Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Nature ; 612(7939): 301-309, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36450978

RESUMO

Clonal haematopoiesis involves the expansion of certain blood cell lineages and has been associated with ageing and adverse health outcomes1-5. Here we use exome sequence data on 628,388 individuals to identify 40,208 carriers of clonal haematopoiesis of indeterminate potential (CHIP). Using genome-wide and exome-wide association analyses, we identify 24 loci (21 of which are novel) where germline genetic variation influences predisposition to CHIP, including missense variants in the lymphocytic antigen coding gene LY75, which are associated with reduced incidence of CHIP. We also identify novel rare variant associations with clonal haematopoiesis and telomere length. Analysis of 5,041 health traits from the UK Biobank (UKB) found relationships between CHIP and severe COVID-19 outcomes, cardiovascular disease, haematologic traits, malignancy, smoking, obesity, infection and all-cause mortality. Longitudinal and Mendelian randomization analyses revealed that CHIP is associated with solid cancers, including non-melanoma skin cancer and lung cancer, and that CHIP linked to DNMT3A is associated with the subsequent development of myeloid but not lymphoid leukaemias. Additionally, contrary to previous findings from the initial 50,000 UKB exomes6, our results in the full sample do not support a role for IL-6 inhibition in reducing the risk of cardiovascular disease among CHIP carriers. Our findings demonstrate that CHIP represents a complex set of heterogeneous phenotypes with shared and unique germline genetic causes and varied clinical implications.


Assuntos
COVID-19 , Doenças Cardiovasculares , Humanos , Hematopoiese Clonal/genética , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/genética
2.
Mol Cell ; 83(13): 2149-2151, 2023 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-37419085
3.
Nature ; 613(7942): 30-31, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36544002
5.
Cell ; 134(3): 378-81, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18692458

RESUMO

Two terms used as frameworks for scientific experimentation--the "hypothesis" and the "model"--carry distinct philosophical assumptions, with important consequences for the practicing scientist.


Assuntos
Ciência/história , Ciência/métodos , História do Século XVI , História do Século XVII , História do Século XVIII , História do Século XX , Projetos de Pesquisa , Ciência/tendências
6.
Nucleic Acids Res ; 48(1): 212-230, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31665437

RESUMO

When replication forks encounter template DNA lesions, the lesion is simply skipped in some cases. The resulting lesion-containing gap must be converted to duplex DNA to permit repair. Some gap filling occurs via template switching, a process that generates recombination-like branched DNA intermediates. The Escherichia coli Uup and RadD proteins function in different pathways to process the branched intermediates. Uup is a UvrA-like ABC family ATPase. RadD is a RecQ-like SF2 family ATPase. Loss of both functions uncovers frequent and RecA-independent deletion events in a plasmid-based assay. Elevated levels of crossing over and repeat expansions accompany these deletion events, indicating that many, if not most, of these events are associated with template switching in postreplication gaps as opposed to simple replication slippage. The deletion data underpin simulations indicating that multiple postreplication gaps may be generated per replication cycle. Both Uup and RadD bind to branched DNAs in vitro. RadD protein suppresses crossovers and Uup prevents nucleoid mis-segregation. Loss of Uup and RadD function increases sensitivity to ciprofloxacin. We present Uup and RadD as genomic guardians. These proteins govern two pathways for resolution of branched DNA intermediates such that potentially deleterious genome rearrangements arising from frequent template switching are averted.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Adenosina Trifosfatases/genética , Proteínas de Bactérias/química , Replicação do DNA , DNA Bacteriano/genética , Proteínas de Ligação a DNA/química , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Transportadores de Cassetes de Ligação de ATP/deficiência , Adenosina Trifosfatases/deficiência , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Ciprofloxacina/farmacologia , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Farmacorresistência Bacteriana/genética , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Genoma Bacteriano , Plasmídeos/química , Plasmídeos/metabolismo , Origem de Replicação , Deleção de Sequência
7.
Crit Rev Biochem Mol Biol ; 54(2): 174-183, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31144559

RESUMO

GDF11 is a secreted factor in the TGFß family of cytokines. Its nearest neighbor evolutionarily is myostatin, a factor discovered as being a negative regulator of skeletal muscle growth. High profile studies several years ago suggested that GDF11 declines with age, and that restoration of systemic GDF11 to 'youthful' levels is beneficial for several age-related conditions. Particularly surprising was a report that supplementation of GDF11 aided skeletal muscle regeneration, as its homolog, myostatin, has the opposite role. Given this apparent contradiction in functionality, multiple independent labs sought to discern differences between the two factors and better elucidate age-related changes in circulating GDF11, with most failing to reproduce the initial finding of declining GDF11 levels, and, importantly, all subsequent studies examining the effects of GDF11 on skeletal muscle described an inhibitory effect on regeneration - and that higher doses induce skeletal muscle atrophy and cachexia. There have also been several studies examining the effect of GDF11 and/or the downstream ActRII pathway on cardiac function, along with several interesting reports on bone. A review of the GDF11 literature, as it relates in particular to aging and skeletal muscle, cardiac and bone biology, is presented.


Assuntos
Envelhecimento , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/fisiologia , Fatores de Diferenciação de Crescimento/metabolismo , Coração/fisiologia , Músculo Esquelético/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/sangue , Fatores de Diferenciação de Crescimento/sangue , Homeostase , Humanos , Miostatina/sangue , Miostatina/metabolismo
8.
J Bacteriol ; 203(18): e0022821, 2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34181484

RESUMO

The XP-D/DinG family of DNA helicases contributes to genomic stability in all three domains of life. Here, we investigate the role of one of these proteins, YoaA, of Escherichia coli. In E. coli, YoaA aids in tolerance to the nucleoside azidothymidine (AZT), a DNA replication inhibitor, and physically interacts with a subunit of the DNA polymerase III holoenzyme, HolC. We map the residues of YoaA required for HolC interaction to its C terminus by yeast two-hybrid analysis. We propose that this interaction competes with HolC's interaction with HolD and the rest of the replisome; YoaA indeed inhibits growth when overexpressed, dependent on this interaction region. By gene fusions, we show that YoaA is repressed by LexA and induced in response to DNA damage as part of the SOS response. Induction of YoaA by AZT is biphasic, with an immediate response after treatment and a slower response that peaks in the late log phase of growth. This growth-phase-dependent induction by AZT is not blocked by lexA3 (Ind-), which normally negates its self-cleavage, implying another means to induce the DNA damage response that responds to the nutritional state of the cell. We propose that YoaA helicase activity increases access to the 3' nascent strand during replication; consistent with this, YoaA appears to aid in the removal of potential A-to-T transversion mutations in ndk mutants, which are prone to nucleotide misincorporation. We provide evidence that YoaA and its paralog DinG may also initiate template switching that leads to deletions between tandem repeats in DNA. IMPORTANCE Maintaining genomic stability is crucial for all living organisms. Replication of DNA frequently encounters barriers that must be removed to complete genome duplication. Balancing DNA synthesis with its repair is critical and not entirely understood at a mechanistic level. The YoaA protein, studied here, is required for certain types of DNA repair and interacts in an alternative manner with proteins that catalyze DNA replication. YoaA is part of the well-studied LexA-regulated response to DNA damage, the SOS response. We describe an unusual feature of its regulation that promotes induction after DNA damage as the culture begins to experience starvation. Replication fork repair integrates both DNA damage and nutritional signals. We also show that YoaA affects genomic stability.


Assuntos
DNA Helicases/genética , DNA Polimerase III/metabolismo , Replicação do DNA , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Dano ao DNA/genética , DNA Helicases/metabolismo , DNA Polimerase III/genética , Reparo do DNA , Escherichia coli/fisiologia , Proteínas de Escherichia coli/metabolismo , Instabilidade Genômica/genética
9.
Proc Natl Acad Sci U S A ; 114(47): 12448-12453, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29109273

RESUMO

The TGF-ß family ligands myostatin, GDF11, and activins are negative regulators of skeletal muscle mass, which have been reported to primarily signal via the ActRIIB receptor on skeletal muscle and thereby induce muscle wasting described as cachexia. Use of a soluble ActRIIB-Fc "trap," to block myostatin pathway signaling in normal or cachectic mice leads to hypertrophy or prevention of muscle loss, perhaps suggesting that the ActRIIB receptor is primarily responsible for muscle growth regulation. Genetic evidence demonstrates however that both ActRIIB- and ActRIIA-deficient mice display a hypertrophic phenotype. Here, we describe the mode of action of bimagrumab (BYM338), as a human dual-specific anti-ActRIIA/ActRIIB antibody, at the molecular and cellular levels. As shown by X-ray analysis, bimagrumab binds to both ActRIIA and ActRIIB ligand binding domains in a competitive manner at the critical myostatin/activin binding site, hence preventing signal transduction through either ActRII. Myostatin and the activins are capable of binding to both ActRIIA and ActRIIB, with different affinities. However, blockade of either single receptor through the use of specific anti-ActRIIA or anti-ActRIIB antibodies achieves only a partial signaling blockade upon myostatin or activin A stimulation, and this leads to only a small increase in muscle mass. Complete neutralization and maximal anabolic response are achieved only by simultaneous blockade of both receptors. These findings demonstrate the importance of ActRIIA in addition to ActRIIB in mediating myostatin and activin signaling and highlight the need for blocking both receptors to achieve a strong functional benefit.


Assuntos
Receptores de Activinas Tipo II/antagonistas & inibidores , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Hipertrofia/induzido quimicamente , Músculo Esquelético/efeitos dos fármacos , Receptores de Activinas Tipo II/metabolismo , Ativinas/metabolismo , Animais , Anticorpos Bloqueadores/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Proteínas Morfogenéticas Ósseas/metabolismo , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Fatores de Diferenciação de Crescimento/metabolismo , Células HEK293 , Humanos , Hipertrofia/patologia , Masculino , Camundongos , Camundongos SCID , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miostatina/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Síndrome de Emaciação/tratamento farmacológico , Síndrome de Emaciação/patologia
10.
J Biol Chem ; 293(30): 11837-11849, 2018 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-29899111

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is among the most prevalent of the adult-onset muscular dystrophies. FSHD causes a loss of muscle mass and function, resulting in severe debilitation and reduction in quality of life. Currently, only the symptoms of FSHD can be treated, and such treatments have minimal benefit. The available options are not curative, and none of the treatments address the underlying cause of FSHD. The genetic, epigenetic, and molecular mechanisms triggering FSHD are now quite well-understood, and it has been shown that expression of the transcriptional regulator double homeobox 4 (DUX4) is necessary for disease onset and is largely thought to be the causative factor in FSHD. Therefore, we sought to identify compounds suppressing DUX4 expression in a phenotypic screen using FSHD patient-derived muscle cells, a zinc finger and SCAN domain-containing 4 (ZSCAN4)-based reporter gene assay for measuring DUX4 activity, and ∼3,000 small molecules. This effort identified molecules that reduce DUX4 gene expression and hence DUX4 activity. Among those, ß2-adrenergic receptor agonists and phosphodiesterase inhibitors, both leading to increased cellular cAMP, effectively decreased DUX4 expression by >75% in cells from individuals with FSHD. Of note, we found that cAMP production reduces DUX4 expression through a protein kinase A-dependent mode of action in FSHD patient myotubes. These findings increase our understanding of how DUX4 expression is regulated in FSHD and point to potential areas of therapeutic intervention.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação para Baixo , Ativação Enzimática , Proteínas de Homeodomínio/genética , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Agonistas Adrenérgicos beta/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Regulação para Baixo/efeitos dos fármacos , Descoberta de Drogas , Ativação Enzimática/efeitos dos fármacos , Humanos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Distrofia Muscular Facioescapuloumeral/tratamento farmacológico , Distrofia Muscular Facioescapuloumeral/metabolismo
11.
J Pharmacol Exp Ther ; 369(2): 188-199, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30819762

RESUMO

The anabolic effects of ß 2-adrenoceptor (ß 2-AR) agonists on skeletal muscle have been demonstrated in various species. However, the clinical use of ß 2-AR agonists for skeletal muscle wasting conditions has been limited by their undesired cardiovascular effects. Here, we describe the preclinical pharmacological profile of a novel 5-hydroxybenzothiazolone (5-HOB) derived ß 2-AR agonist in comparison with formoterol as a representative ß 2-AR agonist that have been well characterized. In vitro, 5-HOB has nanomolar affinity for the human ß 2-AR and selectivity over the ß 1-AR and ß 3-AR. 5-HOB also shows potent agonistic activity at the ß 2-AR in primary skeletal muscle myotubes and induces hypertrophy of skeletal muscle myotubes. Compared with formoterol, 5-HOB demonstrates comparable full-agonist activity on cAMP production in skeletal muscle cells and skeletal muscle tissue-derived membranes. In contrast, a greatly reduced intrinsic activity was determined in cardiomyocytes and cell membranes prepared from the rat heart. In addition, 5-HOB shows weak effects on chronotropy, inotropy, and vascular relaxation compared with formoterol. In vivo, 5-HOB significantly increases hind limb muscle weight in rats with attenuated effects on heart weight and ejection fraction, unlike formoterol. Furthermore, changes in cardiovascular parameters after bolus subcutaneous treatment in rats and rhesus monkeys are significantly lower with 5-HOB compared with formoterol. In conclusion, the pharmacological profile of 5-HOB indicates superior tissue selectivity compared with the conventional ß 2-AR agonist formoterol in preclinical studies and supports the notion that such tissue-selective agonists should be investigated for the safe treatment of muscle-wasting conditions without cardiovascular limiting effects.


Assuntos
Benzotiazóis/química , Benzotiazóis/farmacologia , Sistema Cardiovascular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Receptores Adrenérgicos beta 2/metabolismo , Segurança , Agonistas de Receptores Adrenérgicos beta 2/efeitos adversos , Agonistas de Receptores Adrenérgicos beta 2/química , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Agonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Anabolizantes/efeitos adversos , Anabolizantes/química , Anabolizantes/farmacologia , Anabolizantes/uso terapêutico , Animais , Benzotiazóis/efeitos adversos , Benzotiazóis/uso terapêutico , Células CHO , Cricetulus , Coração/efeitos dos fármacos , Humanos , Hipertrofia/tratamento farmacológico , Cinética , Macaca mulatta , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miócitos Cardíacos/efeitos dos fármacos , Ratos
12.
J Cell Sci ; 129(5): 898-911, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26769899

RESUMO

Neuromuscular junctions (NMJs), the synapses made by motor neurons on muscle fibers, form during embryonic development but undergo substantial remodeling postnatally. Several lines of evidence suggest that α-dystrobrevin, a component of the dystrophin-associated glycoprotein complex (DGC), is a crucial regulator of the remodeling process and that tyrosine phosphorylation of one isoform, α-dystrobrevin-1, is required for its function at synapses. We identified a functionally important phosphorylation site on α-dystrobrevin-1, generated phosphorylation-specific antibodies to it and used them to demonstrate dramatic increases in phosphorylation during the remodeling period, as well as in nerve-dependent regulation in adults. We then identified proteins that bind to this site in a phosphorylation-dependent manner and others that bind to α-dystrobrevin-1 in a phosphorylation-independent manner. They include multiple members of the DGC, as well as α-catulin, liprin-α1, Usp9x, PI3K, Arhgef5 and Grb2. Finally, we show that two interactors, α-catulin (phosphorylation independent) and Grb2 (phosphorylation dependent) are localized to NMJs in vivo, and that they are required for proper organization of neurotransmitter receptors on myotubes.


Assuntos
Proteínas Associadas à Distrofina/metabolismo , Proteína Adaptadora GRB2/metabolismo , Junção Neuromuscular/metabolismo , Neuropeptídeos/metabolismo , Receptores Colinérgicos/metabolismo , alfa Catenina/metabolismo , Animais , Linhagem Celular , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Mapas de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Transporte Proteico , Transmissão Sináptica
13.
Proc Natl Acad Sci U S A ; 111(44): 15756-61, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25336758

RESUMO

Peroxisome proliferator-activated receptor gamma coactivator 1-alpha 4 (PGC-1α4) is a protein isoform derived by alternative splicing of the PGC1α mRNA and has been shown to promote muscle hypertrophy. We show here that G protein-coupled receptor 56 (GPR56) is a transcriptional target of PGC-1α4 and is induced in humans by resistance exercise. Furthermore, the anabolic effects of PGC-1α4 in cultured murine muscle cells are dependent on GPR56 signaling, because knockdown of GPR56 attenuates PGC-1α4-induced muscle hypertrophy in vitro. Forced expression of GPR56 results in myotube hypertrophy through the expression of insulin-like growth factor 1, which is dependent on Gα12/13 signaling. A murine model of overload-induced muscle hypertrophy is associated with increased expression of both GPR56 and its ligand collagen type III, whereas genetic ablation of GPR56 expression attenuates overload-induced muscle hypertrophy and associated anabolic signaling. These data illustrate a signaling pathway through GPR56 which regulates muscle hypertrophy associated with resistance/loading-type exercise.


Assuntos
Regulação da Expressão Gênica/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Condicionamento Físico Animal , Receptores Acoplados a Proteínas G/biossíntese , Transdução de Sinais/fisiologia , Animais , Colágeno Tipo III/biossíntese , Hipertrofia/metabolismo , Fator de Crescimento Insulin-Like I/biossíntese , Masculino , Camundongos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/metabolismo
14.
Crit Rev Biochem Mol Biol ; 49(1): 59-68, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24237131

RESUMO

The molecular mechanisms underlying skeletal muscle maintenance involve interplay between multiple signaling pathways. Under normal physiological conditions, a network of interconnected signals serves to control and coordinate hypertrophic and atrophic messages, culminating in a delicate balance between muscle protein synthesis and proteolysis. Loss of skeletal muscle mass, termed "atrophy", is a diagnostic feature of cachexia seen in settings of cancer, heart disease, chronic obstructive pulmonary disease, kidney disease, and burns. Cachexia increases the likelihood of death from these already serious diseases. Recent studies have further defined the pathways leading to gain and loss of skeletal muscle as well as the signaling events that induce differentiation and post-injury regeneration, which are also essential for the maintenance of skeletal muscle mass. In this review, we summarize and discuss the relevant recent literature demonstrating these previously undiscovered mediators governing anabolism and catabolism of skeletal muscle.


Assuntos
Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Transdução de Sinais , Animais , Caquexia/metabolismo , Caquexia/patologia , Humanos , Hipertrofia/metabolismo , Hipertrofia/patologia , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia
16.
J Mol Cell Cardiol ; 72: 168-76, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24650875

RESUMO

BACKGROUND: Mechanical unloading of the failing human heart induces profound cardiac changes resulting in the reversal of a distorted structure and function. In this process, cardiomyocytes break down unneeded proteins and replace those with new ones. The specificity of protein degradation via the ubiquitin proteasome system is regulated by ubiquitin ligases. Over-expressing the ubiquitin ligase MAFbx/Atrogin-1 in the heart inhibits the development of cardiac hypertrophy, but the role of MAFbx/Atrogin-1 in the unloaded heart is not known. METHODS AND RESULTS: Mechanical unloading, by heterotopic transplantation, decreased heart weight and cardiomyocyte cross-sectional area in wild type mouse hearts. Unexpectedly, MAFbx/Atrogin-1(-/-) hearts hypertrophied after transplantation (n=8-10). Proteasome activity and markers of autophagy were increased to the same extent in WT and MAFbx/Atrogin-1(-/-) hearts after transplantation (unloading). Calcineurin, a regulator of cardiac hypertrophy, was only upregulated in MAFbx/Atrogin-1(-/-) transplanted hearts, while the mTOR pathway was similarly activated in unloaded WT and MAFbx/Atrogin-1(-/-) hearts. MAFbx/Atrogin-1(-/-) cardiomyocytes exhibited increased calcineurin protein expression, NFAT transcriptional activity, and protein synthesis rates, while inhibition of calcineurin normalized NFAT activity and protein synthesis. Lastly, mechanical unloading of failing human hearts with a left ventricular assist device (n=18) also increased MAFbx/Atrogin-1 protein levels and expression of NFAT regulated genes. CONCLUSIONS: MAFbx/Atrogin-1 is required for atrophic remodeling of the heart. During unloading, MAFbx/Atrogin-1 represses calcineurin-induced cardiac hypertrophy. Therefore, MAFbx/Atrogin-1 not only regulates protein degradation, but also reduces protein synthesis, exerting a dual role in regulating cardiac mass.


Assuntos
Cardiomegalia/genética , Transplante de Coração , Proteínas Musculares/genética , Miócitos Cardíacos/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Adulto , Idoso , Animais , Calcineurina/genética , Calcineurina/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/cirurgia , Feminino , Regulação da Expressão Gênica , Coração/fisiopatologia , Coração Auxiliar , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas Musculares/deficiência , Proteínas Musculares/metabolismo , Miócitos Cardíacos/patologia , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Cultura Primária de Células , Proteólise , Proteínas Ligases SKP Culina F-Box/deficiência , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais , Transplante Heterotópico , Remodelação Ventricular
17.
Am J Physiol Endocrinol Metab ; 306(2): E150-6, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24253050

RESUMO

A splice form of IGF-1, IGF-1Eb, is upregulated after exercise or injury. Physiological responses have been ascribed to the 24-amino acid COOH-terminal peptide that is cleaved from the NH3-terminal 70-amino acid mature IGF-1 protein. This COOH-terminal peptide was termed "mechano-growth factor" (MGF). Activities claimed for the MGF peptide included enhancing muscle satellite cell proliferation and delaying myoblast fusion. As such, MGF could represent a promising strategy to improve muscle regeneration. Thus, at our two pharmaceutical companies, we attempted to reproduce the claimed effect of MGF peptides on human and mouse muscle myoblast proliferation and differentiation in vitro. Concentrations of peptide up to 500 ng/ml failed to increase the proliferation of C2C12 cells or primary human skeletal muscle myoblasts. In contrast, all cell types exhibited a proliferative response to mature IGF-1 or full-length IGF-1Eb. MGF also failed to inhibit the differentiation of myoblasts into myotubes. To address whether the response to MGF was lost in these tissue culture lines, we measured proliferation and differentiation of primary mouse skeletal muscle stem cells exposed to MGF. This, too, failed to demonstrate a significant effect. Finally, we tested whether MGF could alter a separate documented in vitro effect of the peptide, activation of p-ERK, but not p-Akt, in cardiac myocytes. Although a robust response to IGF-1 was observed, there were no demonstrated activating responses from the native or a stabilized MGF peptide. These results call in to question whether there is a physiological role for MGF.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Mioblastos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Fator de Crescimento Insulin-Like I/química , Fator de Crescimento Insulin-Like I/farmacologia , Camundongos , Mioblastos/fisiologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Cultura Primária de Células , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Células-Tronco/fisiologia
18.
Annu Rev Pharmacol Toxicol ; 51: 373-95, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20936944

RESUMO

Loss of muscle mass can be the consequence of pathological changes, as observed in muscular dystrophies; or it can be secondary to cachexia-inducing diseases that cause muscle atrophy, such as cancer, heart disease, or chronic obstructive pulmonary disease; or it can be a consequence of aging or simple disuse. Although muscular dystrophies are rare, muscle loss affects millions of people worldwide. We discuss the molecular mechanisms involved in muscular dystrophy and in muscle atrophy and present current strategies aimed at ameliorating these diseases. Finally, we discuss whether lessons learned from studying muscular dystrophies will also be helpful for halting muscle loss secondary to nondystrophic diseases and whether strategies to halt muscle atrophy have potential for the treatment of muscular dystrophies.


Assuntos
Atrofia Muscular/terapia , Doenças Musculares/terapia , Distrofias Musculares/terapia , Envelhecimento , Animais , Caquexia/etiologia , Caquexia/fisiopatologia , Caquexia/terapia , Cardiopatias/complicações , Humanos , Atrofia Muscular/etiologia , Atrofia Muscular/fisiopatologia , Doenças Musculares/fisiopatologia , Distrofias Musculares/fisiopatologia , Neoplasias/complicações , Doença Pulmonar Obstrutiva Crônica/complicações
19.
Cell Biochem Funct ; 32(1): 39-50, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23512667

RESUMO

AIMS: Muscle ring finger (MuRF) proteins have been implicated in the transmission of mechanical forces to nuclear cell signaling pathways through their association with the sarcomere. We recently reported that MuRF1, but not MuRF2, regulates pathologic cardiac hypertrophy in vivo. This was surprising given that MuRF1 and MuRF2 interact with each other and many of the same sarcomeric proteins experimentally. METHODS AND RESULTS: Mice missing all four MuRF1 and MuRF2 alleles [MuRF1/MuRF2 double null (DN)] were born with a massive spontaneous hypertrophic cardiomyopathy and heart failure; mice that were null for one of the genes but heterozygous for the other (i.e. MuRF1(-/-) //MuRF2(+/-) or MuRF1(+/-) //MuRF2(-/-) ) were phenotypically identical to wild-type mice. Microarray analysis of genes differentially-expressed between MuRF1/MuRF2 DN, mice missing three of the four alleles and wild-type mice revealed a significant enrichment of genes regulated by the E2F transcription factor family. More than 85% of the differentially-expressed genes had E2F promoter regions (E2f:DP; P<0.001). Western analysis of E2F revealed no differences between MuRF1/MuRF2 DN hearts and wild-type hearts; however, chromatin immunoprecipitation studies revealed that MuRF1/MuRF2 DN hearts had significantly less binding of E2F1 in the promoter regions of genes previously defined to be regulated by E2F1 (p21, Brip1 and PDK4, P<0.01). CONCLUSIONS: These studies suggest that MuRF1 and MuRF2 play a redundant role in regulating developmental physiologic hypertrophy, by regulating E2F transcription factors essential for normal cardiac development by supporting E2F localization to the nucleus, but not through a process that degrades the transcription factor.


Assuntos
Cardiomegalia/metabolismo , Coração/fisiologia , Proteínas Musculares/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Animais Recém-Nascidos , Cardiomegalia/genética , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Expressão Gênica , Coração/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Regiões Promotoras Genéticas , Sarcômeros/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/genética
20.
J Mol Biol ; 436(1): 168372, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37979908

RESUMO

Brca1 mouse models were first reported in the mid-1990's shortly after cloning the human gene. Since then, many mouse models with a range of mutations have been generated, some mimic patient mutations, others are designed to probe specific protein domains and functions. In this review, we discuss early and recent studies using engineered Brca1 mouse alleles, and their implications for understanding Brca1 protein function in the context of DNA repair, tumorigenesis, and anti-cancer therapeutics.


Assuntos
Proteína BRCA1 , Neoplasias Experimentais , Animais , Humanos , Camundongos , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Reparo do DNA , Mutação , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA