulfate (DSS)-induced colitis that correlates with a 50% loss of expression of TFF3, the principal colonic trefoil. In addition, the response to acute colitis is associated with altered expression of IL-6 and IL-33, but not other inflammatory cytokines. While TFF2 can reduce macrophage responsiveness and block inflammatory cell recruitment to the colon, the major role in limiting the susceptibility to acute colitis appears to be maintenance of barrier function. Bone marrow transfer experiments demonstrate that leukocyte expression of TFF2 is not sufficient for prevention of colitis induction but, rather, that the gastrointestinal epithelium is the primary source of TFF2. Together, these findings illustrate that epithelial TFF2 is an important endogenous regulator of gut mucosal homeostasis that can modulate immune and epithelial compartments. Because of its extreme stability, even in the corrosive gut lumen, TFF2 is an attractive candidate as an oral therapeutic scaffold for future drug development in the treatment of inflammatory bowel disease.


Assuntos
Transplante de Medula Óssea , Colite/metabolismo , Colo/metabolismo , Citocinas/metabolismo , Sulfato de Dextrana , Células Epiteliais/metabolismo , Mediadores da Inflamação/metabolismo , Mucinas/deficiência , Proteínas Musculares/deficiência , Peptídeos/deficiência , Redução de Peso , Animais , Células Cultivadas , Colite/induzido quimicamente , Colite/genética , Colite/imunologia , Colite/patologia , Colite/prevenção & controle , Colo/imunologia , Colo/patologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Interleucina-33 , Interleucina-6/metabolismo , Interleucinas/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/genética , Mucinas/metabolismo , Proteínas Musculares/genética , Peptídeos/genética , Peptídeos/metabolismo , Índice de Gravidade de Doença , Fatores de Tempo , Fator Trefoil-1 , Fator Trefoil-2 , Fator Trefoil-3
6.
Gastroenterology ; 145(6): 1312-22.e1-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23993973

RESUMO

BACKGROUND & AIMS: Chronic inflammation contributes to the pathogenesis of gastric tumorigenesis. The aurora kinase A (AURKA) gene is frequently amplified and overexpressed in gastrointestinal cancers. We investigated the roles of AURKA in inflammation and gastric tumorigenesis. METHODS: We used quantitative real-time reverse transcription polymerase chain reaction, immunofluorescence, immunohistochemistry, luciferase reporter, immunoblot, co-immunoprecipitation, and in vitro kinase assays to analyze AGS and MKN28 gastric cancer cells. We also analyzed Tff1(-/-) mice, growth of tumor xenografts, and human tissues. RESULTS: We correlated increased expression of AURKA with increased levels of tumor necrosis factor-α and inflammation in the gastric mucosa of Tff1(-/-) mice (r = 0.62; P = .0001). MLN8237, an investigational small-molecule selective inhibitor of AURKA, reduced nuclear staining of nuclear factor-κB (NF-κB) p65 in human gastric cancer samples and mouse epithelial cells, suppressed NF-κB reporter activity, and reduced expression of NF-κB target genes that regulate inflammation and cell survival. Inhibition of AURKA also reduced growth of xenograft tumors from human gastric cancer cells in mice and reversed the development of gastric tumors in Tff1(-/-) mice. AURKA was found to regulate NF-κB activity by binding directly and phosphorylating IκBα in cells. Premalignant and malignant lesions from the gastric mucosa of patients had increased levels of AURKA protein and nuclear NF-κB, compared with healthy gastric tissue. CONCLUSIONS: In analyses of gastric cancer cell lines, human tissue samples, and mouse models, we found AURKA to be up-regulated during chronic inflammation to promote activation of NF-κB and tumorigenesis. AURKA inhibitors might be developed as therapeutic agents for gastric cancer.


Assuntos
Adenocarcinoma/metabolismo , Aurora Quinase A/metabolismo , Carcinogênese/metabolismo , Inflamação/metabolismo , Neoplasias Gástricas/metabolismo , Regulação para Cima , Adenocarcinoma/patologia , Animais , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/efeitos dos fármacos , Azepinas/farmacologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Xenoenxertos , Humanos , Técnicas In Vitro , Camundongos Knockout , Camundongos Nus , NF-kappa B/metabolismo , Peptídeos/deficiência , Peptídeos/genética , Peptídeos/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Neoplasias Gástricas/patologia , Fator Trefoil-1
7.
J Immunol ; 189(6): 3078-84, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22896633

RESUMO

IL-12-mediated type 1 inflammation confers host protection against the parasitic protozoan Toxoplasma gondii. However, production of IFN-γ, another type 1 inflammatory cytokine, also drives lethality from excessive injury to the intestinal epithelium. As mechanisms that restore epithelial barrier function following infection remain poorly understood, this study investigated the role of trefoil factor 2 (TFF2), a well-established regulator of mucosal tissue repair. Paradoxically, TFF2 antagonized IL-12 release from dendritic cells (DCs) and macrophages, which protected TFF2-deficient (TFF2(-/-)) mice from T. gondii pathogenesis. Dysregulated intestinal homeostasis in naive TFF2(-/-) mice correlated with increased IL-12/23p40 levels and enhanced T cell recruitment at baseline. Infected TFF2(-/-) mice displayed low rates of parasite replication and reduced gut immunopathology, whereas wild-type (WT) mice experienced disseminated infection and lethal ileitis. p38 MAPK activation and IL-12p70 production was more robust from TFF2(-/-)CD8+ DC compared with WT CD8+ DC and treatment of WT DC with rTFF2 suppressed TLR-induced IL-12/23p40 production. Neutralization of IFN-γ and IL-12 in TFF2(-/-) animals abrogated resistance shown by enhanced parasite replication and infection-induced morbidity. Hence, TFF2 regulated intestinal barrier function and type 1 cytokine release from myeloid phagocytes, which dictated the outcome of oral T. gondii infection in mice.


Assuntos
Regulação para Baixo/imunologia , Mucinas/fisiologia , Proteínas Musculares/fisiologia , Peptídeos/fisiologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Toxoplasmose/parasitologia , Animais , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/genética , Imunidade Celular/genética , Inflamação/imunologia , Inflamação/parasitologia , Inflamação/patologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/parasitologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/deficiência , Proteínas Musculares/deficiência , Peptídeos/deficiência , Fagocitose/genética , Fagocitose/imunologia , Toxoplasma/genética , Toxoplasmose/patologia , Fator Trefoil-2
8.
Nature ; 451(7174): 33-7, 2008 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-18066048

RESUMO

Mating in many species induces a dramatic switch in female reproductive behaviour. In most insects, this switch is triggered by factors present in the male's seminal fluid. How these factors exert such profound effects in females is unknown. Here we identify a receptor for the Drosophila melanogaster sex peptide (SP, also known as Acp70A), the primary trigger of post-mating responses in this species. Females that lack the sex peptide receptor (SPR, also known as CG16752), either entirely or only in the nervous system, fail to respond to SP and continue to show virgin behaviours even after mating. SPR is expressed in the female's reproductive tract and central nervous system. The behavioural functions of SPR map to the subset of neurons that also express the fruitless gene, a key determinant of sex-specific reproductive behaviour. SPR is highly conserved across insects, opening up the prospect of new strategies to control the reproductive and host-seeking behaviours of agricultural pests and human disease vectors.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Peptídeos/metabolismo , Comportamento Sexual Animal/fisiologia , Animais , Sistema Nervoso Central/metabolismo , Sequência Conservada , Copulação/fisiologia , Proteínas de Drosophila/química , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Feminino , Genitália Feminina/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Peptídeos/química , Peptídeos/deficiência , Peptídeos/genética , Receptores de Peptídeos , Especificidade por Substrato , Fatores de Transcrição/metabolismo
9.
Am J Respir Cell Mol Biol ; 49(5): 845-54, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23795648

RESUMO

Pulmonary surfactant protein-C (SP-C) gene-targeted mice (Sftpc(-/-)) develop progressive lung inflammation and remodeling. We hypothesized that SP-C deficiency reduces the ability to suppress repetitive inflammatory injury. Sftpc(+/+) and Sftpc(-/-) mice given three doses of bacterial LPS developed airway and airspace inflammation, which was more intense in the Sftpc(-/-) mice at 3 and 5 days after the final dose. Compared with Sftpc(+/+)mice, inflammatory injury persisted in the lungs of Sftpc(-/-) mice 30 days after the final LPS challenge. Sftpc(-/-) mice showed LPS-induced airway goblet cell hyperplasia with increased detection of Sam pointed Ets domain and FoxA3 transcription factors. Sftpc(-/-) type II alveolar epithelial cells had increased cytokine expression after LPS exposure relative to Sftpc(+/+) cells, indicating that type II cell dysfunction contributes to inflammatory sensitivity. Microarray analyses of isolated type II cells identified a pattern of enhanced expression of inflammatory genes consistent with an intrinsic low-level inflammation resulting from SP-C deficiency. SP-C-containing clinical surfactant extract (Survanta) or SP-C/phospholipid vesicles blocked LPS signaling through the LPS receptor (Toll-like receptor [TLR] 4/CD14/MD2) in human embryonic kidney 293T cells, indicating that SP-C blocks LPS-induced cytokine production by a TLR4-dependent mechanism. Phospholipid vesicles alone did not modify the TLR4 response. In vivo deficiency of SP-C leads to inflammation, increased cytokine production by type II cells, and persistent inflammation after repetitive LPS stimulation.


Assuntos
Endotoxinas , Pulmão/metabolismo , Peptídeos/deficiência , Pneumonia/metabolismo , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Produtos Biológicos/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células Caliciformes/imunologia , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Células HEK293 , Fator 3-gama Nuclear de Hepatócito/metabolismo , Humanos , Hiperplasia , Imunidade Inata , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Receptores de Lipopolissacarídeos/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Peptídeos/genética , Pneumonia/induzido quimicamente , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/patologia , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteína C Associada a Surfactante Pulmonar , Transdução de Sinais , Fatores de Tempo , Receptor 4 Toll-Like/metabolismo
10.
Hepatology ; 55(3): 807-20, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21994122

RESUMO

UNLABELLED: A novel theory in the field of tumor biology postulates that cancer growth is driven by a population of stem-like cells, called tumor-initiating cells (TICs). We previously identified a TIC population derived from hepatocellular carcinoma (HCC) that is characterized by membrane expression of CD133. Here, we describe a novel mechanism by which these cells mediate tumor growth and angiogenesis by systematic comparison of the gene expression profiles between sorted CD133 liver subpopulations through genome-wide microarray analysis. A significantly dysregulated interleukin-8 (IL-8) signaling network was identified in CD133(+) liver TICs obtained from HCC clinical samples and cell lines. IL-8 was found to be overexpressed at both the genomic and proteomic levels in CD133(+) cells isolated from HCC cell lines or clinical samples. Functional studies found enhanced IL-8 secretion in CD133(+) liver TICs to exhibit a greater ability to self-renew, induce tumor angiogenesis, and initiate tumors. In further support of these observations, IL-8 repression in CD133(+) liver TICs by knockdown or neutralizing antibody abolished these effects. Subsequent studies of the IL-8 functional network identified neurotensin (NTS) and CXCL1 to be preferentially expressed in CD133(+) liver TICs. Addition of exogenous NTS resulted in concomitant up-regulation of IL-8 and CXCL1 with simultaneous activation of p-ERK1/2 and RAF-1, both key components of the mitogen-activated protein kinase (MAPK) pathway. Enhanced IL-8 secretion by CD133(+) liver TICs can in turn activate an IL-8-dependent feedback loop that signals through the MAPK pathway. Further, in its role as a liver TIC marker CD133 also plays a functional part in regulating tumorigenesis of liver TICs by way of regulating NTS, IL-8, CXCL1, and MAPK signaling. CONCLUSION: CD133(+) liver TICs promote angiogenesis, tumorigenesis, and self-renewal through NTS-induced activation of the IL-8 signaling cascade.


Assuntos
Antígenos CD/metabolismo , Proliferação de Células , Quimiocina CXCL1/fisiologia , Glicoproteínas/metabolismo , Interleucina-8/fisiologia , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/fisiologia , Neovascularização Patológica/fisiopatologia , Neurotensina/fisiologia , Peptídeos/metabolismo , Transdução de Sinais/fisiologia , Antígeno AC133 , Animais , Antígenos CD/genética , Carcinoma Hepatocelular/irrigação sanguínea , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/cirurgia , Linhagem Celular Tumoral , Células Cultivadas , Retroalimentação Fisiológica , Glicoproteínas/deficiência , Glicoproteínas/genética , Hepatectomia , Humanos , Interleucina-8/deficiência , Interleucina-8/genética , Fígado/irrigação sanguínea , Fígado/patologia , Fígado/cirurgia , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/cirurgia , Camundongos , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno , Células-Tronco Neoplásicas/imunologia , Neurotensina/farmacologia , Peptídeos/deficiência , Peptídeos/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Blood ; 117(20): 5532-40, 2011 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-21422469

RESUMO

Administration of a single dose of anti-CD40L mAb at the time of allogeneic BM transplantation tolerizes peripheral alloreactive T cells and permits establishment of mixed hematopoietic chimerism in mice. Once engrafted, mixed chimeras are systemically tolerant to donor Ags through a central deletion mechanism and will accept any donor organ indefinitely. We previously found that the PD-1/PD-L1 pathway is required for CD8 T-cell tolerance in this model. However, the cell population that must express PD-1 and the role of other inhibitory molecules were unknown. Here, we report that LAG-3 is required for long-term peripheral CD8 but not CD4 T-cell tolerance and that this requirement is CD8 cell-extrinsic. In contrast, adoptive transfer studies revealed a CD8 T cell-intrinsic requirement for CTLA4/B7.1/B7.2 and for PD-1 for CD8 T-cell tolerance induction. We also observed that both PD-L1 and PD-L2 are independently required on donor cells to achieve T-cell tolerance. Finally, we uncovered a requirement for TGF-ß signaling into T cells to achieve peripheral CD8 but not CD4 T-cell tolerance in this in vivo system.


Assuntos
Antígenos CD/imunologia , Antígenos de Superfície/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Linfócitos T CD8-Positivos/imunologia , Tolerância Imunológica , Fator de Crescimento Transformador beta/imunologia , Transferência Adotiva , Animais , Antígenos CD/genética , Antígenos de Superfície/genética , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Antígeno B7-H1 , Transplante de Medula Óssea/imunologia , Linfócitos T CD4-Positivos/imunologia , Ligante de CD40/antagonistas & inibidores , Antígeno CTLA-4 , Feminino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Imunológicos , Peptídeos/deficiência , Peptídeos/genética , Peptídeos/imunologia , Proteína 2 Ligante de Morte Celular Programada 1 , Receptor de Morte Celular Programada 1 , Transdução de Sinais/imunologia , Transplante Homólogo , Proteína do Gene 3 de Ativação de Linfócitos
12.
J Immunol ; 187(3): 1097-105, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21697456

RESUMO

Programmed death-1 ligand 1 (PD-L1) is a coinhibitory molecule that negatively regulates multiple tolerance checkpoints. In the NOD mouse model, PD-L1 regulates the development of diabetes. PD-L1 has two binding partners, programmed death-1 and B7-1, but the significance of the PD-L1:B7-1 interaction in regulating self-reactive T cell responses is not yet clear. To investigate this issue in NOD mice, we have compared the effects of two anti-PD-L1 Abs that have different blocking activities. Anti-PD-L1 mAb 10F.2H11 sterically and functionally blocks only PD-L1:B7-1 interactions, whereas anti-PD-L1 mAb 10F.9G2 blocks both PD-L1:B7-1 and PD-L1:programmed death-1 interactions. Both Abs had potent, yet distinct effects in accelerating diabetes in NOD mice: the single-blocker 10F.2H11 mAb was more effective at precipitating diabetes in older (13-wk-old) than in younger (6- to 7-wk-old) mice, whereas the dual-blocker 10F.9G2 mAb rapidly induced diabetes in NOD mice of both ages. Similarly, 10F.2H11 accelerated diabetes in recipients of T cells from diabetic, but not prediabetic mice, whereas 10F.9G2 was effective in both settings. Both anti-PD-L1 mAbs precipitated diabetes in adoptive transfer models of CD4(+) and CD8(+) T cell-driven diabetes. Taken together, these data demonstrate that the PD-L1:B7-1 pathway inhibits potentially pathogenic self-reactive effector CD4(+) and CD8(+) T cell responses in vivo, and suggest that the immunoinhibitory functions of this pathway may be particularly important during the later phases of diabetogenesis.


Assuntos
Antígeno B7-1/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Inibidores do Crescimento/fisiologia , Glicoproteínas de Membrana/fisiologia , Peptídeos/fisiologia , Transdução de Sinais/imunologia , Transferência Adotiva , Animais , Anticorpos Bloqueadores/administração & dosagem , Antígenos CD/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Antígeno B7-H1 , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/transplante , Diabetes Mellitus Tipo 1/genética , Feminino , Ligantes , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Peptídeos/deficiência , Peptídeos/imunologia , Receptor de Morte Celular Programada 1 , Ligação Proteica/imunologia , Transdução de Sinais/genética
13.
J Immunol ; 186(1): 121-31, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21131428

RESUMO

Antagonism of CXCR4 disrupts the interaction between the CXCR4 receptor on hematopoietic stem cells (HSCs) and the CXCL12 expressed by stromal cells in the bone marrow, which subsequently results in the shedding of HSCs to the periphery. Because of their profound immunomodulatory effects, HSCs have emerged as a promising therapeutic strategy for autoimmune disorders. We sought to investigate the immunomodulatory role of mobilized autologous HSCs, via target of the CXCR4-CXL12 axis, to promote engraftment of islet cell transplantation. Islets from BALB/c mice were transplanted beneath the kidney capsule of hyperglycemic C57BL/6 mice, and treatment of recipients with CXCR4 antagonist resulted in mobilization of HSCs and in prolongation of islet graft survival. Addition of rapamycin to anti-CXCR4 therapy further promoted HSC mobilization and islet allograft survival, inducing a robust and transferable host hyporesponsiveness, while administration of an ACK2 (anti-CD117) mAb halted CXCR4 antagonist-mediated HSC release and restored allograft rejection. Mobilized HSCs were shown to express high levels of the negative costimulatory molecule programmed death ligand 1 (PD-L1), and HSCs extracted from wild-type mice, but not from PD-L1 knockout mice, suppressed the in vitro alloimmune response. Moreover, HSC mobilization in PD-L1 knockout mice failed to prolong islet allograft survival. Targeting the CXCR4-CXCL12 axis thus mobilizes autologous HSCs and promotes long-term survival of islet allografts via a PD-L1-mediated mechanism.


Assuntos
Antígeno B7-1/fisiologia , Quimiocina CXCL12/antagonistas & inibidores , Marcação de Genes , Sobrevivência de Enxerto/imunologia , Transplante de Células-Tronco Hematopoéticas , Transplante das Ilhotas Pancreáticas/imunologia , Glicoproteínas de Membrana/fisiologia , Peptídeos/fisiologia , Receptores CXCR4/antagonistas & inibidores , Animais , Antígeno B7-1/genética , Antígeno B7-H1 , Benzilaminas , Quimiocina CXCL12/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Ciclamos , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/cirurgia , Marcação de Genes/métodos , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/genética , Compostos Heterocíclicos/farmacologia , Transplante das Ilhotas Pancreáticas/patologia , Masculino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Peptídeos/deficiência , Peptídeos/genética , Receptores CXCR4/metabolismo , Transplante Homólogo
14.
J Exp Med ; 203(4): 883-95, 2006 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-16606670

RESUMO

Programmed death 1 (PD-1), an inhibitory receptor expressed on activated lymphocytes, regulates tolerance and autoimmunity. PD-1 has two ligands: PD-1 ligand 1 (PD-L1), which is expressed broadly on hematopoietic and parenchymal cells, including pancreatic islet cells; and PD-L2, which is restricted to macrophages and dendritic cells. To investigate whether PD-L1 and PD-L2 have synergistic or unique roles in regulating T cell activation and tolerance, we generated mice lacking PD-L1 and PD-L2 (PD-L1/PD-L2(-/-) mice) and compared them to mice lacking either PD-L. PD-L1 and PD-L2 have overlapping functions in inhibiting interleukin-2 and interferon-gamma production during T cell activation. However, PD-L1 has a unique and critical role in controlling self-reactive T cells in the pancreas. Our studies with bone marrow chimeras demonstrate that PD-L1/PD-L2 expression only on antigen-presenting cells is insufficient to prevent the early onset diabetes that develops in PD-L1/PD-L2(-/-) non-obese diabetic mice. PD-L1 expression in islets protects against immunopathology after transplantation of syngeneic islets into diabetic recipients. PD-L1 inhibits pathogenic self-reactive CD4+ T cell-mediated tissue destruction and effector cytokine production. These data provide evidence that PD-L1 expression on parenchymal cells rather than hematopoietic cells protects against autoimmune diabetes and point to a novel role for PD-1-PD-L1 interactions in mediating tissue tolerance.


Assuntos
Antígeno B7-1/biossíntese , Tolerância Imunológica , Glicoproteínas de Membrana/biossíntese , Linfócitos T/imunologia , Animais , Antígeno B7-1/genética , Antígeno B7-1/fisiologia , Antígeno B7-H1 , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/genética , Células-Tronco Hematopoéticas/metabolismo , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Interferon gama , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Especificidade de Órgãos/imunologia , Peptídeos/deficiência , Peptídeos/genética , Peptídeos/fisiologia , Proteína 2 Ligante de Morte Celular Programada 1 , Linfócitos T/metabolismo
15.
Gastroenterology ; 140(3): 879-91, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21111741

RESUMO

BACKGROUND & AIMS: Epigenetic alterations have been correlated with field cancerization in human patients, but evidence from experimental models that specific epigenetic changes can initiate cancer has been lacking. Although hormones have been associated with cancer risk, the mechanisms have not been determined. The peptide hormone gastrin exerts a suppressive effect on antral gastric carcinogenesis. METHODS: N-methyl-N-nitrosourea (MNU)-dependent gastric cancer was investigated in hypergastrinemic (INS-GAS), gastrin-deficient (GAS(-/-)), Tff1-deficient (Tff1(+/-)), and wild-type (WT) mice. Epigenetic alterations of the trefoil factor 1 (TFF1) tumor suppressor gene were evaluated in vitro and in vivo. RESULTS: Human intestinal-type gastric cancers in the antrum exhibited progressive TFF1 repression and promoter hypermethylation. Mice treated with MNU exhibited a field defect characterized by widespread Tff1 repression associated with histone H3 lysine 9 methylation and H3 deacetylation at the Tff1 promoter in epithelial cells. In MNU-induced advanced cancers, DNA methylation at the Tff1 promoter was observed. Tumor induction and Tff1 repression were increased in MNU-treated mice by Helicobacter infection. Hypergastrinemia suppressed MNU-dependent tumor initiation and progression in a manner that correlated with gene silencing and epigenetic alterations of Tff1. In contrast, homozygous gastrin-deficient and heterozygous Tff1-deficient mice showed enhanced MNU-dependent field defects and cancer initiation compared with WT mice. In gastric cancer cells, gastrin stimulation partially reversed the epigenetic silencing in the TFF1 promoter. CONCLUSIONS: Initiation of antral gastric cancer is associated with progressive epigenetic silencing of TFF1, which can be suppressed by the hormone gastrin.


Assuntos
Transformação Celular Neoplásica/genética , Gastrinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Peptídeos/genética , Neoplasias Gástricas/prevenção & controle , Proteínas Supressoras de Tumor/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Montagem e Desmontagem da Cromatina , Metilação de DNA , Modelos Animais de Doenças , Feminino , Gastrinas/deficiência , Gastrinas/genética , Infecções por Helicobacter/genética , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter felis/patogenicidade , Histonas/metabolismo , Humanos , Masculino , Metilnitrosoureia , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Peptídeos/deficiência , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Fatores de Tempo , Transfecção , Fator Trefoil-1 , Proteínas Supressoras de Tumor/metabolismo
16.
Eur J Immunol ; 41(4): 1165-75, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21413005

RESUMO

Increased remissions in multiple sclerosis (MS) during pregnancy suggest that elevated levels of sex steroids exert immunoregulatory activity. Estrogen (E2=17ß-estradiol) protects against experimental autoimmune encephalomyelitis (EAE), but the cellular basis for E2-induced protection remains unclear. Studies demonstrate that depletion of B cells prior to induction of EAE exacerbates disease severity, implicating regulatory B cells. We thus evaluated pathogenic and E2-induced protective mechanisms in B-cell-deficient (µMT(-/-)) mice. EAE-protective effects of E2 were abrogated in µMT(-/-) mice, with no reduction in disease severity, cellular infiltration or pro-inflammatory factors in the central nervous system compared to untreated controls. E2 treatment of WT mice selectively upregulated expression of PD-L1 on B cells and increased the percentage of IL-10-producing CD1d(high) CD5(+) regulatory B cells. Upregulation of PD-L1 was critical for E2-mediated protection since E2 did not inhibit EAE in PD-L1(-/-) mice. Direct treatment of B cells with E2 significantly reduced proliferation of MOG(35-55)-specific T cells that required estrogen receptor-α (ERα). These results demonstrate, for the first time, a requirement for B cells in E2-mediated protection against EAE involving direct E2 effects on regulatory B cells mediated through ERα and the PD-1/PD-L1 negative co-stimulatory pathway. E2-primed B cells may represent an important regulatory mechanism in MS and have strong implications for women receiving current MS therapies that cause B-cell depletion.


Assuntos
Linfócitos B/imunologia , Encefalomielite Autoimune Experimental/imunologia , Estrogênios/imunologia , Animais , Antígenos CD1d/imunologia , Antígeno B7-1/imunologia , Antígeno B7-H1 , Antígenos CD5/imunologia , Células Cultivadas , Estradiol/imunologia , Feminino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/deficiência , Peptídeos/imunologia , Baço/imunologia
17.
Hepatology ; 54(1): 216-28, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21503939

RESUMO

UNLABELLED: Ischemia/reperfusion (I/R) injury remains a key risk factor significantly affecting morbidity and mortality after liver transplantation (LT). B7 homolog 1 (B7-H1), a recently identified member of the B7 family, is known to play important roles in regulating local immune responses. We hypothesized that B7-H1 plays crucial roles during innate immune responses induced by hepatic I/R injury, and using B7-H1 knockout (KO) liver grafts, we tested this hypothesis in the mouse LT model with 24 hours of cold storage. Cold I/R injury in wild type (WT)-to-WT LT enhanced constitutive B7-H1 expression on dendritic cells and sinusoidal endothelial cells and promptly induced B7-H1 on hepatocytes. When B7-H1 KO liver grafts were transplanted into WT recipients, serum alanine aminotransferase (ALT) and graft necrosis levels were significantly higher than those after WT-to-WT LT. Augmented tissue injury in B7-H1 KO grafts was associated with increased frequencies and absolute numbers of graft CD3(+) T cells (particularly CD8(+) T cells). B7-H1 KO grafts had significantly fewer annexin V(+) CD8(+) T cells, and this indicated a failure to delete infiltrating CD8(+) T cells. To evaluate the relative contributions of parenchymal cell and bone marrow-derived cell (BMDC) B7-H1 expression, we generated and transplanted into WT recipients chimeric liver grafts lacking B7-H1 on parenchymal cells or BMDCs. A selective B7-H1 deficiency on parenchymal cells or BMDCs resulted in similar levels of ALT and liver injury, and this suggested that parenchymal cell and BMDC B7-H1 expression was involved in liver damage control. Human livers up-regulated B7-H1 expression after LT. CONCLUSION: The study demonstrates that graft tissue expression of B7-H1 plays a critical role in regulating inflammatory responses during LT-induced hepatic I/R injury, and negative coregulatory signals may have an important function in hepatic innate immune responses.


Assuntos
Antígeno B7-1/metabolismo , Isquemia Fria/efeitos adversos , Transplante de Fígado , Glicoproteínas de Membrana/metabolismo , Peptídeos/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose , Antígeno B7-1/genética , Antígeno B7-H1 , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Masculino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Necrose , Peptídeos/deficiência , Peptídeos/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
18.
Blood ; 116(13): 2266-76, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20574047

RESUMO

Oral tolerance is a key feature of intestinal immunity, generating systemic tolerance to fed antigens. However, the molecular mechanism mediating oral tolerance remains unclear. In this study, we examined the role of the B7 family members of costimulatory molecules in the establishment of oral tolerance. Deficiencies of B7-H1 and B7-DC abrogated the oral tolerance, accompanied by enhanced antigen-specific CD4(+) T-cell response and IgG(1) production. Mesenteric lymph node (MLN) dendritic cells (DCs) displayed higher levels of B7-H1 and B7-DC than systemic DCs, whereas they showed similar levels of CD80, CD86, and B7-H2. MLN DCs enhanced the antigen-specific generation of CD4(+)Foxp3(+) inducible regulatory T cells (iT(regs)) from CD4(+)Foxp3(-) T cells rather than CD4(+) effector T cells (T(eff)) relative to systemic DCs, owing to the dominant expression of B7-H1 and B7-DC. Furthermore, the antigen-specific conversion of CD4(+)Foxp3(-) T cells into CD4(+)Foxp3(+) iT(regs) occurred in MLNs greater than in peripheral organs during oral tolerance under steady-state conditions, and such conversion required B7-H1 and B7-DC more than other B7 family members, whereas it was severely impaired under inflammatory conditions. In conclusion, our findings suggest that B7-H1 and B7-DC expressed on MLN DCs are essential for establishing oral tolerance through the de novo generation of antigen-specific CD4(+)Foxp3(+) iT(regs).


Assuntos
Antígeno B7-1/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Tolerância Imunológica , Glicoproteínas de Membrana/metabolismo , Peptídeos/metabolismo , Linfócitos T Reguladores/imunologia , Administração Oral , Animais , Apresentação de Antígeno , Antígenos/administração & dosagem , Antígeno B7-1/genética , Antígeno B7-H1 , Fatores de Transcrição Forkhead/metabolismo , Imunoglobulina G/biossíntese , Técnicas In Vitro , Linfonodos/citologia , Linfonodos/imunologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Mesentério/citologia , Mesentério/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Peptídeos/deficiência , Peptídeos/genética , Proteína 2 Ligante de Morte Celular Programada 1 , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Linfócitos T Reguladores/metabolismo
19.
J Surg Res ; 175(2): 278-88, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21601882

RESUMO

BACKGROUND AND AIM: Recently, the cancer stem cells (CSCs) theory has been proposed, and CD133 has been suggested as a potential marker of CSCs in various cancer types. In the present study, we aimed evaluate CD133 as a potential marker of colorectal CSCs and, for this purpose, isolated CD133(+) and CD133(-) cells from a single colorectal cancer cell line, and compared their features, especially related to the tumor-forming and differentiation abilities, and the sensitivity to chemotherapy. METHODS AND RESULTS: CD133(+) cells had higher in vivo tumor-forming ability than CD133(-) cells, and in culture, they progressively differentiated into CD133(-) cells, but not vice-versa. On the other hand, CD133(-) cells were more resistant to 5-fluorouracil (FU) treatment than CD133(+) cells, and it was found to be dependent on the higher expression of ß1-integrins, and consequently, higher ability to bind collagen. Disruption of the ß1-integrin function abrogated the chemoresistance. CONCLUSION: From the present results, we concluded that colorectal cancer CD133(+) cells, although showing some features of CSCs, are not more resistant to 5-FU than CD133(-) cells. Therefore, definite conclusions can not be drawn yet, but it is strongly suggestive that CD133 should not be used as a single CSC marker of colorectal cancer.


Assuntos
Antígenos CD/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/imunologia , Fluoruracila/uso terapêutico , Glicoproteínas/deficiência , Glicoproteínas/metabolismo , Integrina beta1/fisiologia , Peptídeos/deficiência , Peptídeos/metabolismo , Transdução de Sinais/fisiologia , Antígeno AC133 , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/imunologia , Adenocarcinoma/fisiopatologia , Antígenos CD/genética , Antimetabólitos Antineoplásicos/uso terapêutico , Apoptose/fisiologia , Biomarcadores Tumorais/imunologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/patologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/fisiopatologia , Glicoproteínas/genética , Humanos , Peptídeos/genética , Resultado do Tratamento
20.
Arterioscler Thromb Vasc Biol ; 31(5): 1100-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21393583

RESUMO

OBJECTIVE: Programmed cell death-1 (PD-1) is a member of the CD28 superfamily that delivers negative signals on interaction with its 2 ligands, PD-L1 and PD-L2. We studied the contribution of the PD-1 pathway to regulation of T cells that promote atherosclerotic lesion formation and inflammation. METHODS AND RESULTS: We show that compared with Ldlr-/- control mice, Pd1-/-Ldlr-/- mice developed larger lesions with more abundant CD4+ and CD8+ T cells and macrophages, accompanied by higher levels of serum tumor necrosis factor-α. Iliac lymph node T cells from Pd1-/-Ldlr-/- mice proliferated more to αCD3 or oxidized low-density lipoprotein stimulation compared with controls. CD8+ T cells from Pd1-/-Ldlr-/- mice displayed more cytotoxic activity compared with controls in vivo and in vitro. Administration of a blocking anti-PD-1 antibody increased lesional inflammation in hypercholesterolemic Ldlr-/- mice with more lesional T cells and more activated T cells in paraaortic lymph nodes. The changes in lesional T-cell content when PD-1 was absent or blocked were also observed in bone marrow chimeric Ldlr-/- mice lacking PD-L1 and PD-L2 on hematopoietic cells. CONCLUSIONS: PD-1 has an important role in downregulating proatherogenic T-cell responses, and blockade of this molecule for treatment of viral infections or cancer may increase risk of cardiovascular complications.


Assuntos
Antígenos de Superfície/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Aterosclerose/metabolismo , Inflamação/metabolismo , Ativação Linfocitária , Subpopulações de Linfócitos T/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Antígenos de Superfície/genética , Apoptose , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/patologia , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígeno B7-H1 , Transplante de Medula Óssea , Complexo CD3/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Genes Codificadores dos Receptores de Linfócitos T , Genótipo , Hipercolesterolemia/genética , Hipercolesterolemia/imunologia , Hipercolesterolemia/metabolismo , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Lipoproteínas LDL/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/deficiência , Peptídeos/genética , Fenótipo , Proteína 2 Ligante de Morte Celular Programada 1 , Receptor de Morte Celular Programada 1 , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA