Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Genet ; 17(6): e1009603, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34143769

RESUMO

The inability to maintain a strictly regulated endo(lyso)somal acidic pH through the proton-pumping action of the vacuolar-ATPases (v-ATPases) has been associated with various human diseases including heritable connective tissue disorders. Autosomal recessive (AR) cutis laxa (CL) type 2C syndrome is associated with genetic defects in the ATP6V1E1 gene and is characterized by skin wrinkles or loose redundant skin folds with pleiotropic systemic manifestations. The underlying pathological mechanisms leading to the clinical presentations remain largely unknown. Here, we show that loss of atp6v1e1b in zebrafish leads to early mortality, associated with craniofacial dysmorphisms, vascular anomalies, cardiac dysfunction, N-glycosylation defects, hypotonia, and epidermal structural defects. These features are reminiscent of the phenotypic manifestations in ARCL type 2C patients. Our data demonstrates that loss of atp6v1e1b alters endo(lyso)somal protein levels, and interferes with non-canonical v-ATPase pathways in vivo. In order to gain further insights into the processes affected by loss of atp6v1e1b, we performed an untargeted analysis of the transcriptome, metabolome, and lipidome in early atp6v1e1b-deficient larvae. We report multiple affected pathways including but not limited to oxidative phosphorylation, sphingolipid, fatty acid, and energy metabolism together with profound defects on mitochondrial respiration. Taken together, our results identify complex pathobiological effects due to loss of atp6v1e1b in vivo.


Assuntos
Anormalidades Múltiplas/genética , Cútis Laxa/genética , Células Epiteliais/metabolismo , Pele/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética , Proteínas de Peixe-Zebra/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Animais , Cútis Laxa/metabolismo , Cútis Laxa/patologia , Modelos Animais de Doenças , Endossomos/metabolismo , Endossomos/patologia , Células Epiteliais/patologia , Regulação da Expressão Gênica , Humanos , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Lipidômica , Longevidade/genética , Lisossomos/metabolismo , Lisossomos/patologia , Metaboloma/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Fosforilação Oxidativa , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Pele/patologia , Síndrome , Transcriptoma , ATPases Vacuolares Próton-Translocadoras/deficiência , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/deficiência
2.
Front Immunol ; 10: 1911, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31456807

RESUMO

Proper orchestration of T lymphocyte development is critical, as T cells underlie nearly all responses of the adaptive immune system. Developing thymocytes differentiate in response to environmental cues carried from cell surface receptors to the nucleus, shaping a distinct transcriptional program that defines their developmental outcome. Our recent work has identified a previously undescribed role for the vacuolar ATPase (V-ATPase) in facilitating the development of murine thymocytes progressing toward the CD4+ and CD8+ αß T cell lineages. Vav1Cre recombinase-mediated deletion of the a2 isoform of the V-ATPase (a2V) in mouse hematopoietic cells leads to a specific and profound loss of peripheral CD4+ and CD8+ αß T cells. Utilizing T cell-restricted LckCre and CD4Cre strains, we further traced this deficiency to the thymus and found that a2V plays a cell-intrinsic role throughout intrathymic development. Loss of a2V manifests as a partial obstruction in the double negative stage of T cell development, and later, a near complete failure of positive selection. These data deepen our understanding of the biological mechanisms that orchestrate T cell development and lend credence to the recent focus on V-ATPase as a potential chemotherapeutic target to combat proliferative potential in T cell lymphoblastic leukemias and autoimmune disease.


Assuntos
Linfopoese , Linfócitos T/fisiologia , Timócitos/fisiologia , Timo/citologia , Timo/enzimologia , ATPases Vacuolares Próton-Translocadoras/fisiologia , Animais , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Feminino , Deleção de Genes , Leucopenia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor Notch1/metabolismo , Transdução de Sinais , Timo/imunologia , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética
3.
J Exp Med ; 214(12): 3707-3729, 2017 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-29127204

RESUMO

The biogenesis of the multi-subunit vacuolar-type H+-ATPase (V-ATPase) is initiated in the endoplasmic reticulum with the assembly of the proton pore V0, which is controlled by a group of assembly factors. Here, we identify two hemizygous missense mutations in the extracellular domain of the accessory V-ATPase subunit ATP6AP2 (also known as the [pro]renin receptor) responsible for a glycosylation disorder with liver disease, immunodeficiency, cutis laxa, and psychomotor impairment. We show that ATP6AP2 deficiency in the mouse liver caused hypoglycosylation of serum proteins and autophagy defects. The introduction of one of the missense mutations into Drosophila led to reduced survival and altered lipid metabolism. We further demonstrate that in the liver-like fat body, the autophagic dysregulation was associated with defects in lysosomal acidification and mammalian target of rapamycin (mTOR) signaling. Finally, both ATP6AP2 mutations impaired protein stability and the interaction with ATP6AP1, a member of the V0 assembly complex. Collectively, our data suggest that the missense mutations in ATP6AP2 lead to impaired V-ATPase assembly and subsequent defects in glycosylation and autophagy.


Assuntos
Autofagia , Proteínas de Drosophila/genética , Genes Ligados ao Cromossomo X , Proteínas de Membrana/genética , Mutação/genética , ATPases Translocadoras de Prótons/genética , Receptores de Superfície Celular/genética , ATPases Vacuolares Próton-Translocadoras/genética , Adolescente , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas Sanguíneas/metabolismo , Encéfalo/embriologia , Encéfalo/patologia , Cútis Laxa/complicações , Cútis Laxa/patologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Degradação Associada com o Retículo Endoplasmático , Fibroblastos/patologia , Glicosilação , Humanos , Lactente , Lipídeos/química , Fígado/patologia , Hepatopatias/complicações , Hepatopatias/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional , ATPases Translocadoras de Prótons/deficiência , ATPases Translocadoras de Prótons/metabolismo , Transtornos Psicomotores/complicações , Transtornos Psicomotores/patologia , Receptores de Superfície Celular/química , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/metabolismo , ATPases Vacuolares Próton-Translocadoras/química , ATPases Vacuolares Próton-Translocadoras/deficiência , Adulto Jovem
4.
J Biol Chem ; 290(41): 25045-61, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26272612

RESUMO

GLP1 activates its receptor, GLP1R, to enhance insulin secretion. The activation and transduction of GLP1R requires complex interactions with a host of accessory proteins, most of which remain largely unknown. In this study, we used membrane-based split ubiquitin yeast two-hybrid assays to identify novel GLP1R interactors in both mouse and human islets. Among these, ATP6ap2 (ATPase H(+)-transporting lysosomal accessory protein 2) was identified in both mouse and human islet screens. ATP6ap2 was shown to be abundant in islets including both alpha and beta cells. When GLP1R and ATP6ap2 were co-expressed in beta cells, GLP1R was shown to directly interact with ATP6ap2, as assessed by co-immunoprecipitation. In INS-1 cells, overexpression of ATP6ap2 did not affect insulin secretion; however, siRNA knockdown decreased both glucose-stimulated and GLP1-induced insulin secretion. Decreases in GLP1-induced insulin secretion were accompanied by attenuated GLP1 stimulated cAMP accumulation. Because ATP6ap2 is a subunit required for V-ATPase assembly of insulin granules, it has been reported to be involved in granule acidification. In accordance with this, we observed impaired insulin granule acidification upon ATP6ap2 knockdown but paradoxically increased proinsulin secretion. Importantly, as a GLP1R interactor, ATP6ap2 was required for GLP1-induced Ca(2+) influx, in part explaining decreased insulin secretion in ATP6ap2 knockdown cells. Taken together, our findings identify a group of proteins that interact with the GLP1R. We further show that one interactor, ATP6ap2, plays a novel dual role in beta cells, modulating both GLP1R signaling and insulin processing to affect insulin secretion.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Receptores de Superfície Celular/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Células CHO , Cálcio/metabolismo , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Técnicas de Silenciamento de Genes , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Humanos , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Camundongos , Ligação Proteica , ATPases Translocadoras de Prótons/deficiência , ATPases Translocadoras de Prótons/genética , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética
5.
J Leukoc Biol ; 96(4): 601-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24970860

RESUMO

HIV is known to subvert cellular machinery to enhance its replication. Recently, HIV has been reported to enhance TC renin expression. We hypothesized that HIV induces and maintains high renin expression to promote its own replication in TCs. Renin enhanced HIV replication in TCs in a dose-dependent manner. (P)RR-deficient TCs, as well as those lacking renin, displayed attenuated NF-κB activity and HIV replication. TCs treated with renin and Hpr displayed activation of the (P)RR-PLZF protein signaling cascade. Renin, HIV, and Hpr activated the PI3K pathway. Both renin and Hpr cleaved Agt (a renin substrate) to Ang I and also cleaved Gag polyproteins (protease substrate) to p24. Furthermore, aliskiren, a renin inhibitor, reduced renin- and Hpr-induced cleavage of Agt and Gag polyproteins. These findings indicate that renin contributes to HIV replication in TCs via the (P)RR-PLZF signaling cascade and through cleavage of the Gag polyproteins.


Assuntos
HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Renina/farmacologia , Linfócitos T/metabolismo , Linfócitos T/virologia , Replicação Viral/efeitos dos fármacos , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica , Proteólise/efeitos dos fármacos , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/metabolismo , Renina/metabolismo , Transdução de Sinais , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/metabolismo
6.
J Clin Invest ; 123(10): 4219-31, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24051376

RESUMO

Inactivation of the B1 proton pump subunit (ATP6V1B1) in intercalated cells (ICs) leads to type I distal renal tubular acidosis (dRTA), a disease associated with salt- and potassium-losing nephropathy. Here we show that mice deficient in ATP6V1B1 (Atp6v1b1-/- mice) displayed renal loss of NaCl, K+, and water, causing hypovolemia, hypokalemia, and polyuria. We demonstrated that NaCl loss originated from the cortical collecting duct, where activity of both the epithelial sodium channel (ENaC) and the pendrin/Na(+)-driven chloride/bicarbonate exchanger (pendrin/NDCBE) transport system was impaired. ENaC was appropriately increased in the medullary collecting duct, suggesting a localized inhibition in the cortex. We detected high urinary prostaglandin E2 (PGE2) and ATP levels in Atp6v1b1-/- mice. Inhibition of PGE2 synthesis in vivo restored ENaC protein levels specifically in the cortex. It also normalized protein levels of the large conductance calcium-activated potassium channel and the water channel aquaporin 2, and improved polyuria and hypokalemia in mutant mice. Furthermore, pharmacological inactivation of the proton pump in ß-ICs induced release of PGE2 through activation of calcium-coupled purinergic receptors. In the present study, we identified ATP-triggered PGE2 paracrine signaling originating from ß-ICs as a mechanism in the development of the hydroelectrolytic imbalance associated with dRTA. Our data indicate that in addition to principal cells, ICs are also critical in maintaining sodium balance and, hence, normal vascular volume and blood pressure.


Assuntos
Túbulos Renais Coletores/metabolismo , Potássio na Dieta/sangue , Sódio na Dieta/sangue , Equilíbrio Hidroeletrolítico , Trifosfato de Adenosina/metabolismo , Animais , Aquaporina 2/metabolismo , Dinoprostona/metabolismo , Canais Epiteliais de Sódio/metabolismo , Técnicas In Vitro , Medula Renal/citologia , Medula Renal/metabolismo , Túbulos Renais Coletores/citologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Camundongos , Camundongos Knockout , Comunicação Parácrina , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética
7.
PLoS One ; 8(4): e58599, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23577057

RESUMO

Periodontal disease affects about 80% of adults in America, and is characterized by oral bacterial infection-induced gingival inflammation, oral bone resorption, and tooth loss. Periodontitis is also associated with other diseases such as rheumatoid arthritis, diabetes, and heart disease. Although many efforts have been made to develop effective therapies for this disease, none have been very effective and there is still an urgent need for better treatments and preventative strategies. Herein we explored for the first time the possibility that adeno-associated virus (AAV)-mediated RNAi knockdown could be used to treat periodontal disease with improved efficacy. For this purpose, we used AAV-mediated RNAi knockdown of Atp6i/TIRC7 gene expression to target bone resorption and gingival inflammation simultaneously. Mice were infected with the oral pathogen Porphyromonas gingivalis W50 (P. gingivalis) in the maxillary periodontium to induce periodontitis. We found that Atp6i depletion impaired extracellular acidification and osteoclast-mediated bone resorption. Furthermore, local injection of AAV-shRNA-Atp6i/TIRC7 into the periodontal tissues in vivo protected mice from P. gingivalis infection-stimulated bone resorption by >85% and decreased the T-cell number in periodontal tissues. Notably, AAV-mediated Atp6i/TIRC7 knockdown also reduced the expression of osteoclast marker genes and inflammation-induced cytokine genes. Atp6i(+/-) mice with haploinsufficiency were similarly protected from P. gingivalis infection-stimulated bone loss and gingival inflammation. This suggests that AAV-shRNA-Atp6i/TIRC7 therapeutic treatment may significantly improve the health of millions who suffer from P. gingivalis-mediated periodontal disease.


Assuntos
Reabsorção Óssea/prevenção & controle , Haploinsuficiência , Doenças Periodontais/genética , Doenças Periodontais/terapia , Interferência de RNA , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética , Animais , Reabsorção Óssea/complicações , Reabsorção Óssea/genética , Contagem de Células , Dependovirus/genética , Modelos Animais de Doenças , Espaço Extracelular/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Concentração de Íons de Hidrogênio , Inflamação/complicações , Inflamação/genética , Inflamação/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Osteoclastos/metabolismo , Osteoclastos/patologia , Doenças Periodontais/complicações , Doenças Periodontais/microbiologia , Periodonto/imunologia , Periodonto/metabolismo , Periodonto/microbiologia , Periodonto/patologia , Porphyromonas gingivalis/fisiologia , Linfócitos T/citologia , Transdução Genética
8.
Acta Neuropathol ; 125(3): 439-57, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23315026

RESUMO

X-linked Myopathy with Excessive Autophagy (XMEA) is a childhood onset disease characterized by progressive vacuolation and atrophy of skeletal muscle. We show that XMEA is caused by hypomorphic alleles of the VMA21 gene, that VMA21 is the diverged human ortholog of the yeast Vma21p protein, and that like Vma21p, VMA21 is an essential assembly chaperone of the vacuolar ATPase (V-ATPase), the principal mammalian proton pump complex. Decreased VMA21 raises lysosomal pH which reduces lysosomal degradative ability and blocks autophagy. This reduces cellular free amino acids which leads to downregulation of the mTORC1 pathway, and consequent increased macroautophagy resulting in proliferation of large and ineffective autolysosomes that engulf sections of cytoplasm, merge, and vacuolate the cell. Our results uncover a novel mechanism of disease, namely macroautophagic overcompensation leading to cell vacuolation and tissue atrophy.


Assuntos
Adenosina Trifosfatases/metabolismo , Autofagia/genética , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/prevenção & controle , Doenças Musculares/genética , Doenças Musculares/prevenção & controle , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética , Animais , Células Cultivadas , Humanos , Concentração de Íons de Hidrogênio , Leucina/metabolismo , Doenças por Armazenamento dos Lisossomos/patologia , Lisossomos/genética , Lisossomos/metabolismo , Masculino , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Doenças Musculares/patologia , Mutação/genética , Interferência de RNA/fisiologia , RNA Mensageiro/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Frações Subcelulares/metabolismo , Frações Subcelulares/patologia , Fatores de Tempo , Vacúolos/metabolismo
9.
Am J Physiol Cell Physiol ; 293(1): C199-210, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17392376

RESUMO

An acidic luminal pH in the epididymis contributes to maintaining sperm quiescent during their maturation and storage. The vacuolar H(+)ATPase (V-ATPase), located in narrow and clear cells, is a major contributor to luminal acidification. Mutations in one of the V-ATPase subunits, ATP6v1B1 (B1), cause distal renal tubular acidosis in humans but surprisingly, B1(-/-) mice do not develop metabolic acidosis and are fertile. While B1 is located in the apical membrane of narrow and clear cells, the B2 subunit localizes to subapical vesicles in wild-type mouse, rat and human epididymis. However, a marked increase (84%) in the mean pixel intensity of B2 staining was observed in the apical pole of clear cells by conventional immunofluorescence, and relocalization into their apical membrane was detected by confocal microscopy in B1(-/-) mice compared with B1(+/+). Immunogold electron microscopy showed abundant B2 in the apical microvilli of clear cells in B1(-/-) mice. B2 mRNA expression, determined by real time RT-PCR using laser-microdissected epithelial cells, was identical in both groups. Semiquantitative Western blots from whole epididymis and cauda epididymidis showed no variation of B2 expression. Finally, the luminal pH of the cauda epididymidis was the same in B1(-/-) mice as in B1(+/+) (pH 6.7). These data indicate that whereas overall expression of B2 is not affected in B1(-/-) mice, significant redistribution of B2-containing complexes occurs from intracellular compartments into the apical membrane of clear cells in B1(-/-) mice. This relocation compensates for the absence of functional B1 and maintains the luminal pH in an acidic range that is compatible with fertility.


Assuntos
Membrana Celular/enzimologia , Epididimo/enzimologia , Células Epiteliais/enzimologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Western Blotting , Polaridade Celular , Epididimo/citologia , Epididimo/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Humanos , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Lasers , Masculino , Camundongos , Camundongos Knockout , Microdissecção/métodos , Microscopia Confocal , Microscopia Eletrônica de Transmissão/métodos , Pessoa de Meia-Idade , Transporte Proteico , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética
10.
Am J Physiol Regul Integr Comp Physiol ; 292(5): R2068-76, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17272665

RESUMO

In the skin of zebrafish embryo, the vacuolar H(+)-ATPase (V-ATPase, H(+) pump) distributed mainly in the apical membrane of H(+)-pump-rich cells, which pump internal acid out of the embryo and function similarly to acid-secreting intercalated cells in mammalian kidney. In addition to acid excretion, the electrogenic H(+) efflux via the H(+)-ATPases in the gill apical membrane of freshwater fish was proposed to act as a driving force for Na(+) entry through the apical Na(+) channels. However, convincing molecular physiological evidence in vivo for this model is still lacking. In this study, we used morpholino-modified antisense oligonucleotides to knockdown the gene product of H(+)-ATPase subunit A (atp6v1a) and examined the phenotype of the mutants. The H(+)-ATPase knockdown embryos revealed several abnormalities, including suppression of acid-secretion from skin, growth retardation, trunk deformation, and loss of internal Ca(2+) and Na(+). This finding reveals the critical role of H(+)-ATPase in embryonic acid -secretion and ion balance, as well.


Assuntos
Ácidos/metabolismo , Embrião não Mamífero/metabolismo , ATPases Translocadoras de Prótons/deficiência , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo , Equilíbrio Hidroeletrolítico , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Água Doce , Regulação Enzimológica da Expressão Gênica , Mutação , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , ATPases Translocadoras de Prótons/química , ATPases Translocadoras de Prótons/genética , ATPases Translocadoras de Prótons/metabolismo , Prótons , ATPases Vacuolares Próton-Translocadoras/deficiência , Saco Vitelino/metabolismo , Proteínas de Peixe-Zebra/deficiência
11.
J Immunol ; 173(4): 2342-52, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15294947

RESUMO

The membrane protein T cell immune response cDNA 7 (TIRC7) was recently identified and was shown to play an important role in T cell activation. To characterize the function of TIRC7 in more detail, we generated TIRC7-deficient mice by gene targeting. We observed disturbed T and B cell function both in vitro and in vivo in TIRC7(-/-) mice. Histologically, primary and secondary lymphoid organs showed a mixture of hypo-, hyper-, and dysplastic changes of multiple lymphohemopoietic compartments. T cells from TIRC7(-/-) mice exhibited significantly increased proliferation and expression of IL-2, IFN-gamma, and IL-4 in response to different stimuli. Resting T cells from TIRC7(-/-) mice exhibited decreased CD62L, but increased CD11a and CD44 expression, suggesting an in vivo expansion of memory/effector T cells. Remarkably, activated T cells from TIRC7(-/-) mice expressed lower levels of CTLA-4 in comparison with wild-type cells. B cells from TIRC7-deficient mice exhibited significantly higher in vitro proliferation following stimulation with anti-CD40 Ab or LPS plus IL-4. B cell hyperreactivity was reflected in vivo by elevated serum levels of various Ig classes and higher CD86 expression on B cells. Furthermore, TIRC7 deficiency resulted in an augmented delayed-type hypersensitivity response that was also reflected in increased mononuclear infiltration in the skin obtained from TIRC7-deficient mice food pads. In summary, the data strongly support an important role for TIRC7 in regulating both T and B cell responses.


Assuntos
Linfócitos B/imunologia , Citocinas/imunologia , Ativação Linfocitária/imunologia , Subunidades Proteicas/imunologia , Linfócitos T/imunologia , ATPases Vacuolares Próton-Translocadoras/imunologia , Animais , Formação de Anticorpos/genética , Linfócitos B/enzimologia , Células Cultivadas , Citometria de Fluxo , Marcação de Genes , Hipersensibilidade Tardia/imunologia , Imuno-Histoquímica , Camundongos , Subunidades Proteicas/deficiência , Baço/imunologia , Baço/patologia , Linfócitos T/enzimologia , ATPases Vacuolares Próton-Translocadoras/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA