Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
EClinicalMedicine ; 68: 102433, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38318125

RESUMO

Background: RYR1-related myopathies (RYR1-RM) are caused by pathogenic variants in the RYR1 gene which encodes the type 1 ryanodine receptor (RyR1). RyR1 is the sarcoplasmic reticulum (SR) calcium release channel that mediates excitation-contraction coupling in skeletal muscle. RyR1 sub-conductance, SR calcium leak, reduced RyR1 expression, and oxidative stress often contribute to RYR1-RM pathogenesis. Loss of RyR1-calstabin1 association, SR calcium leak, and increased RyR1 open probability were observed in 17 RYR1-RM patient skeletal muscle biopsies and improved following ex vivo treatment with Rycal compounds. Thus, we initiated a first-in-patient trial of Rycal S48168 (ARM210) in ambulatory adults with genetically confirmed RYR1-RM. Methods: Participants received 120 mg (n = 3) or 200 mg (n = 4) S48168 (ARM210) daily for 29 days. The primary endpoint was safety and tolerability. Exploratory endpoints included S48168 (ARM210) pharmacokinetics (PK), target engagement, motor function measure (MFM)-32, hand grip and pinch strength, timed functional tests, PROMIS fatigue scale, semi-quantitative physical exam strength measurements, and oxidative stress biomarkers. The trial was registered with clinicaltrials.gov (NCT04141670) and was conducted at the National Institutes of Health Clinical Center between October 28, 2019 and December 12, 2021. Findings: S48168 (ARM210) was well-tolerated, did not cause any serious adverse events, and exhibited a dose-dependent PK profile. Three of four participants who received the 200 mg/day dose reported improvements in PROMIS-fatigue at 28 days post-dosing, and also demonstrated improved proximal muscle strength on physical examination. Interpretation: S48168 (ARM210) demonstrated favorable safety, tolerability, and PK, in RYR1-RM affected individuals. Most participants who received 200 mg/day S48168 (ARM210) reported decreased fatigue, a key symptom of RYR1-RM. These results set the foundation for a randomized, double-blind, placebo-controlled proof of concept trial to determine efficacy of S48168 (ARM210) in RYR1-RM. Funding: NINDS and NINR Intramural Research Programs, NIH Clinical Center Bench to Bedside Award (2017-551673), ARMGO Pharma Inc., and its development partner Les Laboratoires Servier.

2.
Clin Transl Sci ; 13(3): 462-472, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31899602

RESUMO

Ubrogepant is a novel, oral calcitonin gene-related peptide (CGRP) receptor antagonist intended for the acute treatment of migraine attacks. Ubrogepant has a chemical structure distinct from previous small-molecule CGRP receptor antagonists that were associated with elevated serum alanine aminotransferase (ALT) in clinical trials. Here, we report overall and hepatic safety data from two placebo-controlled phase I trials of ubrogepant, spray-dried oral compressed tablet (SD-OCT) in healthy male volunteers. Trial A was a pharmacokinetic (PK) trial of single (100-400 mg) and multiple (40-400 mg) ascending doses. Trial B was a dedicated hepatic safety trial assessing daily use of ubrogepant 150 mg for 28 days. Serum ALT (as hepatotoxicity biomarker) and PK data are reported. Ubrogepant was well-tolerated in both trials, with a low incidence of adverse events that did not differ greatly from placebo. Changes in mean ALT levels were minimal and similar to placebo. Over 28 days of treatment, the mean percentage change in ALT from baseline was < 5% at all time points. No participant in either trial demonstrated ALT ≥ 3× upper limit of normal at any time. Ubrogepant SD-OCT demonstrated linear PK appropriate for acute treatment of migraine, with rapid uptake (time of maximum plasma concentration (tmax ): 2-3 hours) and no accumulation with daily use. Overall, there was no evidence of ubrogepant-associated hepatotoxicity with daily doses up to 400 mg for 10 days or with daily ubrogepant 150 mg for 28 days. Supratherapeutic dosing is a useful strategy for characterizing hepatic safety in early drug development.


Assuntos
Alanina Transaminase/sangue , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Transtornos de Enxaqueca/tratamento farmacológico , Piridinas/efeitos adversos , Pirróis/efeitos adversos , Adolescente , Adulto , Doença Hepática Induzida por Substâncias e Drogas/sangue , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Voluntários Saudáveis , Humanos , Testes de Função Hepática , Masculino , Pessoa de Meia-Idade , Piridinas/administração & dosagem , Piridinas/farmacocinética , Pirróis/administração & dosagem , Pirróis/farmacocinética , Valores de Referência , Adulto Jovem
3.
Mol Ther Nucleic Acids ; 18: 580-589, 2019 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-31678734

RESUMO

Systemic delivery of antisense oligonucleotides (AO) for DMD exon skipping has proven effective for reframing DMD mRNA, rescuing dystrophin expression, and slowing disease progression in animal models. In humans with Duchenne muscular dystrophy treated with AOs, low levels of dystrophin have been induced, and modest slowing of disease progression has been observed, highlighting the need for improved efficiency of human skipping drugs. Here, we demonstrate that dantrolene and Rycals S107 and ARM210 potentiate AO-mediated exon skipping of exon 44 or exon 45 in patient-derived myotube cultures with appropriate mutations. Further, dantrolene is shown to boost AO-mediated exon skipping in patient-derived, induced cardiomyocyte cultures. Our findings further validate the ryanodine receptors (RyR) as the likely target responsible for exon skip boosting and demonstrate potential applicability beyond exon 51 skipping. These data provide preclinical support of dantrolene trial as an adjuvant to AO-mediated exon-skipping therapy in humans and identify a novel Rycal, ARM210, for development as a potential exon-skipping booster. Further, they highlight the value of mutation-specific DMD culture models for basic discovery, preclinical drug screening and translation of personalized genetic medicines.

4.
J Clin Pharmacol ; 58(3): 294-303, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29136283

RESUMO

Merck & Co., Inc. (Kenilworth, New Jersey) has recently published an integrated strategy for implementation of dried blood spots (DBS) in late-stage trials for population pharmacokinetic (PK) modeling. We applied this strategy for another late-stage clinical program: ubrogepant (MK-1602), a novel oral calcitonin gene-related peptide receptor antagonist for acute treatment of migraine. At the time of implementation, ubrogepant was entering phase 2 development. DBS was implemented to acquire PK information proximal to an acute migraine event to enable exposure-response modeling. The clinical endpoint was a spontaneous event, which generally occurs outside a clinic visit. Thus, an innovative feature of this trial was facilitating DBS in an outpatient setting. In vitro and bioanalytical tests established initial method feasibility and suitability for further evaluations in the clinic. A quantitative relationship was developed between blood and plasma concentrations from concurrently collected samples in a phase 1 (healthy subjects) and phase 2 (target patient population) study using graphical and population PK approaches. This integrated information was presented to the Food and Drug Administration for regulatory input. Following regulatory concurrence, DBS was poised for use in further clinical studies. Population PK modeling was used to dissect sources of variability contributing to DBS collection in the outpatient setting. What has been learned from this program has informed the broader integrated strategy of Merck & Co., Inc. (Kenilworth, NJ) for DBS implementation in clinical trials and research to improve the precision of PK data collected in an outpatient setting.


Assuntos
Antagonistas do Receptor do Peptídeo Relacionado ao Gene de Calcitonina/sangue , Antagonistas do Receptor do Peptídeo Relacionado ao Gene de Calcitonina/farmacocinética , Transtornos de Enxaqueca/sangue , Transtornos de Enxaqueca/tratamento farmacológico , Piridinas/sangue , Piridinas/farmacocinética , Pirróis/sangue , Pirróis/farmacocinética , Adulto , Antagonistas do Receptor do Peptídeo Relacionado ao Gene de Calcitonina/administração & dosagem , Relação Dose-Resposta a Droga , Teste em Amostras de Sangue Seco , Feminino , Humanos , Pessoa de Meia-Idade , Piridinas/administração & dosagem , Pirróis/administração & dosagem , Inquéritos e Questionários
5.
Drug Dev Ind Pharm ; 43(7): 1173-1177, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28277845

RESUMO

OBJECTIVES: A new improved mometasone furoate (Elocon™) cream with an emulsification system that produces a stable emulsion has been developed. In order to register the product in various markets, it was essential to ensure the cream was topically well tolerated and that it was bioequivalent to the reference product. METHODS: Phase I clinical studies were performed to assess the local safety and tolerability upon multiple dosing of this new cream as well as to assess the single-dose bioequivalence relative to the marketed product. Bioequivalence was assessed using a vasoconstrictive assay (VCA) after a dose-duration pilot study was completed with the marketed Elocon cream. KEY FINDINGS: The new mometasone cream and its vehicle were nonirritating in healthy subjects during 21-day patch application (MCII <0.025). The positive control was moderately irritating in the same study. The pivotal VCA study enrolled 162 subjects with 105 detectors included in the analysis of bioequivalence. In the 105 detectors, the ratio (×100%) of AUEC values at ED50 for test vs. standard (90% CI) was 112.91% (105.55, 120.87), within the bioequivalence criteria of (80, 125). CONCLUSIONS: These studies supported the registration of reformulated mometasone cream in various markets.


Assuntos
Furoato de Mometasona/administração & dosagem , Absorção Cutânea/efeitos dos fármacos , Vasoconstritores/química , Vasoconstritores/farmacologia , Administração Cutânea , Bioensaio , Humanos , Furoato de Mometasona/química , Furoato de Mometasona/farmacologia , Projetos Piloto , Equivalência Terapêutica
6.
Antimicrob Agents Chemother ; 59(6): 3385-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25824210

RESUMO

Posaconazole in oral suspension must be taken multiple times a day with food (preferably a high-fat meal) to ensure adequate exposure among patients. We evaluated the effect of food on the bioavailability of a new delayed-release tablet formulation of posaconazole at the proposed clinical dose of 300 mg once daily in a randomized, open-label, single-dose, two-period crossover study with 18 healthy volunteers. When a single 300-mg dose of posaconazole in tablet form (3 tablets × 100 mg) was administered with a high-fat meal, the posaconazole area under the concentration-time curve from 0 to 72 h (AUC0-72) and maximum concentration in plasma (Cmax) increased 51% and 16%, respectively, compared to those after administration in the fasted state. The median time to Cmax (Tmax) shifted from 5 h in the fasted state to 6 h under fed conditions. No serious adverse events were reported, and no subject discontinued the study due to an adverse event. Six of the 18 subjects reported at least one clinical adverse event; all of these events were mild and short lasting. The results of this study demonstrate that a high-fat meal only modestly increases the mean posaconazole exposure (AUC), ∼1.5-fold, after administration of posaconazole tablets, in contrast to the 4-fold increase in AUC observed previously for a posaconazole oral suspension given with a high-fat meal.


Assuntos
Antifúngicos/farmacocinética , Comprimidos/farmacocinética , Triazóis/farmacocinética , Administração Oral , Adolescente , Adulto , Idoso , Antifúngicos/administração & dosagem , Antifúngicos/sangue , Estudos Cross-Over , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Comprimidos/administração & dosagem , Triazóis/administração & dosagem , Triazóis/sangue , Adulto Jovem
7.
J Clin Pharmacol ; 54(11): 1280-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24895078

RESUMO

We evaluated the effect of prednisone on midazolam and odanacatib pharmacokinetics. In this open-label, 2-period crossover study in healthy male subjects, midazolam 2 mg was administered (Day -1) followed by odanacatib 50 mg (Day 1) during Part 1. In Period 2, prednisone 10 mg once daily (qd) was administered on Days 1-28; odanacatib was co-administered on Day 14 and midazolam 2 mg was co-administered on Days 1 and 28. Subjects were administered midazolam 2 mg on Days 42 and 56. Safety and tolerability were assessed throughout the study. A physiologically-based pharmacokinetic (PBPK) model was also built. There were 15 subjects enrolled; mean age was 31 years. The odanacatib AUC(0- ∞) GMR (90% CI) [odanacatib + prednisone (Day 14, Period 2)/odanacatib alone (Day 1, Period 1] was 1.06 (0.96, 1.17). AUC(0-∞) GMR (90%CI) [midazolam + prednisone (Day 28, Period 2)/midazolam alone (Day -1, Period 1] was 1.08 (0.93,1.26). There were no serious AEs or AEs leading to discontinuation. PBPK modeling showed that prednisone does not cause significant effects on the exposure of sensitive CYP3A4 substrates in vivo at therapeutic doses. Co-administration of prednisone 10 mg qd had no effect on pharmacokinetics of either odanacatib 10 mg or midazolam 2 mg.


Assuntos
Adjuvantes Anestésicos/farmacocinética , Anti-Inflamatórios/farmacocinética , Compostos de Bifenilo/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Midazolam/farmacocinética , Prednisona/farmacocinética , Adjuvantes Anestésicos/administração & dosagem , Adulto , Anti-Inflamatórios/administração & dosagem , Área Sob a Curva , Compostos de Bifenilo/administração & dosagem , Interações Medicamentosas , Meia-Vida , Humanos , Masculino , Midazolam/administração & dosagem , Pessoa de Meia-Idade , Prednisona/administração & dosagem , Adulto Jovem
8.
J Pharmacol Exp Ther ; 347(2): 478-86, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23975906

RESUMO

Calcitonin gene-related peptide (CGRP) is a potent neuropeptide whose agonist interaction with the CGRP receptor (CGRP-R) in the periphery promotes vasodilation, neurogenic inflammation and trigeminovascular sensory activation. This process is implicated in the cause of migraine headaches, and CGRP-R antagonists in clinical development have proven effective in treating migraine-related pain in humans. CGRP-R is expressed on blood vessel smooth muscle and sensory trigeminal neurons and fibers in the periphery as well as in the central nervous system. However, it is not clear what role the inhibition of central CGRP-R plays in migraine pain relief. To this end, the CGRP-R positron emission tomography (PET) tracer [(11)C]MK-4232 (2-[(8R)-8-(3,5-difluorophenyl)-6,8-[6-(11)C]dimethyl-10-oxo-6,9-diazaspiro[4.5]decan-9-yl]-N-[(2R)-2'-oxospiro[1,3-dihydroindene-2,3'-1H-pyrrolo[2,3-b]pyridine]-5-yl]acetamide) was discovered and developed for use in clinical PET studies. In rhesus monkeys and humans, [(11)C]MK-4232 displayed rapid brain uptake and a regional brain distribution consistent with the known distribution of CGRP-R. Monkey PET studies with [(11)C]MK-4232 after intravenous dosing with CGRP-R antagonists validated the ability of [(11)C]MK-4232 to detect changes in CGRP-R occupancy in proportion to drug plasma concentration. Application of [(11)C]MK-4232 in human PET studies revealed that telcagepant achieved only low receptor occupancy at an efficacious dose (140 mg PO). Therefore, it is unlikely that antagonism of central CGRP-R is required for migraine efficacy. However, it is not known whether high central CGRP-R antagonism may provide additional therapeutic benefit.


Assuntos
Acetanilidas/farmacocinética , Analgésicos/farmacocinética , Azepinas/farmacocinética , Encéfalo/metabolismo , Antagonistas do Receptor do Peptídeo Relacionado ao Gene de Calcitonina , Imidazóis/farmacocinética , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Compostos de Espiro/farmacocinética , Acetanilidas/química , Adulto , Analgésicos/uso terapêutico , Animais , Azepinas/uso terapêutico , Encéfalo/diagnóstico por imagem , Radioisótopos de Carbono , Feminino , Humanos , Imidazóis/uso terapêutico , Macaca mulatta , Masculino , Pessoa de Meia-Idade , Transtornos de Enxaqueca/tratamento farmacológico , Transtornos de Enxaqueca/metabolismo , Estrutura Molecular , Ligação Proteica , Compostos Radiofarmacêuticos/química , Especificidade da Espécie , Compostos de Espiro/química , Distribuição Tecidual , Adulto Jovem
9.
J Cell Physiol ; 226(1): 236-47, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20658524

RESUMO

The tyrosine kinase Fyn is a member of the Src family kinases which are important in many integrin-mediated cellular processes including cell adhesion and migration. Fyn has multiple phosphorylation sites which can affect its kinase activity. Among these phosphorylation sites, the serine 21 (S21) residue of Fyn is a protein kinase A (PKA) recognition site within an RxxS motif of the amino terminal SH4 domain of Fyn. In addition, S21 is critical for Fyn kinase-linked cellular signaling. Mutation of S21A blocks PKA phosphorylation of Fyn and alters its tyrosine kinase activity. Expression of Fyn S21A in cells lacking Src family kinases (SYF cell) led to decreased tyrosine phosphorylation of focal adhesion kinase resulting in reduced focal adhesion targeting, which slowed lamellipodia dynamics and thus cell migration. These changes in cell motility were reflected by the fact that cells expressing Fyn S21A were severely deficient in their ability to assemble and disassemble focal adhesions. Taken together, our findings indicate that phosphorylation of S21 within the pPKA recognition site (RxxS motif) of Fyn regulates its tyrosine kinase activity and controls focal adhesion targeting, and that this residue of Fyn is critical for transduction of signals arising from cell-extracellular matrix interactions.


Assuntos
Adesões Focais/fisiologia , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Serina/metabolismo , Sequência de Aminoácidos , Animais , Adesão Celular/fisiologia , Linhagem Celular , Movimento Celular , Regulação Enzimológica da Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Mutação , Fosforilação , Proteínas Proto-Oncogênicas c-fyn/genética , Transdução de Sinais/fisiologia
10.
J Cell Biol ; 187(5): 733-47, 2009 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-19951918

RESUMO

Focal adhesion disassembly is regulated by microtubules (MTs) through an unknown mechanism that involves dynamin. To test whether endocytosis may be involved, we interfered with the function of clathrin or its adaptors autosomal recessive hypercholesteremia (ARH) and Dab2 (Disabled-2) and found that both treatments prevented MT-induced focal adhesion disassembly. Surface labeling experiments showed that integrin was endocytosed in an extracellular matrix-, clathrin-, and ARH- and Dab2-dependent manner before entering Rab5 endosomes. Clathrin colocalized with a subset of focal adhesions in an ARH- and Dab2-dependent fashion. Direct imaging showed that clathrin rapidly accumulated on focal adhesions during MT-stimulated disassembly and departed from focal adhesions with integrin upon their disassembly. In migrating cells, depletion of clathrin or Dab2 and ARH inhibited focal adhesion disassembly and decreased the rate of migration. These results show that focal adhesion disassembly occurs through a targeted mechanism involving MTs, clathrin, and specific clathrin adaptors and that direct endocytosis of integrins from focal adhesions mediates their disassembly in migrating cells.


Assuntos
Clatrina/fisiologia , Endocitose , Adesões Focais/metabolismo , Integrinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transporte Vesicular/antagonistas & inibidores , Animais , Proteínas Reguladoras de Apoptose , Movimento Celular , Polaridade Celular , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Microtúbulos/fisiologia , Células NIH 3T3 , Proteínas do Tecido Nervoso/antagonistas & inibidores , Interferência de RNA
11.
J Cell Physiol ; 214(3): 604-13, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17786957

RESUMO

We examined the consequences of v-Crk expression in mouse embryo fibroblasts deficient Src family kinases or p130CAS. We found that Src kinases are essential for p130CAS/v-Crk signaling leading to FAK phosphorylation and cell migration in which Src is likely to mediate the focal adhesion targeting of v-Crk. SYF cells showed only low levels of FAK phosphorylation and cell migration, even in the presence of v-Crk. Expression of v-Crk restored migration of p130CAS-deficient cells to the level of wild-type cells, most likely through the targeting of v-Crk to focal adhesions by cSrc. In addition, we identified a new v-Crk-interacting protein that mediates v-Crk signaling in p130CAS-deficient cells. Using RT-PCR and caspase cleavage assays, we confirmed that this protein is not p130CAS and is responsible for maintaining v-Crk/Src signaling and migration in these. These findings suggest that focal adhesion targeting of v-Crk is essential in v-Crk-mediated cellular signaling and that v-Crk must form a complex with p130CAS or a p130CAS substitute to transduce signaling from the extracellular matrix.


Assuntos
Movimento Celular , Proteína Substrato Associada a Crk/metabolismo , Embrião de Mamíferos/citologia , Fibroblastos/enzimologia , Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/enzimologia , Proteína Oncogênica v-crk/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/farmacologia , Movimento Celular/efeitos dos fármacos , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/enzimologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Adesões Focais/efeitos dos fármacos , Camundongos , Microinjeções , Dados de Sequência Molecular , Miosinas/química , Miosinas/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Quinases da Família src/metabolismo
12.
Cell Commun Adhes ; 14(5): 169-80, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18163228

RESUMO

The alpha1beta1 and alpha2beta1 integrins belong to a family of cell-surface molecules involved in structural contacts and signal-transduction events across the cell membrane. Employing two-dimensional substrates coated with fluorescently labeled type I collagen, we have discovered a novel subcellular matrix remodeling event that is particular to cells that express the fibrillar collagen receptor alpha2beta1. Cells expressing alpha1beta1 also perform this collagen organization process, but less proficiently. This work will provide a basis for subsequent studies of cell-mediated collagen fibril assembly.


Assuntos
Colágeno Tipo I/metabolismo , Colágenos Fibrilares/metabolismo , Integrina alfa1beta1/metabolismo , Integrina alfa2beta1/metabolismo , Microscopia de Fluorescência/métodos , Animais , Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Colágeno Tipo I/ultraestrutura , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Colágenos Fibrilares/ultraestrutura , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Adesões Focais/metabolismo , Adesões Focais/ultraestrutura , Humanos , Camundongos , Células NIH 3T3 , Ligação Proteica/fisiologia
13.
Mol Biol Cell ; 17(10): 4237-48, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16855018

RESUMO

Integrin receptors, and associated cytoplasmic proteins mediate adhesion, cell signaling and connections to the cytoskeleton. Using fluorescent protein chimeras, we analyzed initial integrin adhesion in spreading fibroblasts with Total Internal Reflection Fluorescence (TIRF) microscopy. Surprisingly, sequential radial projection of integrin and actin containing filopodia formed the initial cell-matrix contacts. These Cdc42-dependent, integrin-containing projections recruited cytoplasmic focal adhesion (FA) proteins in a hierarchical manner; initially talin with integrin and subsequently FAK and paxillin. Radial FA structures then anchored cortical actin bridges between them and subsequently cells reorganized their actin, a process promoted by Src, and characterized by lateral FA reorientation to provide anchor points for actin stress fibers. Finally, the nascent adhesions coalesced until they formed mature FAs.


Assuntos
Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/fisiologia , Integrinas/metabolismo , Pseudópodes/metabolismo , Actinas/metabolismo , Animais , Adesão Celular , Matriz Extracelular/metabolismo , Camundongos , Microscopia de Fluorescência/métodos , Células NIH 3T3 , Paxilina/metabolismo , Talina/metabolismo , Transfecção , Proteína cdc42 de Ligação ao GTP/fisiologia , Quinases da Família src/fisiologia
14.
Mol Cell Biol ; 26(12): 4399-409, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16738308

RESUMO

Src kinase is a crucial mediator of adhesion-related signaling and motility. Src binds to focal adhesion kinase (FAK) through its SH2 domain and subsequently activates it for phosphorylation of downstream substrates. In addition to this binding function, data suggested that the SH2 domain might also perform an important role in targeting Src to focal adhesions (FAs) to enable further substrate phosphorylations. To examine this, we engineered an R175L mutation in cSrc to prevent the interaction with FAK pY397. This constitutively open Src kinase mediated up-regulated substrate phosphorylation in SYF cells but was unable to promote malignant transformation. Significantly, SrcR175L cells also had a profound motility defect and an impaired FA generation capacity. Importantly, we were able to recapitulate wild-type motile behavior and FA formation by directing the kinase to FAs, clearly implicating the SH2 domain in recruitment to FAK and indicating that this targeting capacity, and not simply Src-FAK scaffolding, was critical for normal Src function.


Assuntos
Movimento Celular/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Adesões Focais/fisiologia , Quinases da Família src/química , Quinases da Família src/fisiologia , Substituição de Aminoácidos , Animais , Arginina/química , Linhagem Celular , Galinhas , Proteína-Tirosina Quinases de Adesão Focal/química , Proteína-Tirosina Quinases de Adesão Focal/genética , Genes src , Humanos , Técnicas In Vitro , Mutagênese Sítio-Dirigida , Fosforilação , Domínios de Homologia de src , Quinases da Família src/genética
15.
J Cell Sci ; 119(Pt 6): 1175-83, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16507589

RESUMO

Integrin-mediated ;outside-in' signaling requires the transmission of a conformational change from the extracellular domains to the cytoplasmic domains. Although one component of this conformational change is the separation of the alpha and beta cytoplasmic domains, it is not clear how this separation could result in the initiation of downstream signals necessary for focal adhesion (FA) formation. To address this question, we used a swapped integrin heterodimer, in which the extracellular domains of the alpha and beta chains were attached to their opposing transmembrane and cytoplasmic domains. This receptor was able to bind ligand normally, but could not promote FA formation. We then displaced the beta cytoplasmic domain with either a duplication of its membrane-proximal region or an unrelated alpha-helical spacer. This displacement partially restored FA formation in these swapped receptors and rescued other aspects of integrin-mediated signaling, including cytoskeletal organization, motility and several tyrosine-phosphorylation-dependent signals. We suggest that separation of the cytoplasmic domains leads to alteration of the secondary structure of the distal beta tail, which initiates downstream signals leading to cytoskeletal reorganization.


Assuntos
Adesões Focais , Integrinas/metabolismo , Animais , Adesão Celular , Movimento Celular , Cricetinae , Integrinas/química , Integrinas/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade
16.
Science ; 303(5659): 836-9, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14764879

RESUMO

Microtubule (MT) stabilization is regulated by the small guanosine triphosphate (GTP)-binding protein Rho and its effector, mammalian homolog of Diaphanous (mDia), in migrating cells, but factors responsible for localized stabilization at the leading edge are unknown. We report that integrin-mediated activation of focal adhesion kinase (FAK) at the leading edge is required for MT stabilization by the Rho-mDia signaling pathway in mouse fibroblasts. MT stabilization also involved FAK-regulated localization of a lipid raft marker, ganglioside GM1, to the leading edge. The integrin-FAK signaling pathway may facilitate Rho-mDia signaling through GM1, or through a specialized membrane domain containing GM1, to stabilize MTs in the leading edge of migrating cells.


Assuntos
Membrana Celular/metabolismo , Integrinas/metabolismo , Microdomínios da Membrana/metabolismo , Microtúbulos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Acetilação , Animais , Proteínas de Transporte/metabolismo , Adesão Celular , Linhagem Celular , Colesterol/metabolismo , Fibronectinas/metabolismo , Fibronectinas/farmacologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Forminas , Gangliosídeo G(M1)/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Camundongos , Camundongos Knockout , Microtúbulos/ultraestrutura , Células NIH 3T3 , Fosforilação , Proteínas Tirosina Quinases/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Tubulina (Proteína)/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
17.
Mol Biol Cell ; 14(2): 349-60, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12589038

RESUMO

Integrin receptor signals are costimulatory for mitogenesis with the T-cell receptor during T-cell activation. A subset of integrin receptors can link to the adapter protein Shc and provide a mitogenic stimulus. Using a combination of genetic and pharmacological approaches, we show herein that integrin signaling to Shc in T cells requires the receptor tyrosine phosphatase CD45, the Src family kinase member Lck, and protein kinase C. Our results suggest a model in which integrin-dependent serine phosphorylation of Lck is the critical step that determines the efficiency of Shc tyrosine phosphorylation in T cells. Serine phosphorylation of Lck is dependent on PKC and is also linked to CD45 dephosphorylation. Mutants of Lck that cannot be phosphorylated on the critical serine residues do not signal efficiently to Shc and have greatly reduced kinase activity. This signaling from integrins to Lck may be an important step in the costimulation with the T-cell receptor during lymphocyte activation.


Assuntos
Integrinas/metabolismo , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Proteína Quinase C/metabolismo , Linfócitos T/metabolismo , Tirosina/metabolismo , Anticorpos Monoclonais/metabolismo , Western Blotting , Linhagem Celular , Reagentes de Ligações Cruzadas/farmacologia , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica , Humanos , Células Jurkat , Antígenos Comuns de Leucócito/fisiologia , Modelos Biológicos , Mutação , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-fyn , Serina/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA