Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Exp Rheumatol ; 41(9): 1735-1745, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36700637

RESUMO

OBJECTIVES: PARPs, which are members of the poly(ADP-ribose) polymerase superfamily, promote tumorigenesis and tumour-associated inflammation and are thus therapeutic targets for several cancers. The aim of the present study is to investigate the mechanistic insight into the roles PARPs for inflammation. METHODS: Primary murine macrophages were cultured in the presence or absence of the PARP5 inhibitor NVP-TNKS656 to examine the role of PARP5 for cytokine production. RESULTS: In contrast to the roles of other PARPs for induction of inflammation, we found in the present study that pharmacologic inhibition of PARP5 induces production of inflammatory cytokines in primary murine macrophages. We found that treatment with the PARP5 inhibitor NVP-TNKS656 in macrophages enhanced steady-state and LPS-mediated cytokine production through degradation of IκBα and subsequent nuclear translocation of NF-κB. We also found that pharmacologic inhibition of PARP5 stabilises the adaptor protein 3BP2, a substrate of PARP5, and that accelerated cytokine production induced by PARP5 inhibition was rescued in 3BP2-deleted macrophages. Additionally, we found that LPS increases the expression of 3BP2 and AXIN1, a negative regulator of ß-catenin, through suppression of PARP5 transcripts in macrophages, leading to further activation of cytokine production and inhibition of ß-catenin-mediated cell proliferation, respectively. Lastly, we found that PARP5 inhibition in macrophages promotes osteoclastogenesis through stabilisation of 3BP2 and AXIN1, leading to activation of SRC and suppression of ß-catenin, respectively. CONCLUSIONS: Our results show that pharmacologic inhibition of PARP5 against cancers unexpectedly induces adverse autoinflammatory side effects through activation of innate immunity, unlike inhibition of other PARPs.


Assuntos
Lipopolissacarídeos , beta Catenina , Humanos , Camundongos , Animais , beta Catenina/uso terapêutico , Lipopolissacarídeos/farmacologia , Osteogênese , NF-kappa B/metabolismo , Citocinas/metabolismo , Inflamação , Poli(ADP-Ribose) Polimerase-1/uso terapêutico
2.
J Clin Invest ; 132(7)2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35362478

RESUMO

Dysregulation of Toll-like receptor (TLR) signaling contributes to the pathogenesis of autoimmune diseases. Here, we provide genetic evidence that tankyrase, a member of the poly(ADP-ribose) polymerase (PARP) family, negatively regulates TLR2 signaling. We show that mice lacking tankyrase in myeloid cells developed severe systemic inflammation with high serum inflammatory cytokine levels. We provide mechanistic evidence that tankyrase deficiency resulted in tyrosine phosphorylation and activation of TLR2 and show that phosphorylation of tyrosine 647 within the TIR domain by SRC and SYK kinases was critical for TLR2 stabilization and signaling. Last, we show that the elevated cytokine production and inflammation observed in mice lacking tankyrase in myeloid cells were dependent on the adaptor protein 3BP2, which is required for SRC and SYK activation. These data demonstrate that tankyrase provides a checkpoint on the TLR-mediated innate immune response.


Assuntos
Doenças Autoimunes , Inflamação , Tanquirases , Receptor 2 Toll-Like , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Doenças Autoimunes/genética , Inflamação/genética , Camundongos , Transdução de Sinais , Quinase Syk/metabolismo , Tanquirases/genética , Tanquirases/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo
3.
Blood Adv ; 5(16): 3120-3133, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34406376

RESUMO

How hematopoietic stem cells (HSCs) coordinate their divisional axis and whether this orientation is important for stem cell-driven hematopoiesis is poorly understood. Single-cell RNA sequencing data from patients with Shwachman-Diamond syndrome (SDS), an inherited bone marrow failure syndrome, show that ARHGEF2, a RhoA-specific guanine nucleotide exchange factor and determinant of mitotic spindle orientation, is specifically downregulated in SDS hematopoietic stem and progenitor cells (HSPCs). We demonstrate that transplanted Arhgef2-/- fetal liver and bone marrow cells yield impaired hematopoietic recovery and a production deficit from long-term HSCs, phenotypes that are not the result of differences in numbers of transplanted HSCs, their cell cycle status, level of apoptosis, progenitor output, or homing ability. Notably, these defects are functionally restored in vivo by overexpression of ARHGEF2 or its downstream activated RHOA GTPase. By using live imaging of dividing HSPCs, we show an increased frequency of misoriented divisions in the absence of Arhgef2. ARHGEF2 knockdown in human HSCs also impairs their ability to regenerate hematopoiesis, culminating in significantly smaller xenografts. Together, these data demonstrate a conserved role for Arhgef2 in orienting HSPC division and suggest that HSCs may divide in certain orientations to establish hematopoiesis, the loss of which could contribute to HSC dysfunction in bone marrow failure.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Apoptose , Células da Medula Óssea , Humanos , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fuso Acromático
4.
Front Oncol ; 11: 665273, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34136397

RESUMO

Activity of transcription factors is normally regulated through interaction with other transcription factors, chromatin remodeling proteins and transcriptional co-activators. In distinction to these well-established transcriptional controls of gene expression, we have uncovered a unique activation model of transcription factors between tyrosine kinase ABL and RUNX2, an osteoblastic master transcription factor, for cancer invasion. We show that ABL directly binds to, phosphorylates, and activates RUNX2 through its SH2 domain in a kinase activity-dependent manner and that the complex formation of these proteins is required for expression of its target gene MMP13. Additionally, we show that the RUNX2 transcriptional activity is dependent on the number of its tyrosine residues that are phosphorylated by ABL. In addition to regulation of RUNX2 activity, we show that ABL transcriptionally enhances RUNX2 expression through activation of the bone morphogenetic protein (BMP)-SMAD pathway. Lastly, we show that ABL expression in highly metastatic breast cancer MDA-MB231 cells is associated with their invasive capacity and that ABL-mediated invasion is abolished by depletion of endogenous RUNX2 or MMP13. Our genetic and biochemical evidence obtained in this study contributes to a mechanistic insight linking ABL-mediated phosphorylation and activation of RUNX2 to induction of MMP13, which underlies a fundamental invasive capacity in cancer and is different from the previously described model of transcriptional activation.

5.
Sci Rep ; 11(1): 8502, 2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33875686

RESUMO

Bone is a highly dynamic organ that undergoes remodeling equally regulated by osteoblast-mediated bone formation and osteoclast-mediated bone resorption. To clarify the regulation of osteoblastogenesis, primary murine osteoblasts are required for an in vitro study. Primary osteoblasts are isolated from neonatal calvariae through digestion with collagenase. However, the number of cells collected from one pup is not sufficient for further in vitro experiments, leading to an increase in the use of euthanized pups. We hypothesized that the viscosity of digested calvariae and digestion solution supplemented with collagenase results in cell clumping and reduction of isolated cells from bones. We simply added Benzonase, a genetically engineered endonuclease that shears all forms of DNAs/RNAs, in order to reduce nucleic acid-mediated viscosity. We found that addition of Benzonase increased the number of collected osteoblasts by three fold compared to that without Benzonase through reduction of viscosity. Additionally, Benzonase has no effect on cellular identity and function. The new osteoblast isolation protocol with Benzonase minimizes the number of neonatal pups required for an in vitro study and expands the concept that isolation of other populations of cells including osteocytes that are difficult to be purified could be modified by Benzonase.


Assuntos
Diferenciação Celular , Proliferação de Células , Endonucleases/metabolismo , Osteoblastos/citologia , Osteogênese , Crânio/citologia , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Crânio/metabolismo
6.
J Clin Invest ; 131(7)2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33561012

RESUMO

Ovarian cancer (OC) is the most deadly gynecological malignancy, with unmet clinical need for new therapeutic approaches. The relaxin peptide is a pleiotropic hormone with reproductive functions in the ovary. Relaxin induces cell growth in several types of cancer, but the role of relaxin in OC is poorly understood. Here, using cell lines and xenograft models, we demonstrate that relaxin and its associated GPCR RXFP1 form an autocrine signaling loop essential for OC in vivo tumorigenesis, cell proliferation, and viability. We determined that relaxin signaling activates expression of prooncogenic pathways, including RHO, MAPK, Wnt, and Notch. We found that relaxin is detectable in patient-derived OC tumors, ascites, and serum. Further, inflammatory cytokines IL-6 and TNF-α activated transcription of relaxin via recruitment of STAT3 and NF-κB to the proximal promoter, initiating an autocrine feedback loop that potentiated expression. Inhibition of RXFP1 or relaxin increased cisplatin sensitivity of OC cell lines and abrogated in vivo tumor formation. Finally, we demonstrate that a relaxin-neutralizing antibody reduced OC cell viability and sensitized cells to cisplatin. Collectively, these data identify the relaxin/RXFP1 autocrine loop as a therapeutic vulnerability in OC.


Assuntos
Comunicação Autócrina , Carcinogênese/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Relaxina/metabolismo , Via de Sinalização Wnt , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia
7.
J Immunol ; 205(12): 3300-3310, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33199537

RESUMO

Gout is a painful arthritic inflammatory disease caused by buildup of monosodium urate (MSU) crystals in the joints. Colchicine, a microtubule-depolymerizing agent that is used in prophylaxis and treatment of acute gout flare, alleviates the painful inflammatory response to MSU crystals. Using i.p. and intra-articular mouse models of gout-like inflammation, we found that GEF-H1/GEF-H1/AHRGEF2, a microtubule-associated Rho-GEF, was necessary for the inhibitory effect of colchicine on neutrophil recruitment. GEF-H1 was required for neutrophil polarization in response to colchicine, characterized by uropod formation, accumulation of F-actin and myosin L chain at the leading edge, and accumulation of phosphorylated myosin L chain, flotillin-2, and P-selectin glycoprotein ligand-1 (PSGL-1) in the uropod. Wild-type neutrophils that were pre-exposed to colchicine failed to roll or accumulate on activated endothelial monolayers, whereas GEF-H1 knockout (GEF-H1-/-) neutrophils were unaffected by treatment with colchicine. In vivo, colchicine blocked MSU-induced recruitment of neutrophils to the peritoneum and the synovium in wild-type mice, but not in GEF-H1-/- mice. Inhibition of macrophage IL-1ß production by colchicine was independent of GEF-H1, supporting a neutrophil-intrinsic mode of action. Our results suggest that the anti-inflammatory effects of colchicine in acute gout-like inflammation can be accounted for by inhibition of neutrophil-rolling interactions with the inflamed vasculature and occurs through GEF-H1-dependent neutrophil stimulation by colchicine. These results contribute to our understanding of the therapeutic action of colchicine, and could inform the application of this drug in other conditions.


Assuntos
Colchicina/farmacologia , Gota , Migração e Rolagem de Leucócitos , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos , Fatores de Troca de Nucleotídeo Guanina Rho/imunologia , Actinas/genética , Actinas/imunologia , Animais , Modelos Animais de Doenças , Gota/tratamento farmacológico , Gota/genética , Gota/imunologia , Gota/patologia , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Migração e Rolagem de Leucócitos/genética , Migração e Rolagem de Leucócitos/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Cadeias Leves de Miosina , Neutrófilos/imunologia , Neutrófilos/patologia , Fatores de Troca de Nucleotídeo Guanina Rho/genética
8.
Sci Rep ; 9(1): 3590, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30837567

RESUMO

Genomic rearrangements are a hallmark of cancer biology and progression, allowing cells to rapidly transform through alterations in regulatory structures, changes in expression patterns, reprogramming of signaling pathways, and creation of novel transcripts via gene fusion events. Though functional gene fusions encoding oncogenic proteins are the most dramatic outcomes of genomic rearrangements, we investigated the relationship between rearrangements evidenced by fusion transcripts and local expression changes in cancer using transcriptome data alone. 9,953 gene fusion predictions from 418 primary serious ovarian cancer tumors were analyzed, identifying depletions of gene fusion breakpoints within coding regions of fused genes as well as an N-terminal enrichment of breakpoints within fused genes. We identified 48 genes with significant fusion-associated upregulation and furthermore demonstrate that significant regional overexpression of intact genes in patient transcriptomes occurs within 1 megabase of 78 novel gene fusions that function as central markers of these regions. We reveal that cancer transcriptomes select for gene fusions that preserve protein and protein domain coding potential. The association of gene fusion transcripts with neighboring gene overexpression supports rearrangements as mechanism through which cancer cells remodel their transcriptomes and identifies a new way to utilize gene fusions as indicators of regional expression changes in diseased cells with only transcriptomic data.


Assuntos
Pontos de Quebra do Cromossomo , Regulação Neoplásica da Expressão Gênica , Fusão Gênica , Proteínas de Fusão Oncogênica , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Transcriptoma , Biomarcadores Tumorais/genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Gradação de Tumores
9.
Sci Signal ; 10(503)2017 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-29089450

RESUMO

The PAR-1-MARK pathway controls cell polarity through the phosphorylation of microtubule-associated proteins. Rho-Rac guanine nucleotide exchange factor 2 (ARHGEF2), which activates Ras homolog family member A (RHOA), is anchored to the microtubule network and sequestered in an inhibited state through binding to dynein light chain Tctex-1 type 1 (DYNLT1). We showed in mammalian cells that liver kinase B1 (LKB1) activated the microtubule affinity-regulating kinase 3 (MARK3), which in turn phosphorylated ARHGEF2 at Ser151 This modification disrupted the interaction between ARHGEF2 and DYNLT1 by generating a 14-3-3 binding site in ARHGEF2, thus causing ARHGEF2 to dissociate from microtubules. Phosphorylation of ARHGEF2 by MARK3 stimulated RHOA activation and the formation of stress fibers and focal adhesions, and was required for organized cellular architecture in three-dimensional culture. Protein phosphatase 2A (PP2A) dephosphorylated Ser151 in ARHGEF2 to restore the inhibited state. Thus, we have identified a regulatory switch controlled by MARK3 that couples microtubules to the actin cytoskeleton to establish epithelial cell polarity through ARHGEF2.


Assuntos
Citoesqueleto de Actina/metabolismo , Polaridade Celular/fisiologia , Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Células COS , Chlorocebus aethiops , Dineínas/genética , Dineínas/metabolismo , Adesões Focais/metabolismo , Células HEK293 , Humanos , Fosforilação , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Proteínas Serina-Treonina Quinases/genética , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Serina/metabolismo , Fibras de Estresse/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
10.
J Clin Invest ; 127(7): 2612-2625, 2017 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-28581440

RESUMO

Cleidocranial dysplasia (CCD) is an autosomal dominant human disorder characterized by abnormal bone development that is mainly due to defective intramembranous bone formation by osteoblasts. Here, we describe a mouse strain lacking the E3 ubiquitin ligase RNF146 that shows phenotypic similarities to CCD. Loss of RNF146 stabilized its substrate AXIN1, leading to impairment of WNT3a-induced ß-catenin activation and reduced Fgf18 expression in osteoblasts. We show that FGF18 induces transcriptional coactivator with PDZ-binding motif (TAZ) expression, which is required for osteoblast proliferation and differentiation through transcriptional enhancer associate domain (TEAD) and runt-related transcription factor 2 (RUNX2) transcription factors, respectively. Finally, we demonstrate that adipogenesis is enhanced in Rnf146-/- mouse embryonic fibroblasts. Moreover, mice with loss of RNF146 within the osteoblast lineage had increased fat stores and were glucose intolerant with severe osteopenia because of defective osteoblastogenesis and subsequent impaired osteocalcin production. These findings indicate that RNF146 is required to coordinate ß-catenin signaling within the osteoblast lineage during embryonic and postnatal bone development.


Assuntos
Desenvolvimento Ósseo , Displasia Cleidocraniana/metabolismo , Metabolismo Energético , Osteoblastos/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo , Animais , Proteína Axina/biossíntese , Proteína Axina/genética , Displasia Cleidocraniana/genética , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Humanos , Camundongos , Camundongos Knockout , Osteocalcina/biossíntese , Osteocalcina/genética , Ubiquitina-Proteína Ligases/genética , beta Catenina/genética , beta Catenina/metabolismo
11.
J Clin Invest ; 126(12): 4482-4496, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27797343

RESUMO

Cellular identity in metazoan organisms is frequently established through lineage-specifying transcription factors, which control their own expression through transcriptional positive feedback, while antagonizing the developmental networks of competing lineages. Here, we have uncovered a distinct positive feedback loop that arises from the reciprocal stabilization of the tyrosine kinase ABL and the transcriptional coactivator TAZ. Moreover, we determined that this loop is required for osteoblast differentiation and embryonic skeletal formation. ABL potentiated the assembly and activation of the RUNX2-TAZ master transcription factor complex that is required for osteoblastogenesis, while antagonizing PPARγ-mediated adipogenesis. ABL also enhanced TAZ nuclear localization and the formation of the TAZ-TEAD complex that is required for osteoblast expansion. Last, we have provided genetic data showing that regulation of the ABL-TAZ amplification loop lies downstream of the adaptor protein 3BP2, which is mutated in the craniofacial dysmorphia syndrome cherubism. Our study demonstrates an interplay between ABL and TAZ that controls the mesenchymal maturation program toward the osteoblast lineage and is mechanistically distinct from the established model of lineage-specific maturation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Núcleo Celular/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Núcleo Celular/genética , Querubismo/genética , Querubismo/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , PPAR gama/genética , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas c-abl/genética , Transativadores
12.
J Cell Biol ; 215(1): 107-119, 2016 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-27738004

RESUMO

Leukocyte crawling and transendothelial migration (TEM) are potentiated by shear stress caused by blood flow. The mechanism that couples shear stress to migration has not been fully elucidated. We found that mice lacking GEF-H1 (GEF-H1-/-), a RhoA-specific guanine nucleotide exchange factor (GEF), displayed limited migration and recruitment of neutrophils into inflamed tissues. GEF-H1-/- leukocytes were deficient in in vivo crawling and TEM in the postcapillary venules. We demonstrated that although GEF-H1 deficiency had little impact on the migratory properties of neutrophils under static conditions, shear stress triggered GEF-H1-dependent spreading and crawling of neutrophils and relocalization of GEF-H1 to flotillin-2-rich uropods. Our results identify GEF-H1 as a component of the shear stress response machinery in neutrophils required for a fully competent immune response to bacterial infection.


Assuntos
Movimento Celular , Inflamação/patologia , Neutrófilos/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Estresse Mecânico , Animais , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células HL-60 , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Modelos Biológicos , Músculos/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Polimerização/efeitos dos fármacos , Fatores de Troca de Nucleotídeo Guanina Rho/deficiência , Sepse/patologia
13.
Cell ; 147(6): 1324-39, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22153076

RESUMO

Cherubism is an autosomal-dominant syndrome characterized by inflammatory destructive bony lesions resulting in symmetrical deformities of the facial bones. Cherubism is caused by mutations in Sh3bp2, the gene that encodes the adaptor protein 3BP2. Most identified mutations in 3BP2 lie within the peptide sequence RSPPDG. A mouse model of cherubism develops hyperactive bone-remodeling osteoclasts and systemic inflammation characterized by expansion of the myelomonocytic lineage. The mechanism by which cherubism mutations alter 3BP2 function has remained obscure. Here we show that Tankyrase, a member of the poly(ADP-ribose)polymerase (PARP) family, regulates 3BP2 stability through ADP-ribosylation and subsequent ubiquitylation by the E3-ubiquitin ligase RNF146 in osteoclasts. Cherubism mutations uncouple 3BP2 from Tankyrase-mediated protein destruction, which results in its stabilization and subsequent hyperactivation of the SRC, SYK, and VAV signaling pathways.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Querubismo/metabolismo , Transdução de Sinais , Tanquirases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Querubismo/genética , Modelos Animais de Doenças , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/metabolismo , Osteoclastos/metabolismo , Estabilidade Proteica , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-vav/metabolismo , Deleção de Sequência , Quinase Syk , Tanquirases/genética , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitinação
14.
J Clin Invest ; 121(8): 3244-57, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21765218

RESUMO

A fine balance between bone resorption by osteoclasts and bone formation by osteoblasts maintains bone homeostasis. In patients with cherubism, gain-of-function mutations in 3BP2, which is encoded by SH3-domain binding protein 2 (SH3BP2), cause cystic lesions with activated osteoclasts that lead to craniofacial abnormalities. However, little is known about the function of wild-type 3BP2 in regulating bone homeostasis. Here we have shown that 3BP2 is required for the normal function of both osteoblasts and osteoclasts. Initial analysis showed that Sh3bp2-/-mice developed osteoporosis as a result of reduced bone formation despite the fact that bone resorption was impaired. We demonstrated using reciprocal bone marrow chimeras, a cell-intrinsic defect of the osteoblast and osteoclast compartments in vivo. Further, Sh3bp2-/- osteoblasts failed to mature and form mineralized nodules in vitro, while Sh3bp2-/- osteoclasts spread poorly and were unable to effectively degrade dentine matrix in vitro. Finally, we showed that 3BP2 was required for Abl activation in osteoblasts and Src activation in osteoclasts, and demonstrated that the in vitro defect of each cell type was restored by the respective expression of activated forms of these kinases. These findings reveal an unanticipated role for the 3BP2 adapter protein in osteoblast function and in coordinating bone homeostatic signals in both osteoclast and osteoblast lineages.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Regulação da Expressão Gênica , Osteoclastos/metabolismo , Osteoporose/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Medula Óssea/metabolismo , Reabsorção Óssea , Linhagem da Célula , Integrinas , Masculino , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo
15.
Mol Cell Biol ; 27(8): 3109-22, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17283041

RESUMO

3BP2 is a pleckstrin homology domain- and Src homology 2 (SH2) domain-containing adapter protein that is mutated in the rare human bone disorder cherubism and which has also been implicated in immunoreceptor signaling. However, a function for this protein has yet to be established. Here we show that mice lacking 3BP2 exhibited a perturbation in the peritoneal B1 and splenic marginal-zone B-cell compartments and diminished thymus-independent type 2 antigen response. 3BP2(-/-) B cells demonstrated a proliferation defect in response to antigen receptor cross-linking and a heightened sensitivity to B-cell receptor-induced death via a caspase-3-dependent apoptotic pathway. We show that 3BP2 binds via its SH2 domain to the CD19 signaling complex and is required for optimum Syk phosphorylation and calcium flux.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Formação de Anticorpos/imunologia , Linfócitos B/imunologia , Ativação Linfocitária/imunologia , Timo/imunologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linfócitos B/citologia , Antígenos CD5/imunologia , Contagem de Células , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Regulação da Expressão Gênica , Humanos , Imunização , Camundongos , Camundongos Endogâmicos C57BL , Peritônio/citologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Baço/citologia
16.
J Immunol ; 174(1): 180-94, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15611240

RESUMO

In concert with the TCR, CD28 promotes T cell survival by regulating the expression of the antiapoptotic protein Bcl-x(L). The mechanism by which CD28 mediates the induction of Bcl-x(L) remains unknown. We show that although signaling through the TCR is sufficient to stimulate transcription of Bcl-x(L) mRNA, CD28, by activating PI3K and mammalian target of rapamycin, provides a critical signal that regulates the translation of Bcl-x(L) transcripts. We observe that CD28 induced 4E-binding protein-1 phosphorylation, an inhibitor of the translational machinery, and that CD28 costimulation directly augmented the translation of a Bcl-x(L) 5'-untranslated region reporter construct. Lastly, costimulation by CD28 shifted the distribution of Bcl-x(L) mRNA transcripts from the pretranslation complex to the translationally active polyribosomes. These results demonstrate that CD28 relieves the translational inhibition of Bcl-x(L) in a PI3K/mammalian target of rapamycin-dependent manner.


Assuntos
Antígenos CD28/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Biossíntese de Proteínas/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Linfócitos T/imunologia , Animais , Northern Blotting , Antígenos CD28/genética , Antígenos CD28/imunologia , Complexo CD3/imunologia , Complexo CD3/metabolismo , Morte Celular/fisiologia , Células Cultivadas , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Humanos , Imunossupressores/farmacologia , Interleucina-2/biossíntese , Interleucina-2/imunologia , Células Jurkat , Camundongos , Camundongos Transgênicos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/imunologia , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirolimo/farmacologia , Linfócitos T/metabolismo , Transfecção , Proteína bcl-X
17.
Blood ; 102(12): 4115-22, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12907450

RESUMO

SOCS1-/- mice die prematurely of increased interferon-gamma (IFNgamma) signaling with severe thymic atrophy and accelerated maturation of T cells. However, it was unclear whether the thymic defects were caused by SOCS1 deficiency or by increased IFNgamma signaling. Using SOCS1-/- IFNgamma-/- mice, we show in this study that SOCS1 deficiency skews thymocyte development toward CD8 lineage independently of IFNgamma. Fetal thymic organ cultures and intrathymic transfer of CD4-CD8- precursors into Rag1-/- mice show that the lineage skewing in SOCS1-/- mice is a T-cell autonomous defect. Interestingly, SOCS1 is not required for attenuating interleukin-7 (IL-7) signaling at the CD4-CD8- stage but is essential for regulating IL-15 and IL-2 signaling in CD8+ thymocytes. IL-15 selectively stimulates SOCS1-/- CD8+ thymocytes, inducing sustained signal transducer and activator of transcription 5 (STAT5) phosphorylation and massive proliferation. IL-15 also strongly up-regulates Bcl-xL and CD44 in CD8+ thymocytes lacking SOCS1. The SOCS1 gene is induced in CD4+ thymocytes by gammac cytokines, whereas CD8+ thymocytes constitutively express SOCS1 mRNA even in the absence of cytokine stimulation. Because many different cell types express IL-15, our results strongly suggest that SOCS1 functions as an indispensable attenuator of IL-15 receptor signaling in developing CD8+ thymocytes.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Proteínas de Transporte/fisiologia , Proteínas do Leite , Receptores de Interleucina-2/metabolismo , Proteínas Repressoras , Transdução de Sinais , Timo/citologia , Animais , Proteínas de Transporte/genética , Linhagem da Célula , Proteínas de Ligação a DNA/metabolismo , Receptores de Hialuronatos/biossíntese , Interferon gama/genética , Interferon gama/fisiologia , Interleucina-3/metabolismo , Interleucina-7/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Receptores de Interleucina-15 , Fator de Transcrição STAT5 , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina , Transativadores/metabolismo , Proteína bcl-X
18.
J Immunol ; 171(5): 2435-45, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12928391

RESUMO

T lymphocyte survival, proliferation, and death in the periphery are dependent on several cytokines. Many of these cytokines induce the expression of suppressor of cytokine signaling-1 (SOCS1), a feedback inhibitor of JAK kinases. However, it is unclear whether the cytokines that regulate T lymphocyte homeostasis are critically regulated by SOCS1 in vivo. Using SOCS1(-/-)IFN-gamma(-/-) mice we show that SOCS1 deficiency causes a lymphoproliferative disorder characterized by decreased CD4/CD8 ratio due to chronic accumulation of CD8+CD44(high) memory phenotype T cells. SOCS1-deficient CD8+ T cells express elevated levels of IL-2Rbeta, show increased proliferative response to IL-15 and IL-2 in vitro, and undergo increased bystander proliferation and vigorous homeostatic expansion in vivo. Sorted CD8+CD44(high) T cells from SOCS1(-/-)IFN-gamma(-/-) mice respond 5 times more strongly than control cells, indicating that SOCS1 is a critical regulator of IL-15R signaling. Consistent with this idea, IL-15 stimulates sustained STAT5 phosphorylation in SOCS1-deficient CD8+ T cells. IL-15 strongly induces TNF-alpha production in SOCS1-deficient CD8+ T cells, indicating that SOCS1 is also a critical regulator of CD8+ T cell activation by IL-15. However, IL-15 and IL-2 induce comparable levels of Bcl-2 and Bcl-x(L) in SOCS1-deficient and SOCS1-sufficient CD8+ T cells, suggesting that cytokine receptor signals required for inducing proliferation and cell survival signals are not identical. These results show that SOCS1 differentially regulates common gamma-chain cytokine signaling in CD8+ T cells and suggest that CD8+ T cell homeostasis is maintained by distinct mechanisms that control cytokine-mediated survival and proliferation signals.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Proteínas de Transporte/fisiologia , Receptores de Hialuronatos/biossíntese , Memória Imunológica , Receptores de Interleucina-2/antagonistas & inibidores , Receptores de Interleucina-2/fisiologia , Proteínas Repressoras , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Efeito Espectador/genética , Efeito Espectador/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Proteínas de Transporte/genética , Divisão Celular/genética , Divisão Celular/imunologia , Células Clonais , Relação Dose-Resposta Imunológica , Proteínas de Homeodomínio/genética , Homeostase/genética , Homeostase/imunologia , Memória Imunológica/genética , Imunofenotipagem , Interferon gama/deficiência , Interferon gama/genética , Interleucina-15/farmacologia , Transtornos Linfoproliferativos/genética , Transtornos Linfoproliferativos/imunologia , Transtornos Linfoproliferativos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interleucina-15 , Transdução de Sinais/genética , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia
19.
Mol Cell Biol ; 23(9): 3067-78, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12697809

RESUMO

Genetic studies have implicated the cytosolic juxtamembrane region of the Kit receptor tyrosine kinase as an autoinhibitory regulatory domain. Mutations in the juxtamembrane domain are associated with cancers, such as gastrointestinal stromal tumors and mastocytosis, and result in constitutive activation of Kit. Here we elucidate the biochemical mechanism of this regulation. A synthetic peptide encompassing the juxtamembrane region demonstrates cooperative thermal denaturation, suggesting that it folds as an autonomous domain. The juxtamembrane peptide directly interacted with the N-terminal ATP-binding lobe of the kinase domain. A mutation in the juxtamembrane region corresponding to an oncogenic form of Kit or a tyrosine-phosphorylated form of the juxtamembrane peptide disrupted the stability of this domain and its interaction with the N-terminal kinase lobe. Kinetic analysis of the Kit kinase harboring oncogenic mutations in the juxtamembrane region displayed faster activation times than the wild-type kinase. Addition of exogenous wild-type juxtamembrane peptide to active forms of Kit inhibited its kinase activity in trans, whereas the mutant peptide and a phosphorylated form of the wild-type peptide were less effective inhibitors. Lastly, expression of the Kit juxtamembrane peptide in cells which harbor an oncogenic form of Kit inhibited cell growth in a Kit-specific manner. Together, these results show the Kit kinase is autoinhibited through an intramolecular interaction with the juxtamembrane domain, and tyrosine phosphorylation and oncogenic mutations relieved the regulatory function of the juxtamembrane domain.


Assuntos
Citosol/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Trifosfato de Adenosina/metabolismo , Regulação Alostérica/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Divisão Celular/genética , Linhagem Celular Transformada , Membrana Celular/metabolismo , Ativação Enzimática , Dados de Sequência Molecular , Mutação , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Fosforilação , Dobramento de Proteína , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-kit/química , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Tirosina/metabolismo
20.
J Immunol ; 169(9): 5010-20, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12391216

RESUMO

Suppressor of cytokine signaling 1 (SOCS1) is rapidly induced following stimulation by several cytokines. SOCS1 negatively regulates cytokine receptor signal transduction by inhibiting Janus family tyrosine kinases. Lack of such feedback regulation underlies the premature death of SOCS1(-/-) mice due to unbridled IFN-gamma signaling. We used mouse embryo fibroblasts derived from SOCS1(-/-) mice to investigate the role of SOCS1 in IFN-gamma signaling pathways. SOCS1(-/-) fibroblasts were exquisitely sensitive to the IFN-gamma-mediated growth arrest and showed sustained STAT1 phosphorylation. However, SOCS1(-/-) fibroblasts were inefficient in MHC class II surface expression following IFN-gamma stimulation, despite a marked induction of the MHC class II transactivator and MHC class II gene expression. Retroviral transduction of wild-type SOCS1 relieved the growth-inhibitory effects of IFN-gamma in SOCS1(-/-) fibroblasts by inhibiting STAT1 activation. SOCS1R105K, carrying a mutation within the phosphotyrosine-binding pocket of the Src homology 2 domain, did not inhibit STAT1 phosphorylation, yet considerably inhibited IFN-gamma-mediated growth arrest. Strikingly, expression of SOCS1R105K restored the IFN-gamma-induced MHC class II expression in SOCS1(-/-) cells, indicating that expression of SOCS1 facilitates MHC class II expression in fibroblasts. Our results show that SOCS1, in addition to its negative regulatory role of inhibiting Janus kinases, has an unanticipated positive regulatory function in retarding the degradation of IFN-gamma-induced MHC class II proteins in fibroblasts.


Assuntos
Proteínas de Transporte/fisiologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Antígenos de Histocompatibilidade Classe II/biossíntese , Interferon gama/fisiologia , Proteínas Repressoras , Transdução de Sinais/imunologia , Células 3T3 , Animais , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Divisão Celular/genética , Divisão Celular/imunologia , Linhagem Celular Transformada , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Fibroblastos/citologia , Regulação da Expressão Gênica/imunologia , Genes MHC da Classe II , Inibidores do Crescimento/agonistas , Inibidores do Crescimento/antagonistas & inibidores , Inibidores do Crescimento/fisiologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Interferon gama/agonistas , Interferon gama/antagonistas & inibidores , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Transporte Proteico/imunologia , Fator de Transcrição STAT1 , Transdução de Sinais/genética , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina , Transativadores/metabolismo , Regulação para Cima/genética , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...