Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Immunol ; 196(4): 1964-76, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26755821

RESUMO

Immune responses against cancer cells are often hindered by immunosuppressive mechanisms that are developed in the tumor microenvironment. Induction of a hyporesponsive state in tumor Ag-specific T cells is one of the major events responsible for the inability of the adaptive immune system to mount an efficient antitumor response and frequently contributes to lessen the efficacy of immunotherapeutic approaches. Treatment of localized tumors by focused ultrasound (FUS) is a minimally invasive therapy that uses a range of input energy for in situ tumor ablation through the generation of thermal and cavitation effect. Using a murine B16 melanoma tumor model, we show that a variant of FUS that delivers a reduced level of energy at the focal point and generates mild mechanical and thermal stress in target cells has the ability to increase immunogenic presentation of tumor Ags, which results in reversal of tumor-induced T cell tolerance. Furthermore, we show that the combination of nonablative low-energy FUS with an ablative hypofractionated radiation therapy results in synergistic control of primary tumors and leads to a dramatic reduction in spontaneous pulmonary metastases while prolonging recurrence-free survival only in immunocompetent mice.


Assuntos
Melanoma Experimental/imunologia , Neoplasias Cutâneas/imunologia , Linfócitos T/imunologia , Evasão Tumoral/imunologia , Terapia por Ultrassom/métodos , Animais , Terapia Combinada , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Metástase Neoplásica/patologia , Metástase Neoplásica/terapia , Radioterapia , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia
2.
Proc Natl Acad Sci U S A ; 110(24): 9879-84, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23716685

RESUMO

T-cell costimulation and coinhibition generated by engagement of the B7 family and their receptor CD28 family are of central importance in regulating the T-cell response, making these pathways very attractive therapeutic targets. Here we describe HERV-H LTR-associating protein 2 (HHLA2) as a member of the B7 family that shares 10-18% amino acid identity and 23-33% similarity to other human B7 proteins and phylogenetically forms a subfamily with B7x and B7-H3 within the family. HHLA2 is expressed in humans but not in mice, which is unique within the B7 and CD28 families. HHLA2 protein is constitutively expressed on the surface of human monocytes and is induced on B cells after stimulation with LPS and IFN-γ. HHLA2 does not interact with other known members of the CD28 family or the B7 family, but does bind a putative receptor that is constitutively expressed not only on resting and activated CD4 and CD8 T cells but also on antigen-presenting cells. HHLA2 inhibits proliferation of both CD4 and CD8 T cells in the presence of T-cell receptor signaling. In addition, HHLA2 significantly reduces cytokine production by T cells including IFN-γ, TNF-α, IL-5, IL-10, IL-13, IL-17A, and IL-22. Thus, we have identified a unique B7 pathway that is able to inhibit human CD4 and CD8 T-cell proliferation and cytokine production. This unique human T-cell coinhibitory pathway may afford unique strategies for the treatment of human cancers, autoimmune disorders, infection, and transplant rejection and may help to design better vaccines.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunoglobulinas/imunologia , Células 3T3 , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Antígenos B7/genética , Antígenos B7/imunologia , Antígenos B7/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Citoplasma/metabolismo , Evolução Molecular , Citometria de Fluxo , Humanos , Imunoglobulinas/genética , Imunoglobulinas/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
3.
J Immunol ; 190(7): 3806-14, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23455497

RESUMO

B7x (B7-H4 or B7S1) is an inhibitory member of the B7 family of T cell costimulation. It is expressed in low levels in healthy peripheral tissues, such as the lung epithelium, but is overexpressed in a variety of human cancers with negative clinical associations, including metastasis. However, the function of B7x in the context of cancer, whether expressed on cancer cells or on surrounding "host" tissues, has not been elucidated in vivo. We used the 4T1 metastatic breast cancer model and B7x knockout (B7x (-/-)) mice to investigate the effect of host tissue-expressed B7x on cancer. We found that 4T1 cells were B7x negative in vitro and in vivo, and B7x(-/-) mice had significantly fewer lung 4T1 tumor nodules than did wild-type mice. Furthermore, B7x(-/-) mice showed significantly enhanced survival and a memory response to tumor rechallenge. Mechanistic studies revealed that the presence of B7x correlated with reduced general and tumor-specific T cell cytokine responses, as well as with an increased infiltration of immunosuppressive cells, including tumor-associated neutrophils, macrophages, and regulatory T cells, into tumor-bearing lungs. Importantly, tumor-associated neutrophils strongly bound B7x protein and inhibited the proliferation of both CD4 and CD8 T cells. These results suggest that host B7x may enable metastasizing cancer cells to escape local antitumor immune responses through interactions with the innate and adaptive immune systems. Thus, targeting the B7x pathway holds much promise for improving the efficacy of immunotherapy for metastatic cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias Pulmonares/secundário , Inibidor 1 da Ativação de Células T com Domínio V-Set/genética , Animais , Antígenos Ly/metabolismo , Neoplasias da Mama/imunologia , Neoplasias da Mama/mortalidade , Antígeno CD11b/metabolismo , Linhagem Celular Tumoral , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Memória Imunológica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/mortalidade , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/metabolismo , Fenótipo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Inibidor 1 da Ativação de Células T com Domínio V-Set/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set/metabolismo
4.
J Immunol ; 189(6): 3054-63, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22855708

RESUMO

B7x (B7-H4 or B7S1), a member of the B7 family, inhibits in vitro T cell proliferation and cytokine production by binding to an unidentified receptor on activated T cells, but its in vivo function remains largely unclear. We show that B7x protein was expressed in epithelial cells of the lung, but not in lymphoid tissues. To investigate the role of B7x in the lung, we determined the susceptibility of B7x-deficient (B7x(-/-)) mice to a lethal pulmonary infection with Streptococcus pneumoniae. B7x(-/-), but not B7-H3-deficient, mice were significantly more resistant to S. pneumoniae pulmonary infection than their wild-type (Wt) counterparts. B7x(-/-) mice had significantly lower bacterial burdens and levels of inflammatory cytokines in lungs as early as 12 h postinfection. They also had milder immunopathology that was localized in alveolar spaces, whereas Wt mice had severe inflammation that was perivascular. Control of infection in B7x(-/-) mice was associated with a marked increase in activated CD4 and CD8 T cells and fewer neutrophils in lungs, whereas the susceptible Wt mice had the opposite cellular profile. In B7x(-/-)Rag1(-/-) mice that lack T cells, reduction in bacterial burden was no longer observed. Control of S. pneumoniae and the increased survival observed was specific to the lung, because systemically infected B7x(-/-) mice were not resistant to infection. These data indicate that lung-expressed B7x negatively regulates T cells, and that in its absence, in B7x(-/-) mice, an enhanced T cell response contributed to reduced lethality in a pulmonary infection model with S. pneumoniae.


Assuntos
Pneumonia Pneumocócica/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set/fisiologia , Animais , Resistência à Doença/genética , Resistência à Doença/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Tecido Linfoide/imunologia , Tecido Linfoide/microbiologia , Tecido Linfoide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Avaliação de Resultados em Cuidados de Saúde , Pneumonia Pneumocócica/mortalidade , Pneumonia Pneumocócica/patologia , Análise de Sobrevida , Linfócitos T/imunologia , Linfócitos T/microbiologia , Linfócitos T/patologia , Distribuição Tecidual/genética , Distribuição Tecidual/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set/deficiência , Inibidor 1 da Ativação de Células T com Domínio V-Set/genética
5.
J Immunol ; 189(8): 4165-74, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22972920

RESUMO

B7x (B7-H4 or B7S1) is the seventh member of the B7 family, and its in vivo function remains largely unknown. Despite new genetic data linking the B7x gene with autoimmune diseases, how exactly it contributes to peripheral tolerance and autoimmunity is unclear. In this study, we showed that B7x protein was not detected on APCs or T cells in both human and mice, which is unique in the B7 family. Because B7x protein is expressed in some peripheral cells such as pancreatic ß cells, we used a CD8 T cell-mediated diabetes model (AI4αß) in which CD8 T cells recognize an endogenous self-Ag, and found that mice lacking B7x developed more severe diabetes than control AI4αß mice. Conversely, mice overexpressing B7x in the ß cells (Rip-B7xAI4αß) were diabetes free. Furthermore, adoptive transfer of effector AI4αß CD8 T cells induced diabetes in control mice, but not in Rip-B7xAI4αß mice. Mechanistic studies revealed that pathogenic effector CD8 T cells were capable of migrating to the pancreas but failed to robustly destroy tissue when encountering local B7x in Rip-B7xAI4αß mice. Although AI4αß CD8 T cells in Rip-B7xAI4αß and AI4αß mice showed similar cytotoxic function, cell death, and global gene expression profiles, these cells had greater proliferation in AI4αß mice than in RIP-B7xAI4αß mice. These results suggest that B7x in nonlymphoid organs prevents peripheral autoimmunity partially through inhibiting proliferation of tissue-specific CD8 T cells, and that local overexpression of B7x on pancreatic ß cells is sufficient to abolish CD8 T cell-induced diabetes.


Assuntos
Autoantígenos/sangue , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Pâncreas/imunologia , Pâncreas/patologia , Inibidor 1 da Ativação de Células T com Domínio V-Set/sangue , Animais , Linfócitos T CD8-Positivos/transplante , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/prevenção & controle , Resistência à Doença , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Pâncreas/metabolismo , Inibidor 1 da Ativação de Células T com Domínio V-Set/biossíntese , Inibidor 1 da Ativação de Células T com Domínio V-Set/genética
6.
J Immunol ; 185(1): 624-33, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20530261

RESUMO

Mast cells exert protective effects in experimental antiglomerular basement membrane-induced glomerulonephritis (GN), yet the responsible mediators have not been identified. In this study, we investigated the role of mouse mast cell protease (mMCP)-4, the functional homolog of human chymase, using mMCP-4-deficient mice. Compared with wild type animals, mMCP-4-deficient mice exhibited lower proteinuria, blood creatinine, and blood urea nitrogen levels, indicating an aggravating role of mMCP-4. Kidney histology confirmed less severe renal damage in mMCP-4-deficient mice with reduced deposits, glomerular and interstitial cellularity, and fibrosis scores. High amounts of mMCP-4 were detected in renal capsules, but not in the whole kidney, from wild type mice. Its expression in renal capsules was markedly decreased after GN induction, suggesting that locally released enzyme by degranulated mast cells could contribute to the functional and physiopathological hallmarks of GN. Supporting a proinflammatory role, glomerular and interstitial macrophage and T cell infiltration, levels of proinflammatory TNF and MCP-1 mRNA, and the expression of the profibrotic peptide angiotensin II together with type I collagen were markedly downregulated in kidneys of mMCP-4-deficient mice. We conclude that mMCP-4 chymase, contrary to the global anti-inflammatory action of mast cells, aggravates GN by promoting kidney inflammation. These results highlight the complexity of mast cell-mediated inflammatory actions and suggest that chymase inhibition may represent a novel therapeutic target in GN.


Assuntos
Doença Antimembrana Basal Glomerular/enzimologia , Doença Antimembrana Basal Glomerular/patologia , Doenças do Complexo Imune/enzimologia , Doenças do Complexo Imune/patologia , Mediadores da Inflamação/fisiologia , Serina Endopeptidases/fisiologia , Animais , Doença Antimembrana Basal Glomerular/imunologia , Células Cultivadas , Fibrose , Doenças do Complexo Imune/imunologia , Testes de Função Renal , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
7.
PLoS One ; 15(6): e0233578, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32497097

RESUMO

The B7 family represents one of the best-studied subgroups within the Ig superfamily, yet new interactions continue to be discovered. However, this binding promiscuity represents a major challenge for defining the biological contribution of each specific interaction. We developed a strategy for addressing these challenges by combining cell microarray and high-throughput FACS methods to screen for promiscuous binding events, map binding interfaces, and generate functionally selective reagents. Applying this approach to the interactions of mPD-L1 with its receptor mPD-1 and its ligand mB7-1, we identified the binding interface of mB7-1 on mPD-L1 and as a result generated mPD-L1 mutants with binding selectivity for mB7-1 or mPD-1. Next, using a panel of mB7-1 mutants, we mapped the binding sites of mCTLA-4, mCD28 and mPD-L1. Surprisingly, the mPD-L1 binding site mapped to the dimer interface surface of mB7-1, placing it distal from the CTLA-4/CD28 recognition surface. Using two independent approaches, we demonstrated that mPD-L1 and mB7-1 bind in cis, consistent with recent reports from Chaudhri A et al. and Sugiura D et al. We further provide evidence that while CTLA-4 and CD28 do not directly compete with PD-L1 for binding to B7-1, they can disrupt the cis PD-L1:B7-1 complex by reorganizing B7-1 on the cell surface. These observations offer new functional insights into the regulatory mechanisms associated with this group of B7 family proteins and provide new tools to elucidate their function in vitro and in vivo.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Proteínas Mutantes/metabolismo , Animais , Antígenos de Superfície/metabolismo , Antígeno B7-1/genética , Antígeno B7-H1/genética , Sítios de Ligação , Antígenos CD28/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Antígeno CTLA-4/metabolismo , Células HEK293 , Humanos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Transfecção
8.
Biochim Biophys Acta ; 1783(10): 1781-91, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18588921

RESUMO

Human neutrophil granule exocytosis mobilizes a complex set of secretory granules. This involves different combinations of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins to facilitate membrane fusion. The control mechanisms governing the late fusion steps are still poorly understood. Here, we have analyzed SNARE-interacting Sec1/Munc18 (SM) family members. We found that human neutrophils express Munc18-2 and Munc18-3 isoforms and that Munc18-2 interacts with the target-SNARE syntaxin 3. Munc18-2 was associated preferentially with primary granules but could also be found with secondary and tertiary granules, while Munc18-3 was majorily associated with secondary and tertiary granules. Ultrastructural analysis showed that both Munc18-2 and Munc18-3 were often located in close proximity to their respective SNARE-binding partners syntaxin 3 and syntaxin 4. Both isoforms were also found in plasma membrane fractions and in the cytosol, where they associate with cytoskeletal elements. Upon stimulation, Munc18-2 and Munc18-3 redistributed and became enriched on granules and in the plasma membrane. Munc18-2 primary granule exocytosis can be blocked by introduction of Munc18-2-specific antibodies indicating a crucial role in primary granule fusion. Our results suggest that Munc18-2 acts as a regulator of primary granule exocytosis, while Munc18-3 may preferentially regulate the fusion of secondary granules.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Exocitose , Proteínas Munc18/metabolismo , Neutrófilos/metabolismo , Permeabilidade da Membrana Celular , Células Cultivadas , Humanos , Microscopia Eletrônica de Transmissão , Proteínas Munc18/genética , Proteínas Munc18/ultraestrutura , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico
9.
EBioMedicine ; 17: 30-44, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28233730

RESUMO

Programmed Cell Death-1 (PD-1) is an inhibitory immune receptor, which plays critical roles in T cell co-inhibition and exhaustion upon binding to its ligands PD-L1 and PD-L2. We report the crystal structure of the human PD-1 ectodomain and the mapping of the PD-1 binding interface. Mutagenesis studies confirmed the crystallographic interface, and resulted in mutant PD-1 receptors with altered affinity and ligand-specificity. In particular, a high-affinity mutant PD-1 (HA PD-1) exhibited 45 and 30-fold increase in binding to PD-L1 and PD-L2, respectively, due to slower dissociation rates. This mutant (A132L) was used to engineer a soluble chimeric Ig fusion protein for cell-based and in vivo studies. HA PD-1 Ig showed enhanced binding to human dendritic cells, and increased T cell proliferation and cytokine production in a mixed lymphocyte reaction (MLR) assay. Moreover, in an experimental model of murine Lewis lung carcinoma, HA PD-1 Ig treatment synergized with radiation therapy to decrease local and metastatic tumor burden, as well as in the establishment of immunological memory responses. Our studies highlight the value of structural considerations in guiding the design of a high-affinity chimeric PD-1 Ig fusion protein with robust immune modulatory properties, and underscore the power of combination therapies to selectively manipulate the PD-1 pathway for tumor immunotherapy.


Assuntos
Carcinoma Pulmonar de Lewis/terapia , Imunoterapia/métodos , Receptor de Morte Celular Programada 1/química , Animais , Linhagem Celular Tumoral , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/metabolismo , Feminino , Células HEK293 , Humanos , Imunoglobulinas/genética , Imunoglobulinas/imunologia , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Ligação Proteica , Domínios Proteicos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia
10.
Clin Chim Acta ; 457: 12-7, 2016 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-27012514

RESUMO

BACKGROUND: Different types of cancer cells are previously shown to accumulate intracellular cholesterol. However, the data on intracellular cholesterol levels in leukemia cells provide contradictory evidence. Various previous works indicate either increase, decrease or no difference in total cholesterol levels between leukemia cells and healthy peripheral blood mononuclear cells (PBMCs). METHODS: We studied the intracellular cholesterol levels in acute myeloid leukemia (AML) and acute lymphocytic leukemia (ALL) cells and compared with that in PBMCs from the healthy subjects. RESULTS: We observed that the PBMCs from AML (n=7) and ALL (n=7) patients displayed significantly lower intracellular levels of total cholesterol in comparison to PBMCs from the healthy subjects (n=26). Consistent with the patient data the ALL (CCRF-CEM and MOLT-3) and AML (KG-1 and THP-1) cell lines also displayed significantly lower intracellular levels of total cholesterol. We confirmed this observation using multiple methodological approaches. Both ALL and AML cell lines also displayed significantly lower levels of free cholesterol and cholesteryl ester contents in comparison to normal hematopoietic cells. We observed that >90% of the total cholesterol in leukemia cells as well as in normal PBMCs was present in the form of cholesteryl esters. It was also observed that the lower levels of cholesterol in leukemia cells are not affected by exogenous cholesterol availability. CONCLUSIONS: Present study provides convincing evidence to prove that the cellular free cholesterol and cholesteryl ester content is significantly reduced in leukemia cells in comparison to normal hematopoietic cells in circulation. Moreover, it was shown that the lower levels of cholesterol in leukemia cells are not affected by exogenous cholesterol availability.


Assuntos
Colesterol/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Estudos de Casos e Controles , Linhagem Celular Tumoral , Colesterol/sangue , Humanos , Leucemia Mieloide Aguda/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia
11.
Clin Chim Acta ; 452: 129-33, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26549657

RESUMO

Numerous studies have reported alterations in the plasma lipid profiles of leukemia patients. However, there are several inconsistencies in these reports. The present review highlights and compiles findings from different research groups regarding association of plasma lipoprotein levels with the risk of developing leukemia. We have also discussed the clinical significance of plasma lipid profiles in management of leukemia. Furthermore, the potential role of plasma lipids in promoting leukemogenesis is also highlighted.


Assuntos
Biomarcadores Tumorais/sangue , Leucemia/sangue , Lipídeos/sangue , Humanos , Leucemia/diagnóstico , Leucemia/patologia
12.
Clin Chim Acta ; 450: 356-61, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26386164

RESUMO

Human cells can acquire cholesterol from the circulation but also have the ability to synthesize it via de novo cholesterogenesis (DC). Cholesterol absorption and de novo cholesterogenesis are the key processes that modulate cholesterol homeostasis in the human body. The endogenous biosynthesis of cholesterol substantially contributes to the whole-body cholesterol pool. Additionally, dysregulation of this pathway is associated with diverse medical conditions. The present review focuses on our current understanding of the cholesterogenic pathway and the various different factors regulating this pathway. It also highlights dysregulation of this pathway in various physiological and pathological conditions including cardiovascular diseases, type II diabetes, obesity and viral infections.


Assuntos
Colesterol/biossíntese , Animais , Colesterol/metabolismo , Homeostase , Humanos , Terapia de Alvo Molecular
13.
Metabolism ; 63(7): 895-902, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24814684

RESUMO

BACKGROUND: De novo lipogenesis (DNL) is a complex and highly regulated metabolic pathway. In normal conditions DNL converts excess carbohydrate into fatty acids that are then esterified to storage triacylglycerols (TGs). These TGs could later provide energy via ß-oxidation. In human body this pathway is primarily active in liver and adipose tissue. However, it is considered to be a minor contributor to the serum lipid homeostasis. Deregulations in the lipogenic pathway are associated with diverse pathological conditions. SCOPE OF REVIEW: The present review focuses on our current understanding of the lipogenic pathway with special reference to the causes and consequences of aberrant DNL. MAJOR CONCLUSIONS: The deregulation of DNL in the major lipogenic tissues of the human body is often observed in various metabolic anomalies - including obesity, non-alcoholic fatty liver disease and metabolic syndrome. In addition to that de novo lipogenesis is reported to be exacerbated in cancer tissues, virus infected cells etc. These observations suggest that inhibitors of the DNL pathway might serve as therapeutically significant compounds. The effectiveness of these inhibitors in treatment of cancer and obesity has been suggested by previous works. GENERAL SIGNIFICANCE: De novo lipogenesis - which is an intricate and highly regulated pathway - can lead to adverse metabolic consequences when deregulated. Therapeutic targeting of this pathway may open a new window of opportunity for combating various lipogenesis-driven pathological conditions - including obesity, cancer and certain viral infections.


Assuntos
Tecido Adiposo/metabolismo , Metabolismo Energético , Fígado Gorduroso/metabolismo , Lipogênese , Fígado/metabolismo , Síndrome Metabólica/metabolismo , Obesidade/metabolismo , Tecido Adiposo/efeitos dos fármacos , Animais , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Metabolismo Energético/efeitos dos fármacos , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/etiologia , Humanos , Resistência à Insulina , Reguladores do Metabolismo de Lipídeos/uso terapêutico , Lipogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/etiologia , Terapia de Alvo Molecular , Hepatopatia Gordurosa não Alcoólica , Obesidade/tratamento farmacológico , Obesidade/etiologia , Infecções Oportunistas/metabolismo , Infecções Oportunistas/prevenção & controle , Infecções Oportunistas/virologia
14.
Cell Rep ; 6(4): 625-32, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24529703

RESUMO

B7-H1 (PD-L1) on immune cells plays an important role in T cell coinhibition by binding its receptor PD-1. Here, we show that both human and mouse intestinal epithelium express B7-H1 and that B7-H1-deficient mice are highly susceptible to dextran sodium sulfate (DSS)- or trinitrobenzenesulfonic acid (TNBS)-induced gut injury. B7-H1 deficiency during intestinal inflammation leads to high mortality and morbidity, which are associated with severe pathological manifestations in the colon, including loss of epithelial integrity and overgrowth of commensal bacteria. Results from bone marrow chimeric and knockout mice show that B7-H1 expressed on intestinal parenchyma, but not on hematopoietic cells, controls intestinal inflammation in an adaptive immunity-independent fashion. Finally, we demonstrate that B7-H1 dampened intestinal inflammation by inhibiting tumor necrosis factor α (TNF-α) production and by stimulating interleukin 22 secretion from CD11c(+)CD11b(+) lamina propria cells. Thus, our data uncover a mechanism through which intestinal tissue-expressed B7-H1 functions as an essential ligand for innate immune cells to prevent gut inflammation.


Assuntos
Colite/metabolismo , Mucosa Intestinal/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Animais , Células da Medula Óssea/metabolismo , Colite/induzido quimicamente , Colite/imunologia , Humanos , Imunidade Inata , Inflamação/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/genética , Dodecilsulfato de Sódio/toxicidade , Ácido Trinitrobenzenossulfônico/toxicidade , Fator de Necrose Tumoral alfa/metabolismo , Interleucina 22
15.
Cell Rep ; 9(3): 1089-98, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25437562

RESUMO

B7x (B7-H4 or B7S1) is a member of the B7 family that can inhibit T cell function. B7x protein is absent in most normal human tissues and immune cells, but it is overexpressed in human cancers and often correlates with negative clinical outcome. The expression pattern and function of B7x suggest that it may be a potent immunosuppressive pathway in human cancers. Here, we determined the crystal structure of the human B7x immunoglobulin variable (IgV) domain at 1.59 Å resolution and mapped the epitopes recognized by monoclonal antibodies. We developed an in vivo system to screen therapeutic monoclonal antibodies against B7x and found that the clone 1H3 significantly inhibited growth of B7x-expressing tumors in vivo via multiple mechanisms. Furthermore, the surviving mice given 1H3 treatment were resistant to tumor rechallenge. Our data suggest that targeting B7x on tumors is a promising cancer immunotherapy and humanized 1H3 may be efficacious for immunotherapy of human cancers.


Assuntos
Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Inibidor 1 da Ativação de Células T com Domínio V-Set/química , Inibidor 1 da Ativação de Células T com Domínio V-Set/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cristalografia por Raios X , Modelos Animais de Doenças , Mapeamento de Epitopos , Epitopos/química , Epitopos/imunologia , Humanos , Terapia de Imunossupressão , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Camundongos Endogâmicos BALB C , Modelos Moleculares , Neoplasias/patologia , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície , Análise de Sobrevida , Linfócitos T/imunologia , Microambiente Tumoral/efeitos dos fármacos , Inibidor 1 da Ativação de Células T com Domínio V-Set/metabolismo
16.
Discov Med ; 12(63): 119-28, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21878189

RESUMO

T cell costimulatory and coinhibitory pathways are essential orchestrators and regulators of the adaptive immune response. In recent years, the costimulatory CD28 receptor and B7 ligand families have been expanded to include a total of four and seven members, respectively. Several polymorphisms, mutations, and deletions in both regulatory and protein-coding regions of these genes have subsequently been discovered and evaluated for genetic linkage to various human diseases. Here, we review this evidence as we discuss T cell costimulation and coinhibition in the context of genetic susceptibility to autoimmunity, cancer, and other diseases. As we gain further insight into the functional significance and mechanism of these immunoregulatory pathways by both genetic and immunological approaches, these receptors and ligands are poised to become key targets for immunotherapy.


Assuntos
Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Autoimunidade/genética , Autoimunidade/fisiologia , Antígenos B7/genética , Antígenos B7/metabolismo , Antígenos CD28/genética , Antígenos CD28/metabolismo , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
17.
Immunol Rev ; 217: 79-95, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17498053

RESUMO

Inflammatory kidney disease involves a complex network of interactions between resident kidney and infiltrating hematopoietic cells. Mast cells (MCs) are constitutively found in kidneys in small numbers but increase considerably in various renal diseases. While this increase is usually interpreted as a sign of pathological involvement, recent data using MC-deficient animals show their ability to restore kidney homeostasis. In anti-glomerular basement membrane antibody-induced glomerulonephritis, MCs are protective by initiating repair and remodeling functions counteracting the devastating effects of glomerular injury. Protection may also include immunoregulatory capacities to limit autoreactive T-cell responses. MCs also control tubulointerstitial fibrosis by activating tissue remodeling and neutralizing fibrotic factors. Release of mediators by MCs during inflammation, however, could also promote unwanted responses that ultimately lead to destruction of kidney structure, as exemplified by data showing either protection or aggravation in related renal disease models. Similarly, while the action of proteases may initially be beneficial, the generation of fibrosis-promoting angiotensin II by chymase also shows the limits of adaptive responses to achieve homeostasis. Thus, it is likely the physiological context involving the interaction with other cells and inflammatory mediators that determines the final action of MCs in the development of kidney disease.


Assuntos
Mastócitos/imunologia , Nefrite/imunologia , Fibrose , Humanos , Rim/imunologia , Nefrite Intersticial/imunologia , Nefrite Intersticial/patologia
18.
J Immunol ; 176(9): 5607-15, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16622030

RESUMO

Mast cells are detrimental in several inflammatory diseases; however, their physiological roles are also increasingly recognized. Recent data suggest that mast cells may also be involved in renal diseases. We therefore used congenitally mast cell-deficient W/W(v) mice and normal +/+ littermates to assess their role in anti-glomerular basement membrane-induced glomerulonephritis. Following administration of anti-glomerular basement membrane Abs, W/W(v) mice exhibited increased mortality as compared with +/+ mice owing to rapid deterioration of renal function. Reconstitution of the mast cell population in W/W(v) mice restored protection. This was independent of activating FcgammaR, as protection was also obtained using mast cells deficient in FcRgamma. Comparative histological analysis of kidneys showed that deterioration of renal function was caused by the presence of thick layers of subendothelial glomerular deposits in W/W(v) mice, while +/+ mice or mast cell-reconstituted W/W(v) mice showed significantly less. Deposits appeared during the early phase of disease and persisted thereafter, and were accompanied by enhanced macrophage recruitment. Immunohistochemical analysis revealed increased amounts of fibrin and type I collagen in W/W(v) mice, which were also unable to maintain high tissue plasminogen activator and urinary-type plasminogen activator activity in urine in the heterologous phase of disease. Our results indicate that mast cells by their ability to mediate remodeling and repair functions are protective in immune complex-mediated glomerulonephritis.


Assuntos
Glomerulonefrite/patologia , Mastócitos , Animais , Anticorpos/imunologia , Proliferação de Células , Colágeno/metabolismo , Cricetinae , Suscetibilidade a Doenças , Células Endoteliais/citologia , Fibrina/metabolismo , Membrana Basal Glomerular/imunologia , Glomerulonefrite/embriologia , Glomerulonefrite/imunologia , Glomerulonefrite/metabolismo , Macrófagos , Camundongos , Receptores de IgG/metabolismo , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA