Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Shock ; 59(4): 646-656, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36719431

RESUMO

ABSTRACT: Background: Sepsis is a life-threatening medical emergency, frequently complicated with intensive care unit-acquired weakness syndrome (ICU-AW). ICU-AW patients display flaccid weakness of the limbs, especially in the proximal limb muscles. However, little is known regarding its pathogenesis. Here, we aimed to identify the potential signaling pathway involved in ICU-AW regulation and identify a potential therapeutic drug for intervention. Methods: Both in vivo and in vitro septic mice were used. For the in vivo septic mice, either cecum ligation and puncture or intraperitoneal injection of LPS was conducted in mice. The body weight and muscle mass were then measured and recorded. Muscle strength was evaluated by limb grip strength test. The expression of proteins extracted from cells and muscles was checked through Western blot analysis. Quantitative reverse transcription-polymerase chain reaction was carried out to test the transcriptional level of genes. Senescence-associated ß-galactosidase (SA-ß-gal) staining and Sirius red for collagen staining were conducted. Metformin, as an antiaging agent, was then tested for any attenuation of sepsis-related symptoms. For in vitro sepsis modeling, myoblasts were treated with LPS, analyzed for senescence-related protein expression, and subsequently retested upon metformin treatment. Results: We found that both the weight and strength of muscle were dramatically reduced in cecum ligation and puncture- or LPS-induced septic mice. RNA-seq analysis revealed that various cellular senescent genes were involved in sepsis. In line with this, expression of senescence-related genes, p53 and p21 were both upregulated. Both SA-ß-gal and Sirius red for collagen staining were enhanced in tibialis anterior muscles. Notably, inhibition of p53 expression by siRNA prominently reduced the number of SA-ß-gal-positive myoblasts upon LPS treatment. This indicated sepsis-induced cellular senescence to be dependent on p53. Consistent with the function of metformin in antiaging, metformin attenuated cellular senescence in both murine myoblasts and skeletal muscles during sepsis. Muscle strength of septic mice was improved upon metformin treatment. Metformin intervention is therefore proposed as a potential therapeutic strategy for ICU-AW. Conclusion: Taken together, we revealed a previously unappreciated linkage between cellular senescence and sepsis-induced muscle weakness and propose metformin as a potential therapeutic drug for the treatment of ICU-AW.


Assuntos
Metformina , Sepse , Camundongos , Animais , Metformina/farmacologia , Metformina/uso terapêutico , Proteína Supressora de Tumor p53/metabolismo , Lipopolissacarídeos/toxicidade , Senescência Celular , Debilidade Muscular/tratamento farmacológico , Debilidade Muscular/etiologia , Sepse/complicações , Sepse/tratamento farmacológico
2.
World J Clin Cases ; 10(9): 2916-2922, 2022 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-35434094

RESUMO

BACKGROUND: Undifferentiated pleomorphic sarcoma (UPS) is a type of soft tissue sarcoma, the histologic origin and differentiation direction of which are still unclear. There are few treatment options for UPS other than surgery. Herein we describe a patient who had multiple recurrences of UPS postoperatively, but R0 resection was achieved by local hyperthermia combined with chemotherapy, thus providing a new treatment approach for similar situations. CASE SUMMARY: A 65-year-old man sought evaluation from a physician for a mass on his right back. After surgery, the pathologic diagnosis was fibrosarcoma. During the follow-up evaluations until 2021, the patient had four relapses of varying degrees. Postoperative pathology confirmed the recurrence of UPS on the right back. In March 2021, he underwent local hyperthermia combined with two cycles of chemotherapy for recurring lesions. After magnetic resonance imaging re-examination and preoperative examination, the patient chose surgery again. During the operation, the tumors were easy to excise, the amount of bleeding decreased significantly, and the pathologic evaluation confirmed that one of the specimens was an R0 excision. CONCLUSION: Local hyperthermia combined with chemotherapy enables R0 resection to be achieved in patients with advanced UPS recurrence.

3.
Cell Death Differ ; 27(8): 2344-2362, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32051546

RESUMO

Rab5 is a master regulator for endosome biogenesis and transport while its in vivo physiological function remains elusive. Here, we find that Rab5a is upregulated in several in vivo and in vitro myogenesis models. By generating myogenic Rab5a-deficient mice, we uncover the essential roles of Rab5a in regulating skeletal muscle regeneration. We further reveal that Rab5a promotes myoblast differentiation and directly interacts with insulin receptor substrate 1 (IRS1), an essential scaffold protein for propagating IGF signaling. Rab5a interacts with IRS1 in a GTP-dependent manner and this interaction is enhanced upon IGF-1 activation and myogenic differentiation. We subsequently identify that the arginine 207 and 222 of IRS1 and tyrosine 82, 89, and 90 of Rab5a are the critical amino acid residues for mediating the association. Mechanistically, Rab5a modulates IRS1 activation by coordinating the association between IRS1 and the IGF receptor (IGFR) and regulating the intracellular membrane targeting of IRS1. Both myogenesis-induced and IGF-evoked AKT-mTOR signaling are dependent on Rab5a. Myogenic deletion of Rab5a also reduces the activation of AKT-mTOR signaling during skeletal muscle regeneration. Taken together, our study uncovers the physiological function of Rab5a in regulating muscle regeneration and delineates the novel role of Rab5a as a critical switch controlling AKT-mTOR signaling by activating IRS1.


Assuntos
Diferenciação Celular , Proteínas Substratos do Receptor de Insulina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Músculo Esquelético/fisiologia , Mioblastos/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regeneração/fisiologia , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Células HEK293 , Membro Posterior/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular/genética , Mioblastos/metabolismo , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima/genética , Proteínas rab5 de Ligação ao GTP/genética
4.
Aging Cell ; 18(5): e13003, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31313490

RESUMO

Cellular senescence plays both beneficial and detrimental roles in embryonic development and tissue regeneration, while the underlying mechanism remains elusive. Recent studies disclosed the emerging roles of heat-shock proteins in regulating muscle regeneration and homeostasis. Here, we found that Hsp90ß, but not Hsp90α isoform, was significantly upregulated during muscle regeneration. RNA-seq analysis disclosed a transcriptional elevation of p21 in Hsp90ß-depleted myoblasts, which is due to the upregulation of p53. Moreover, knockdown of Hsp90ß in myoblasts resulted in p53-dependent cellular senescence. In contrast to the notion that Hsp90 interacts with and protects mutant p53 in cancer, Hsp90ß preferentially bound to wild-type p53 and modulated its degradation via a proteasome-dependent manner. Moreover, Hsp90ß interacted with MDM2, the chief E3 ligase of p53, to regulate the stability of p53. In line with these in vitro studies, the expression level of p53-p21 axis was negatively correlated with Hsp90ß in aged mice muscle. Consistently, administration of 17-AAG, a Hsp90 inhibitor under clinical trial, impaired muscle regeneration by enhancing injury-induced senescence in vivo. Taken together, our finding revealed a previously unappreciated role of Hsp90ß in regulating p53 stability to suppress senescence both in vitro and in vivo.


Assuntos
Senescência Celular , Proteínas de Choque Térmico HSP90/metabolismo , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular , Proteínas de Choque Térmico HSP90/química , Camundongos , Proteínas Proto-Oncogênicas c-mdm2/química
5.
Stem Cells ; 36(4): 527-539, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29315990

RESUMO

Tendon repair is a clinical challenge because of the limited understanding on tenogenesis. The synthesis of type I collagen (Collagen I) and other extracellular matrix are essential for tendon differentiation and homeostasis. Current studies on tenogenesis focused mostly on the tenogenic transcriptional factors while the signaling controlling tenogenesis on translational level remains largely unknown. Here, we showed that mechanistic target of rapamycin (mTOR) signaling was activated by protenogenic growth factor, transforming growth factors beta1, and insulin-like growth factor-I. The expression of mTOR was upregulated during tenogenesis of mesenchymal stem cells (MSCs). Moreover, mTOR was downregulated in human tendinopathy tissues and was inactivated upon statin treatment. Both inhibition and depletion of AKT or mTOR significantly reduced type I collagen production and impaired tenogenesis of MSCs. Tendon specific-ablation of mTOR resulted in tendon defect and reduction of Collagen I. However, there is no evident downregulation of tendon associated collagens at the transcription level. Our study demonstrated that AKT-mTOR axis is a key mediator of tendon differentiation and provided a novel therapeutic target for tendinopathy and tendon injuries. Stem Cells 2018;36:527-539.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Tendões/metabolismo , Animais , Células-Tronco Mesenquimais/citologia , Camundongos , Tendões/citologia , Fator de Crescimento Transformador beta1/metabolismo
6.
J Invest Surg ; 29(5): 254-9, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27010682

RESUMO

BACKGROUND: Endovenous laser therapy (EVLT) is safe and effective for lower limb venous ulcers. However, severe necrosis and infection in the ulcer area are contraindications of puncture and EVLT. Local bath with ozone gas has been shown to improve the condition of ulcer areas. The aim of this study was to evaluate the clinical efficacy of ozone gas bath combined with EVLT in comparison with EVLT alone for the treatment for lower limb venous ulcers. PATIENTS AND METHODS: Ninety-two patients with venous ulcers were randomized to receive ozone gas bath combined with EVLT (OEVLT group) or EVLT alone (EVLT group). In the OEVLT group, the venous ulcers were preconditioned with ozone gas bath prior to EVLT. The minimum follow-up time was 12 months. The two groups were compared in terms of complete occlusion of the treated veins, ulcer healing ratio, ratio of ulcer recurrence, patient satisfaction, complications, and side effects. RESULTS: There was no significant difference in venous occlusion between the two groups. The ratio of ulcer healing in the OEVLT group was significantly higher than the EVLT group at 12 months follow-up. Patients in the OEVLT group showed better satisfaction and a lower recurrence ratio than the OEVLT group. No severe complications or side effects occurred in either groups. CONCLUSIONS: Ozone gas bath combined with EVLT showed improved efficacy for the treatment of lower limb venous ulcers and lower recurrence ratio comparison with EVLT alone. This procedure is a safe and technically feasible.


Assuntos
Banhos/métodos , Terapia a Laser/métodos , Ozônio/uso terapêutico , Úlcera Varicosa/cirurgia , Úlcera Varicosa/terapia , Idoso , Terapia Combinada , Feminino , Gases/uso terapêutico , Humanos , Perna (Membro) , Masculino , Pessoa de Meia-Idade , Recidiva , Resultado do Tratamento , Cicatrização
7.
Stem Cells ; 34(4): 1083-96, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26851078

RESUMO

Calcification of soft tissues, such as heart valves and tendons, is a common clinical problem with limited therapeutics. Tissue specific stem/progenitor cells proliferate to repopulate injured tissues. But some of them become divergent to the direction of ossification in the local pathological microenvironment, thereby representing a cellular target for pharmacological approach. We observed that HIF-2alpha (encoded by EPAS1 inclined form) signaling is markedly activated within stem/progenitor cells recruited at calcified sites of diseased human tendons and heart valves. Proinflammatory microenvironment, rather than hypoxia, is correlated with HIF-2alpha activation and promoted osteochondrogenic differentiation of tendon stem/progenitor cells (TSPCs). Abnormal upregulation of HIF-2alpha served as a key switch to direct TSPCs differentiation into osteochondral-lineage rather than teno-lineage. Notably, Scleraxis (Scx), an essential tendon specific transcription factor, was suppressed on constitutive activation of HIF-2alpha and mediated the effect of HIF-2alpha on TSPCs fate decision. Moreover, pharmacological inhibition of HIF-2alpha with digoxin, which is a widely utilized drug, can efficiently inhibit calcification and enhance tenogenesis in vitro and in the Achilles's tendinopathy model. Taken together, these findings reveal the significant role of the tissue stem/progenitor cells fate decision and suggest that pharmacological regulation of HIF-2alpha function is a promising approach for soft tissue calcification treatment.


Assuntos
Tendão do Calcâneo/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Calcinose/tratamento farmacológico , Terapia de Tecidos Moles , Tendão do Calcâneo/crescimento & desenvolvimento , Tendão do Calcâneo/patologia , Idoso , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Calcinose/genética , Calcinose/patologia , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Microambiente Celular/efeitos dos fármacos , Condrogênese/genética , Digoxina/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Ratos , Cardiopatia Reumática/genética , Cardiopatia Reumática/patologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia
8.
Ther Clin Risk Manag ; 10: 17-26, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24379677

RESUMO

BACKGROUND: Coadministration of 1,4-dihydropyridine calcium channel blockers (DHP-CCBs) with statins (or 3-hydroxy-3-methylglutaryl-coenzyme A [HMG-CoA] reductase inhibitors) is common for patients with hypercholesterolemia and hypertension. To reduce the risk of myopathy, in 2011, the US Food and Drug Administration (FDA) Drug Safety Communication set a new dose limitation for simvastatin, for patients taking simvastatin concomitantly with amlodipine. However, there is no such dose limitation for atorvastatin for patients receiving amlodipine. The combination pill formulation of amlodipine/atorvastatin is available on the market. There been no systematic review of the pharmacokinetic drug-drug interaction (DDI) profile of DHP-CCBs with statins, the underlying mechanisms for DDIs of different degree, or the corresponding management of clinical risk. METHODS: The relevant literature was identified by performing a PubMed search, covering the period from January 1987 to September 2013. Studies in the field of drug metabolism and pharmacokinetics that described DDIs between DHP-CCB and statin or that directly compared the degree of DDIs associated with cytochrome P450 (CYP)3A4-metabolized statins or DHP-CCBs were included. The full text of each article was critically reviewed, and data interpretation was performed. RESULTS: There were three circumstances related to pharmacokinetic DDIs in the combined use of DHP-CCB and statin: 1) statin is comedicated as the precipitant drug (pravastatin-nimodipine and lovastatin-nicardipine); 2) statin is comedicated as the object drug (isradipine-lovastatin, lacidipine-simvastatin, amlodipine-simvastatin, benidipine-simvastatin, azelnidipine- simvastatin, lercanidipine-simvastatin, and amlodipine-atorvastatin); and 3) mutual interactions (lercanidipine-fluvastatin). Simvastatin has an extensive first-pass effect in the intestinal wall, whereas atorvastatin has a smaller intestinal first-pass effect. The interaction with simvastatin seems mainly driven by CYP3A4 inhibition at the intestinal level, whereas the interaction with atorvastatin is more due to hepatic CYP3A4 inhibition. The interaction of CYP3A4 inhibitor with simvastatin has been more pronounced compared with atorvastatin. From the current data, atorvastatin seems to be a safer CYP3A4-statin for comedication with DHP-CCB. There is no convincing evidence that amlodipine is an unusual DHP-CCB, either as a precipitant drug or as an object drug, from the perspective of CYP3A4-mediated drug metabolism. Amlodipine may have interactions with CYP3A5 in addition to CYP3A4, which may explain its particular characteristics in comparison with other DHP-CCBs. The degree of DDIs between the DHP-CCB and statin and the clinical outcome depends on many factors, such as the kind of statin, physicochemical proprieties of the DHP-CCB, the dose of either the precipitant drug or the object drug, the sex of the patient (eg, isradipine-lovastatin), route of drug administration (eg, oral versus intravenous nicardipine-lovastatin), the administration schedule (eg, nonconcurrent dosing method versus concurrent dosing method), and the pharmacogenetic status (eg, CYP3A5-nonexpressers versus CYP3A5-expressers). CONCLUSION: Clinical professionals should enhance risk management regarding the combination use of two classes of drugs by increasing their awareness of the potential changes in therapeutic efficacy and adverse drug reactions, by rationally prescribing alternatives, by paying attention to dose adjustment and the administration schedule, and by review of the appropriateness of physician orders. Further study is needed - the DDIs between DHP-CCBs and statins have not all been studied in humans, from either a pharmacokinetic or a clinical perspective; also, the strength of the different pharmacokinetic interactions of DHP-CCBs with statins should be addressed by systematic investigations.

9.
PLoS One ; 7(3): e33863, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22479462

RESUMO

The CRAL_TRIO protein domain, which is unique to the Sec14 protein superfamily, binds to a diverse set of small lipophilic ligands. Similar domains are found in a range of different proteins including neurofibromatosis type-1, a Ras GTPase-activating Protein (RasGAP) and Rho guanine nucleotide exchange factors (RhoGEFs). Proteins containing this structural protein domain exhibit a low sequence similarity and ligand specificity while maintaining an overall characteristic three-dimensional structure. We have previously demonstrated that the BNIP-2 and Cdc42GAP Homology (BCH) protein domain, which shares a low sequence homology with the CRAL_TRIO domain, can serve as a regulatory scaffold that binds to Rho, RhoGEFs and RhoGAPs to control various cell signalling processes. In this work, we investigate 175 BCH domain-containing proteins from a wide range of different organisms. A phylogenetic analysis with ~100 CRAL_TRIO and similar domains from eight representative species indicates a clear distinction of BCH-containing proteins as a novel subclass within the CRAL_TRIO/Sec14 superfamily. BCH-containing proteins contain a hallmark sequence motif R(R/K)h(R/K)(R/K)NL(R/K)xhhhhHPs ('h' is large and hydrophobic residue and 's' is small and weekly polar residue) and can be further subdivided into three unique subtypes associated with BNIP-2-N, macro- and RhoGAP-type protein domains. A previously unknown group of genes encoding 'BCH-only' domains is also identified in plants and arthropod species. Based on an analysis of their gene-structure and their protein domain context we hypothesize that BCH domain-containing genes evolved through gene duplication, intron insertions and domain swapping events. Furthermore, we explore the point of divergence between BCH and CRAL-TRIO proteins in relation to their ability to bind small GTPases, GAPs and GEFs and lipid ligands. Our study suggests a need for a more extensive analysis of previously uncharacterized BCH, 'BCH-like' and CRAL_TRIO-containing proteins and their significance in regulating signaling events involving small GTPases.


Assuntos
Proteínas de Transporte/química , Proteínas Ativadoras de GTPase/química , Animais , Bases de Dados de Proteínas , Ordem dos Genes , Humanos , Modelos Moleculares , Filogenia , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
10.
J Surg Res ; 171(1): e47-53, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21924739

RESUMO

BACKGROUND: Subarachnoid hemorrhage (SAH) can induce a persistent inflammatory response, histopathologic changes in the gut. This study investigated whether progesterone administration modulates intestinal proinflammatory cytokine expression and structure alternations following SAH in male rats. MATERIALS AND METHODS: A total of 48 male rats were randomly divided into four groups: control group (n = 12), SAH group (n = 12), SAH+vehicle group (n = 12) and SAH+progesterone group (n = 12). We measured intestinal wet/dry weight ratio; the concentrations of IL-1ß, TNF-α, and IL-6 by enzyme-linked immunosorbent assay; intestinal mucosal morphologic changes by histopathologic study and electron microscopy. RESULTS: Administration of progesterone following SAH could increase the appetite scores of SAH rats and decrease concentrations of proinflammatory cytokines and wet/dry weight ratio in the gut. SAH-induced damage of gut structure was ameliorated after progesterone supplementation. CONCLUSIONS: The results of the present study suggest that the therapeutic benefit of post-SAH progesterone supplementation might be due to its inhibitory effects on intestinal proinflammatory cytokine expression and gut structure amelioration.


Assuntos
Enterite/tratamento farmacológico , Mucosa Intestinal , Progesterona/farmacologia , Hemorragia Subaracnóidea/complicações , Animais , Apetite/efeitos dos fármacos , Modelos Animais de Doenças , Enterite/etiologia , Enterite/patologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Masculino , Tamanho do Órgão/efeitos dos fármacos , Progestinas/farmacologia , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
11.
J Cell Sci ; 122(Pt 3): 414-24, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19158340

RESUMO

Deleted in liver cancer 1 (DLC1) is a multi-modular Rho-GTPase-activating protein (RhoGAP) and a tumor suppressor. Besides its RhoGAP domain, functions of other domains in DLC1 remain largely unknown. By protein precipitation and mass spectrometry, we identified eukaryotic elongation factor 1A1 (EF1A1) as a novel partner for the sterile alpha motif (SAM) domain of DLC1 but not the SAM domain of DLC2. The solution structure of DLC1 SAM revealed a new monomeric fold with four parallel helices, similar to that of DLC2 SAM but distinct from other SAM domains. Mutating F38, L39 and F40 within a hydrophobic patch retained its overall structure but abolished its interaction with EF1A1 with F38 and L39 forming an indispensable interacting motif. DLC1 SAM did not localize to and was not required for DLC1 to suppress the turnover of focal adhesions. Instead, DLC1 SAM facilitated EF1A1 distribution to the membrane periphery and ruffles upon growth factor stimulation. Compared with wild-type DLC1, the non-interactive DLC1 mutant is less potent in suppressing cell migration, whereas overexpression of the DLC1 SAM domain alone, but not the non-interactive mutant SAM or DLC2 SAM, greatly enhanced cell migration. This finding reveals a novel contribution of the SAM-EF1A1 interaction as a potentially important GAP-independent modulation of cell migration by DLC1.


Assuntos
Movimento Celular/fisiologia , Fator 1 de Elongação de Peptídeos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Proteínas Ativadoras de GTPase , Humanos , Camundongos , Dados de Sequência Molecular , Mutação/genética , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Ratos , Alinhamento de Sequência , Transfecção , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética
12.
J Cell Sci ; 119(Pt 16): 3337-50, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16899818

RESUMO

Human Cayman ataxia and mouse or rat dystonia are linked to mutations in the genes ATCAY (Atcay) that encode BNIP-H or Caytaxin, a brain-specific member of the BNIP-2 family. To explore its possible role(s) in neuronal function, we used protein precipitation and matrix-assisted laser desorption/ionisation mass spectrometry and identified kidney-type glutaminase (KGA) as a novel partner of BNIP-H. KGA converts glutamine to glutamate, which could serve as an important source of neurotransmitter. Co-immunoprecipitation with specific BNIP-H antibody confirmed that endogenous BNIP-H and KGA form a physiological complex in the brain, whereas binding studies showed that they interact with each other directly. Immunohistochemistry and in situ hybridisation revealed high BNIP-H expression in hippocampus and cerebellum, broadly overlapping with the expression pattern previously reported for KGA. Significantly, BNIP-H expression was activated in differentiating neurons of the embryonic carcinoma cell line P19 whereas its overexpression in rat pheochromocytoma PC12 cells relocalised KGA from the mitochondria to neurite terminals. It also reduced the steady-state levels of glutamate by inhibiting KGA enzyme activity. These results strongly suggest that through binding to KGA, BNIP-H could regulate glutamate synthesis at synapses during neurotransmission. Thus, loss of BNIP-H function could render glutamate excitotoxicity or/and deregulated glutamatergic activation, leading to ataxia, dystonia or other neurological disorders.


Assuntos
Glutamatos/metabolismo , Glutaminase/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/enzimologia , Sequência de Aminoácidos , Animais , Encéfalo/citologia , Carcinoma Embrionário/metabolismo , Carcinoma Embrionário/patologia , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Rim/metabolismo , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Coelhos , Ratos
13.
J Biol Chem ; 278(46): 45903-14, 2003 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-12944407

RESUMO

RhoA, Cdc42, and Rac1 are small GTPases that regulate cytoskeletal reorganization leading to changes in cell morphology and cell motility. Their signaling pathways are activated by guanine nucleotide exchange factors and inactivated by GTPase-activating proteins (GAPs). We have identified a novel RhoGAP, BPGAP1 (for BNIP-2 and Cdc42GAP Homology (BCH) domain-containing, Proline-rich and Cdc42GAP-like protein subtype-1), that is ubiquitously expressed and shares 54% sequence identity to Cdc42GAP/p50RhoGAP. BP-GAP1 selectively enhanced RhoA GTPase activity in vivo although it also interacted strongly with Cdc42 and Rac1. "Pull-down" and co-immunoprecipitation studies indicated that it formed homophilic or heterophilic complexes with other BCH domain-containing proteins. Fluorescence studies of epitope-tagged BPGAP1 revealed that it induced pseudopodia and increased migration of MCF7 cells. Formation of pseudopodia required its BCH and GAP domains but not the proline-rich region, and was differentially inhibited by coexpression of the constitutively active mutant of RhoA, or dominant negative mutants of Cdc42 and Rac1. However, the mutant without the proline-rich region failed to confer any increase in cell migration despite the induction of pseudopodia. Our findings provide evidence that cell morphology changes and migration are coordinated via multiple domains in BPGAP1 and present a novel mode of regulation for cell dynamics by a RhoGAP protein.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas Ativadoras de GTPase/química , Proteínas Ativadoras de GTPase/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/fisiologia , Proteínas de Saccharomyces cerevisiae , Proteína cdc42 de Ligação ao GTP/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Western Blotting , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , DNA Complementar/metabolismo , Eletroforese em Gel de Poliacrilamida , Epitopos/química , GTP Fosfo-Hidrolases/química , GTP Fosfo-Hidrolases/metabolismo , Glutationa Transferase/metabolismo , Humanos , Proteínas de Membrana/química , Microscopia de Fluorescência , Modelos Biológicos , Dados de Sequência Molecular , Proteínas de Transferência de Fosfolipídeos , Testes de Precipitina , Prolina/química , Isoformas de Proteínas , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Transfecção , Proteína cdc42 de Ligação ao GTP/metabolismo
14.
J Biol Chem ; 277(9): 7483-92, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11741952

RESUMO

We have cloned the cDNAs for two novel human proteins, designated BNIP-Salpha and beta (for BNIP-2 Similar) that are homologous to BNIP-2, a previously known Bcl-2 and E1B-associated protein. The BNIP-S gene encodes two protein isoforms; the longer protein (BNIP-Salpha) contains a complete BNIP-2 and Cdc42GAP Homology (BCH) domain, a novel protein domain that we recently identified, whereas its shorter variant (BNIP-Sbeta) lacks the full BCH domain as a result of an alternative RNA splicing that introduces a nonsense intron. Primer-specific reverse-transcription PCR revealed that both BNIP-Salpha and BNIP-Sbeta mRNA are differentially expressed in various cells and tissues. The expression of BNIP-Salpha or the complete BCH domain, but not BNIP-Sbeta, causes extensive apoptosis in cells. Furthermore, BNIP-Salpha can form a homophilic complex via a unique sequence motif within its BCH domain, and deletion of this interacting motif prevents its pro-apoptotic effect. These results indicate the presence of two BNIP-S splicing variants as cellular regulators and that the BCH domain of BNIP-Salpha confers a novel apoptotic function. The significance of this is discussed.


Assuntos
Apoptose , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Proteínas de Membrana , Proteína cdc42 de Ligação ao GTP/química , Processamento Alternativo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Anexina A5/farmacologia , Sequência de Bases , Western Blotting , Morte Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Clonagem Molecular , DNA Complementar/metabolismo , Glutationa Transferase/metabolismo , Humanos , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas de Transferência de Fosfolipídeos , Ligação Proteica , Isoformas de Proteínas , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Tempo , Distribuição Tecidual , Células Tumorais Cultivadas , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA