Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 40(48): 6614-6626, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34621020

RESUMO

KIT/PDGFRA oncogenic tyrosine kinase signaling is the central oncogenic event in most gastrointestinal stromal tumors (GIST), which are human malignant mesenchymal neoplasms that often feature myogenic differentiation. Although targeted inhibition of KIT/PDGFRA provides substantial clinical benefit, GIST cells adapt to KIT/PDGFRA driver suppression and eventually develop resistance. The specific molecular events leading to adaptive resistance in GIST remain unclear. By using clinically representative in vitro and in vivo GIST models and GIST patients' samples, we found that the E3 ubiquitin ligase Atrogin-1 (FBXO32)-the main effector of muscular atrophy in cachexia-resulted in the most critical gene derepressed in response to KIT inhibition, regardless the type of KIT primary or secondary mutation. Atrogin-1 in GISTs is transcriptionally controlled by the KIT-FOXO3a axis, thus indicating overlap with Atrogin-1 regulation mechanisms in nonneoplastic muscle cells. Further, Atrogin-1 overexpression was a GIST-cell-specific pro-survival mechanism that enabled the adaptation to KIT-targeted inhibition by apoptosis evasion through cell quiescence. Buttressed on these findings, we established in vitro and in vivo the preclinical proof-of-concept for co-targeting KIT and the ubiquitin pathway to maximize the therapeutic response to first-line imatinib treatment.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Mesilato de Imatinib/farmacologia , Proteínas Musculares/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Sulfetos/farmacologia , Sulfonamidas/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Quimioterapia Combinada , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/metabolismo , Neoplasias Gastrointestinais/patologia , Tumores do Estroma Gastrointestinal/metabolismo , Tumores do Estroma Gastrointestinal/patologia , Humanos , Camundongos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Balkan Med J ; 38(1): 43-49, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32975519

RESUMO

BACKGROUND: Multiple myeloma remains a virtually incurable hematologic malignancy, which is featured with the aberrant growth of malignant plasma cells. AIMS: To elucidate the functions of miR-19a-3p in multiple myeloma. STUDY DESIGN: Cell study. METHODS: Cell counting kit-8 assay was performed to detect cell viability, and flow cytometry was conducted to detect cell apoptosis. Bioinformatics analysis predicted miR-19a-3p-associated biological function, pathway, core regulatory network, and target genes. Luciferase reporter assay verified the target sequence of miR-19a-3p regulating FBXO32. RESULTS: miR-19a-3p is upregulated in multiple myeloma cells (p<0.01) and patients with multiple myeloma (p<0.001). Overexpressed miR-19a-3p significantly increased cell viability (p<0.05) and inhibited cell apoptosis (p<0.01). FBXO32 is a target gene of miR-19a-3p (p<0.01). Moreover, FBXO32 is downregulated in MM, and it significantly decreased cell viability (p<0.05) and promoted cell apoptosis (p<0.01). FBXO32 significantly rescued the influence of miR-19a-3p-inhibiting cell apoptosis (p<0.05). CONCLUSION: miR-19a-3p promoted cell proliferation and inhibited cell apoptosis by degrading the target FBXO32 mRNA in multiple myeloma.


Assuntos
MicroRNAs/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Proteínas Musculares/antagonistas & inibidores , Oncogenes/efeitos dos fármacos , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Adulto , Apoptose/efeitos dos fármacos , Feminino , Humanos , Masculino , MicroRNAs/uso terapêutico , Pessoa de Meia-Idade , Mieloma Múltiplo/fisiopatologia , Proteínas Musculares/uso terapêutico , Oncogenes/fisiologia , Proteínas Ligases SKP Culina F-Box/uso terapêutico
3.
Chem Biol Interact ; 304: 43-51, 2019 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-30849338

RESUMO

Accumulations of uremic toxins has been widely recognized as the major trigger of skeletal muscle loss in chronic kidney disease (CKD), which is defined as uremic sarcopenia. Current study was aimed to examine the effects of representative uremic toxin, indoxyl sulfate (IS), on C2C12 myotubes. The incubation of IS (from 0.1 mM to 1.2 mM) exerted the reduction in myotube diameter without cell survival impairment. Elevated oxidative stress and mitogen-activated protein kinase (MAPKs) phosphorylation were observed after IS stimulation for 1 and 24 h. After N-acetylcysteine (NAC) treatment as antioxidants, the recovery in IS-induced decrease myotube diameter and ERK phosphorylation was observed. This findings were implicit the transduction of p-ERK in IS-induced ROS toxicity. Moreover, the increase of LC3ß was found closely with IS treatment in C2C12 myotubes. The reverse effect of NAC on LC3ß expression revealed the ROS-responsibility in autophagy regulation of CKD myopathy. The evaluation of IS-treated proteasome system showed increased phospho-myosin light chain, along with the upregulation of muscle atrophy F-box (MAFbx) mRNA and protein. This alteration in MAFbx was also identified in nephrectomy-induced CKD model. Besides, the inhibition of p-JNK was capable to attenuate IS-induced upward change in MAFbx protein expression. These findings indicated that IS-mediated myotube atrophy may manipulate through ROS-ERK axis and JNK-MAFbx regulation in C2C12 cells.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Indicã/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Musculares/antagonistas & inibidores , Atrofia Muscular/induzido quimicamente , Espécies Reativas de Oxigênio/metabolismo , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Camundongos , Proteínas Musculares/metabolismo , Atrofia Muscular/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Relação Estrutura-Atividade
4.
Biol Reprod ; 100(4): 896-906, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30535233

RESUMO

The mechanism of maternal protein degradation during preimplantation development has not been clarified yet. It is thought that a lot of maternal proteins are degraded by the ubiquitin-proteasome system. In this study, we focused on the role of the SCF (Skp1-Cullin-F-box) complexes during early bovine embryogenesis. We inhibited them using MLN4924, an inhibitor of SCF complex ligases controlled by neddylation. Oocytes maturated in MLN4924 could be fertilized, but we found no cumulus cell expansion and a high number of polyspermy after in vitro fertilization. We also found a statistically significant deterioration of development after MLN4924 treatment. After treatment with MLN4924 from the four-cell to late eight-cell stage, we found a statistically significant delay in their development; some of the treated embryos were, however, able to reach the blastocyst stage later. We found reduced levels of mRNA of EGA markers PAPOLA and U2AF1A, which can be related to this developmental delay. The cultivation with MLN4924 caused a significant increase in protein levels in MLN4924-treated oocytes and embryos; no such change was found in cumulus cells. To detect the proteins affected by MLN4924 treatment, we performed a Western blot analysis of selected proteins (SMAD4, ribosomal protein S6, centromeric protein E, P27, NFKB inhibitor alpha, RNA-binding motif protein 19). No statistically significant increase in protein levels was detected in either treated embryos or oocytes. In summary, our study shows that SCF ligases are necessary for the correct maturation of oocytes, cumulus cell expansion, fertilization, and early preimplantation development of cattle.


Assuntos
Blastocisto/efeitos dos fármacos , Ciclopentanos/farmacologia , Desenvolvimento Embrionário/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Oogênese/efeitos dos fármacos , Pirimidinas/farmacologia , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Animais , Blastocisto/citologia , Blastocisto/fisiologia , Bovinos , Células Cultivadas , Embrião de Mamíferos , Feminino , Técnicas de Maturação in Vitro de Oócitos/métodos , Técnicas de Maturação in Vitro de Oócitos/veterinária , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/metabolismo , Oócitos/citologia , Oócitos/fisiologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas Ligases SKP Culina F-Box/fisiologia , Fatores de Tempo
5.
Nature ; 563(7733): 652-656, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30464344

RESUMO

The strigolactones, a class of plant hormones, regulate many aspects of plant physiology. In the inhibition of shoot branching, the α/ß hydrolase D14-which metabolizes strigolactone-interacts with the F-box protein D3 to ubiquitinate and degrade the transcription repressor D53. Despite the fact that multiple modes of interaction between D14 and strigolactone have recently been determined, how the hydrolase functions with D3 to mediate hormone-dependent D53 ubiquitination remains unknown. Here we show that D3 has a C-terminal α-helix that can switch between two conformational states. The engaged form of this α-helix facilitates the binding of D3 and D14 with a hydrolysed strigolactone intermediate, whereas the dislodged form can recognize unmodified D14 in an open conformation and inhibits its enzymatic activity. The D3 C-terminal α-helix enables D14 to recruit D53 in a strigolactone-dependent manner, which in turn activates the hydrolase. By revealing the structural plasticity of the SCFD3-D14 ubiquitin ligase, our results suggest a mechanism by which the E3 coordinates strigolactone signalling and metabolism.


Assuntos
Compostos Heterocíclicos com 3 Anéis/metabolismo , Lactonas/metabolismo , Oryza/enzimologia , Oryza/metabolismo , Reguladores de Crescimento de Plantas/metabolismo , Proteínas Ligases SKP Culina F-Box/química , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais , Compostos Heterocíclicos com 3 Anéis/química , Lactonas/química , Modelos Moleculares , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Reguladores de Crescimento de Plantas/química , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Relação Estrutura-Atividade , Ubiquitina , Ubiquitinação
6.
Structure ; 26(9): 1226-1236.e3, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30033217

RESUMO

Skp1-Cul1-F-box (SCF) E3 ligases constitute the largest and best-characterized family of the multisubunit E3 ligases with important cellular functions and numerous disease links. The specificity of an SCF E3 ligase is established by one of the 69 human F-box proteins that are recruited to Cul1 through the Skp1 adaptor. We previously reported generation of ubiquitin variants (UbVs) targeting Fbw7 and Fbw11, which inhibit ligase activity by binding at the F-box-Skp1 interface to competitively displace Cul1. In the present study, we employed an optimized engineering strategy to generate specific binding UbVs against 17 additional Skp1-F-box complexes. We validated our design strategy and uncovered the structural basis of binding specificity by crystallographic analyses of representative UbVs bound to Skp1-Fbl10 and Skp1-Fbl11. Our study highlights the power of combining phage display with structure-based design to develop UbVs targeting specific protein surfaces.


Assuntos
Proteínas Ligases SKP Culina F-Box/química , Proteínas Ligases SKP Culina F-Box/metabolismo , Ubiquitinas/química , Ubiquitinas/farmacologia , Sítios de Ligação , Técnicas de Visualização da Superfície Celular , Cristalografia por Raios X , Proteínas Culina/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Proteínas F-Box/metabolismo , Feminino , Células HEK293 , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Modelos Moleculares , Ligação Proteica , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade
7.
Life Sci ; 205: 91-96, 2018 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-29753766

RESUMO

AIMS: Muscle mass is regulated by the balance between the synthesis and degradation of muscle proteins. Loss of skeletal muscle mass is associated with an increased risk of developing metabolic diseases such as obesity and type 2 diabetes mellitus. The aim of this study was to clarify the effects of licorice flavonoid oil on muscle mass in KK-Ay/Ta mice. MAIN METHODS: Male genetically type II diabetic KK-Ay/Ta mice received 0, 1, or 1.5 g/kg BW of licorice flavonoid oil by mouth once daily for 4 weeks. After 4 weeks, the femoral and soleus muscles were collected for western blotting for evaluation of the mTOR/p70 S6K, p38/FoxO3a, and Akt/FoxO3a signaling pathways. KEY FINDINGS: Ingestion of licorice flavonoid oil significantly enhanced femoral muscle mass without affecting body weight in KK-Ay/Ta mice. Licorice flavonoid oil also decreased expression of MuRF1 and atrogin-1, which are both markers of muscle atrophy. The mechanisms by which licorice flavonoid oil enhances muscle mass include activation of mTOR and p70 S6K, and regulation of phosphorylation of FoxO3a. SIGNIFICANCE: Ingestion of licorice flavonoids may help to prevent muscle atrophy.


Assuntos
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Flavonoides/farmacologia , Glycyrrhiza/química , Músculo Esquelético/efeitos dos fármacos , Óleos de Plantas/farmacologia , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/biossíntese , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Proteína Forkhead Box O3/biossíntese , Proteína Forkhead Box O3/genética , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/patologia , Tamanho do Órgão/efeitos dos fármacos , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Proteínas Ligases SKP Culina F-Box/biossíntese , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/biossíntese , Proteínas com Motivo Tripartido , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
8.
Open Biol ; 8(2)2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29445034

RESUMO

Deregulation of centriole duplication has been implicated in cancer and primary microcephaly. Accordingly, it is important to understand how key centriole duplication factors are regulated. E3 ubiquitin ligases have been implicated in controlling the levels of several duplication factors, including PLK4, STIL and SAS-6, but the precise mechanisms ensuring centriole homeostasis remain to be fully understood. Here, we have combined proteomics approaches with the use of MLN4924, a generic inhibitor of SCF E3 ubiquitin ligases, to monitor changes in the cellular abundance of centriole duplication factors. We identified human STIL as a novel substrate of SCF-ßTrCP. The binding of ßTrCP depends on a DSG motif within STIL, and serine 395 within this motif is phosphorylated in vivo SCF-ßTrCP-mediated degradation of STIL occurs throughout interphase and mutations in the DSG motif causes massive centrosome amplification, attesting to the physiological importance of the pathway. We also uncover a connection between this new pathway and CDK2, whose role in centriole biogenesis remains poorly understood. We show that CDK2 activity protects STIL against SCF-ßTrCP-mediated degradation, indicating that CDK2 and SCF-ßTrCP cooperate via STIL to control centriole biogenesis.


Assuntos
Centríolos/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Ciclopentanos/farmacologia , Células HEK293 , Homeostase , Humanos , Interfase , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação , Fosforilação , Proteólise , Proteômica , Pirimidinas/farmacologia , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Serina/metabolismo
9.
PLoS Genet ; 14(1): e1007165, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29370161

RESUMO

Intellectual disability (ID), one of the most common human developmental disorders, can be caused by genetic mutations in Cullin 4B (Cul4B) and cereblon (CRBN). CRBN is a substrate receptor for the Cul4A/B-DDB1 ubiquitin ligase (CRL4) and can target voltage- and calcium-activated BK channel for ER retention. Here we report that ID-associated CRL4CRBN mutations abolish the interaction of the BK channel with CRL4, and redirect the BK channel to the SCFFbxo7 ubiquitin ligase for proteasomal degradation. Glioma cell lines harbouring CRBN mutations record density-dependent decrease of BK currents, which can be restored by blocking Cullin ubiquitin ligase activity. Importantly, mice with neuron-specific deletion of DDB1 or CRBN express reduced BK protein levels in the brain, and exhibit similar impairment in learning and memory, a deficit that can be partially rescued by activating the BK channel. Our results reveal a competitive targeting of the BK channel by two ubiquitin ligases to achieve exquisite control of its stability, and support changes in neuronal excitability as a common pathogenic mechanism underlying CRL4CRBN-associated ID.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Aprendizagem/fisiologia , Memória/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Proteólise , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Células Cultivadas , Feminino , Células HEK293 , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Ligases SKP Culina F-Box/metabolismo , Complexos Ubiquitina-Proteína Ligase/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
10.
J Nutr Sci Vitaminol (Tokyo) ; 63(5): 339-348, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29225319

RESUMO

Insulin resistance reduces insulin-induced muscle protein synthesis and accelerates muscle protein degradation. Ginseng ingestion has been reported to improve insulin resistance through the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. We hypothesized that panaxatriol (PT) derived from ginseng in combination with aerobic exercise (EX) may further promote protein synthesis and suppress protein degradation, and subsequently maintain muscle mass through the amelioration of insulin resistance. KKAy insulin-resistant mice were divided into control, panaxatriol only (PT), exercise only (EX), and EX+PT groups. EX and EX+PT ran on the treadmill for 45 min at 15 m/min 5 d/wk for 6 wk. PT and EX+PT groups were fed a standard diet containing 0.2% PT for 6 wk. Homeostasis model assessment for insulin resistance (HOMA-R) values was significantly improved after exercise for 6 wk. Moreover, EX+PT mice showed improved HOMA-R as compared to EX mice. p70S6K phosphorylation after a 4 h fast was significantly higher in EX than in the non-exercise control, and it was higher in EX+PT mice than in EX mice. Atrogin1 mRNA expression was significantly lower in EX than in the non-exercise control, and was significantly lowered further by PT treatment. EX and EX+PT mice showed higher soleus muscle mass and cross-sectional area (CSA) of the soleus myofibers than control animals, with higher values noted for both parameters in EX+PT than in EX. These results suggest that aerobic exercise and PT ingestion may contribute to maintain skeletal muscle mass through the amelioration of insulin resistance.


Assuntos
Diabetes Mellitus Tipo 2/terapia , Suplementos Nutricionais , Ginsenosídeos/uso terapêutico , Hipoglicemiantes/uso terapêutico , Resistência à Insulina , Atrofia Muscular/prevenção & controle , Condicionamento Físico Animal , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Terapia Combinada , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/dietoterapia , Diabetes Mellitus Tipo 2/metabolismo , Regulação da Expressão Gênica , Inulina/sangue , Masculino , Camundongos Mutantes , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/complicações , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Panax/química , Fosforilação , Raízes de Plantas/química , Processamento de Proteína Pós-Traducional , Proteólise , RNA Mensageiro/metabolismo , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo
11.
Nutrients ; 9(9)2017 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-28930190

RESUMO

Stimulation of the ubiquitin-proteasome pathway-especially E3 ubiquitin ligases Atrogin-1 and MuRF1-is associated with muscle loss in diabetes. Elevated lipid metabolites impair myogenesis. Oligonol, a low molecular weight polyphenol derived from lychee, exhibited anti-diabetic and anti-obesity properties, suggesting it could be a proper supplement for attenuating muscle loss. Dietary (10 weeks) oligonol supplementation (20 or 200 mg/kg diet) on the skeletal muscle loss was investigated in diabetic db/db mice. Transcription factors NF-κB and FoxO3a involved in regulation of Atrogin-1 and MuRF1 were also investigated. Attenuation of muscle loss by oligonol (both doses) was associated with down-regulation of Atrogin-1 and MuRF1 gene expression. Oligonol supplementation decreased NF-κB expression in the nuclear fraction compared with db/db mice without oligonol supplement. Upregulation of sirtuin1 (SIRT1) expression prevented FoxO3a nuclear localization in db/db mice supplemented with oligonol. Marked increases in AMPKα activity and Ppara mRNA expression leading to lower lipid accumulation by oligonol provided additional benefits for attenuating muscle loss. Oligonol limited palmitate-induced senescent phenotype and cell cycle arrest and suppressed Atrogin-1 and MuRF1 mRNA expression in palmitate-treated C2C12 muscle cells, thus contributing to improving the impaired myotube formation. In conclusion, oligonol-mediated downregulation of Atrogin-1 and MuRF1 gene expression alleviates muscle loss and improves the impaired myotube formation, indicating that oligonol supplementation may be useful for the attenuation of myotube loss.


Assuntos
Catequina/análogos & derivados , Diabetes Mellitus Experimental/fisiopatologia , Litchi/química , Proteínas Musculares/antagonistas & inibidores , Fenóis/farmacologia , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Sarcopenia/tratamento farmacológico , Proteínas com Motivo Tripartido/antagonistas & inibidores , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Animais , Catequina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Frutas/química , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peso Molecular , Proteínas Musculares/genética , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Polifenóis/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Sirtuína 1/genética , Sirtuína 1/metabolismo , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/genética
12.
Am J Physiol Endocrinol Metab ; 312(6): E495-E507, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28246104

RESUMO

Cushing's syndrome is caused by overproduction of the adrenocorticotropic hormone (ACTH), which stimulates the adrenal grand to make cortisol. Skeletal muscle wasting occurs in pathophysiological response to Cushing's syndrome. The forkhead box (FOX) protein family has been implicated as a key regulator of muscle loss under conditions such as diabetes and sepsis. However, the mechanistic role of the FOXO family in ACTH-induced muscle atrophy is not understood. We hypothesized that FOXO3a plays a role in muscle atrophy through expression of the E3 ubiquitin ligases, muscle RING finger protein-1 (MuRF-1), and atrogin-1 in Cushing's syndrome. For establishment of a Cushing's syndrome animal model, Sprague-Dawley rats were implanted with osmotic minipumps containing ACTH (40 ng·kg-1·day-1). ACTH infusion significantly reduced muscle weight. In ACTH-infused rats, MuRF-1, atrogin-1, and FOXO3a were upregulated and the FOXO3a promoter was targeted by the glucocorticoid receptor (GR). Transcriptional activity and expression of FOXO3a were significantly decreased by the GR antagonist RU486. Treatment with RU486 reduced MuRF-1 and atrogin-1 expression in accordance with reduced enrichment of FOXO3a and Pol II on the promoters. Knockdown of FOXO3a prevented dexamethasone-induced MuRF-1 and atrogin-1 expression. These results indicate that FOXO3a plays a role in muscle atrophy through expression of MuRF-1 and atrogin-1 in Cushing's syndrome.


Assuntos
Síndrome de Cushing/metabolismo , Modelos Animais de Doenças , Proteína Forkhead Box O3/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/etiologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Síndrome de Cushing/patologia , Síndrome de Cushing/fisiopatologia , Proteína Forkhead Box O3/agonistas , Proteína Forkhead Box O3/antagonistas & inibidores , Proteína Forkhead Box O3/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Glucocorticoides/farmacologia , Antagonistas de Hormônios/farmacologia , Masculino , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Proteínas Musculares/agonistas , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Interferência de RNA , Ratos Sprague-Dawley , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/metabolismo , Elementos de Resposta/efeitos dos fármacos , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Proteínas Ligases SKP Culina F-Box/genética , Proteínas com Motivo Tripartido/agonistas , Proteínas com Motivo Tripartido/antagonistas & inibidores , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética
13.
EMBO J ; 36(3): 260-273, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28007894

RESUMO

The F-box protein FBXW7 is the substrate-recruiting subunit of an SCF ubiquitin ligase and a major tumor-suppressor protein that is altered in several human malignancies. Loss of function of FBXW7 results in the stabilization of numerous proteins that orchestrate cell proliferation and survival. Little is known about proteins that directly regulate the function of this protein. In the current work, we have mapped the interactome of the enigmatic pseudophosphatase STYX We reasoned that a catalytically inactive phosphatase might have adopted novel mechanisms of action. The STYX interactome contained several F-box proteins, including FBXW7. We show that STYX binds to the F-box domain of FBXW7 and disables its recruitment into the SCF complex. Therefore, STYX acts as a direct inhibitor of FBXW7, affecting the cellular levels of its substrates. Furthermore, we find that levels of STYX and FBXW7 are anti-correlated in breast cancer patients, which affects disease prognosis. We propose the STYX-FBXW7 interaction as a promising drug target for future investigations.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas F-Box/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/patologia , Proteína 7 com Repetições F-Box-WD , Humanos
14.
Appl Physiol Nutr Metab ; 41(12): 1240-1247, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27841025

RESUMO

l-Carnitine was recently found to downregulate the ubiquitin proteasome pathway (UPP) and increase insulin-like growth factor 1 concentrations in animal models. However, the effect of l-carnitine administration on disuse muscle atrophy induced by hindlimb suspension has not yet been studied. Thus, we hypothesized that l-carnitine may have a protective effect on muscle atrophy induced by hindlimb suspension via the Akt1/mTOR and/or UPP. Male Wistar rats were assigned to 3 groups: hindlimb suspension group, hindlimb suspension with l-carnitine administration (1250 mg·kg-1·day-1) group, and pair-fed group adjusted hindlimb suspension. l-Carnitine administration for 2 weeks of hindlimb suspension alleviated the decrease in weight and fiber size in the soleus muscle. In addition, l-carnitine suppressed atrogin-1 mRNA expression, which has been reported to play a pivotal role in muscle atrophy. The present study shows that l-carnitine has a protective effect against soleus muscle atrophy caused by hindlimb suspension and decreased E3 ligase messenger RNA expression, suggesting the possibility that l-carnitine protects against muscle atrophy, at least in part, through the inhibition of the UPP. These observations suggest that l-carnitine could serve as an effective supplement in the decrease of muscle atrophy caused by weightlessness in the fields of clinical and rehabilitative research.


Assuntos
Carnitina/uso terapêutico , Suplementos Nutricionais , Repressão Enzimática , Proteínas Musculares/antagonistas & inibidores , Músculo Esquelético/metabolismo , Transtornos Musculares Atróficos/prevenção & controle , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Animais , Biomarcadores/metabolismo , Elevação dos Membros Posteriores/efeitos adversos , Imuno-Histoquímica , Masculino , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Atrofia Muscular/etiologia , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Atrofia Muscular/prevenção & controle , Transtornos Musculares Atróficos/etiologia , Transtornos Musculares Atróficos/metabolismo , Transtornos Musculares Atróficos/patologia , Complexo de Endopeptidases do Proteassoma , Inibidores de Proteassoma/uso terapêutico , Distribuição Aleatória , Ratos , Ratos Wistar , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Ausência de Peso/efeitos adversos
15.
PLoS Genet ; 12(10): e1006361, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27776126

RESUMO

SKN-1/Nrf are the primary antioxidant/detoxification response transcription factors in animals and they promote health and longevity in many contexts. SKN-1/Nrf are activated by a remarkably broad-range of natural and synthetic compounds and physiological conditions. Defining the signaling mechanisms that regulate SKN-1/Nrf activation provides insights into how cells coordinate responses to stress. Nrf2 in mammals is regulated in part by the redox sensor repressor protein named Keap1. In C. elegans, the p38 MAPK cascade in the intestine activates SKN-1 during oxidative stress by promoting its nuclear accumulation. Interestingly, we find variation in the kinetics of p38 MAPK activation and tissues with SKN-1 nuclear accumulation among different pro-oxidants that all trigger strong induction of SKN-1 target genes. Using genome-wide RNAi screening, we identify new genes that are required for activation of the core SKN-1 target gene gst-4 during exposure to the natural pro-oxidant juglone. Among 10 putative activators identified in this screen was skr-1/2, highly conserved homologs of yeast and mammalian Skp1, which function to assemble protein complexes. Silencing of skr-1/2 inhibits induction of SKN-1 dependent detoxification genes and reduces resistance to pro-oxidants without decreasing p38 MAPK activation. Global transcriptomics revealed strong correlation between genes that are regulated by SKR-1/2 and SKN-1 indicating a high degree of specificity. We also show that SKR-1/2 functions upstream of the WD40 repeat protein WDR-23, which binds to and inhibits SKN-1. Together, these results identify a novel p38 MAPK independent signaling mechanism that activates SKN-1 via SKR-1/2 and involves WDR-23.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Inativação Metabólica/genética , Longevidade/genética , Proteínas Ligases SKP Culina F-Box/genética , Receptores de Ativinas Tipo I/genética , Animais , Antioxidantes/metabolismo , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/biossíntese , Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/biossíntese , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Fosforilação , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Acta Biochim Biophys Sin (Shanghai) ; 48(9): 827-32, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27521792

RESUMO

Control of cardiac muscle mass is thought to be determined by a dynamic balance of protein synthesis and degradation. Recent studies have demonstrated that atrophy-related forkhead box O 3a (FOXO3a)/muscle atrophy F-box (MAFbx) signaling pathway plays a central role in the modulation of proteolysis and exert inhibitory effect on cardiomyocyte hypertrophy. In this study, we tested the hypothesis that adenosine monophosphate-activated protein kinase (AMPK) activation attenuates cardiomyocyte hypertrophy by regulating FOXO3a/MAFbx signaling pathway and its downstream protein degradation. The results showed that activation of AMPK with 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) attenuated cardiomyocyte hypertrophy induced by angiotensin II (Ang II). The antihypertrophic effects of AICAR were blunted by AMPK inhibitor Compound C. In addition, AMPK dramatically increased the activity of transcription factor FOXO3a, up-regulated the expression of its downstream ubiquitin ligase MAFbx, and enhanced cardiomyocyte proteolysis. Meanwhile, the effects of AMPK on protein degradation and cardiomyocyte hypertrophy were blocked after MAFbx was silenced by transfection of cardiomyocytes with MAFbx-siRNA. These results indicate that AMPK plays an important role in the inhibition of cardiomyocyte hypertrophy by activating protein degradation via FOXO3a/MAFbx signaling pathway.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteína Forkhead Box O3/metabolismo , Proteínas Musculares/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Crescimento Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Miócitos Cardíacos/efeitos dos fármacos , Proteólise/efeitos dos fármacos , RNA Interferente Pequeno/genética , Ratos , Ribonucleotídeos/farmacologia , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Proteínas Ligases SKP Culina F-Box/genética , Transdução de Sinais/efeitos dos fármacos
17.
Pharmacol Res ; 111: 34-42, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27238229

RESUMO

In the last decade, the ubiquitin proteasome system (UPS), in general, and E3 ubiquitin ligases, in particular, have emerged as valid drug targets for the development of novel anti-cancer therapeutics. Cullin RING Ligases (CRLs), which can be classified into eight groups (CRL1-8) and comprise approximately 200 members, represent the largest family of E3 ubiquitin ligases which facilitate the ubiquitination-derived proteasomal degradation of a myriad of functionally and structurally diverse substrates. S phase kinase-associated protein 1 (Skp1)-Cullin1-F-Box protein (SCF) complexes are the best characterized among CRLs, which play crucial roles in numerous cellular processes and physiological dysfunctions, such as in cancer biology. Currently, there is growing interest in developing SCF-targeting anti-cancer therapies for clinical application. Indeed, the research in this field has seen some progress in the form of cullin neddylation- and Skp2-inhibitors. However, it still remains an underdeveloped area and needs to design new strategies for developing improved form of therapy. In this review, we venture a novel strategy that rational pharmacological targeting of Skp1, a central regulator of SCF complexes, may provide a novel avenue for SCF-oriented anti-cancer therapy, expected: (i) to simultaneously address the critical roles that multiple SCF oncogenic complexes play in cancer biology, (ii) to selectively target cancer cells with minimal normal cell toxicity, and (iii) to offer multiple chemical series, via therapeutic interventions at the Skp1 binding interfaces in SCF complex, thereby maximizing chances of success for drug discovery. In addition, we also discuss the challenges that might be posed regarding rational pharmacological interventions against Skp1.


Assuntos
Antineoplásicos/uso terapêutico , Descoberta de Drogas/métodos , Inibidores Enzimáticos/uso terapêutico , Neoplasias/tratamento farmacológico , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Animais , Clonagem Molecular , Desenho de Fármacos , Humanos , Terapia de Alvo Molecular , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Conformação Proteica , Proteínas Ligases SKP Culina F-Box/química , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
18.
Biosci Biotechnol Biochem ; 80(8): 1499-504, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27055887

RESUMO

To investigate the intracellular signaling mechanisms by which clenbuterol reduces muscle protein degradation, we examined the phosphorylation level and intracellular localization of FOXO1 in the sartorius muscle of neonatal chicks. One-day-old chicks were given a single intraperitoneal injection of clenbuterol (0.1 mg/kg body weight). Three hours after injection, AKT protein was phosphorylated in the sartorius muscle by clenbuterol injection. Coincidentally, clenbuterol increased cytosolic level of phosphorylated FOXO1 protein, while it decreased nuclear level of FOXO1 protein in the sartorius muscle. Furthermore, clenbuterol decreased the expression of mRNAs for muscle-specific ubiquitin ligases (atrogin-1/MAFbx and MuRF1) in the sartorius muscle accompanied by decreased plasma 3-methylhistidine concentration, an index of muscle protein degradation, at 3 h after injection. These results suggested that, in the sartorius muscle of the chicks, clenbuterol changed the intracellular localization of phosphorylated FOXO1, and consequently decreased protein degradation via suppressing the expression of genes encoding muscle-specific ubiquitin ligases.


Assuntos
Proteínas Aviárias/genética , Clembuterol/farmacologia , Proteína Forkhead Box O1/genética , Músculo Esquelético/efeitos dos fármacos , Proteínas Ligases SKP Culina F-Box/genética , Simpatomiméticos/farmacologia , Ubiquitina-Proteína Ligases/genética , Animais , Animais Recém-Nascidos , Proteínas Aviárias/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Galinhas , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Injeções Intraperitoneais , Metilistidinas/sangue , Músculo Esquelético/metabolismo , Fosforilação/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Proteínas Ligases SKP Culina F-Box/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/metabolismo
19.
Proc Natl Acad Sci U S A ; 113(13): 3527-32, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26976582

RESUMO

Skp1-Cul1-F-box (SCF) E3 ligases play key roles in multiple cellular processes through ubiquitination and subsequent degradation of substrate proteins. Although Skp1 and Cul1 are invariant components of all SCF complexes, the 69 different human F-box proteins are variable substrate binding modules that determine specificity. SCF E3 ligases are activated in many cancers and inhibitors could have therapeutic potential. Here, we used phage display to develop specific ubiquitin-based inhibitors against two F-box proteins, Fbw7 and Fbw11. Unexpectedly, the ubiquitin variants bind at the interface of Skp1 and F-box proteins and inhibit ligase activity by preventing Cul1 binding to the same surface. Using structure-based design and phage display, we modified the initial inhibitors to generate broad-spectrum inhibitors that targeted many SCF ligases, or conversely, a highly specific inhibitor that discriminated between even the close homologs Fbw11 and Fbw1. We propose that most F-box proteins can be targeted by this approach for basic research and for potential cancer therapies.


Assuntos
Proteínas Culina/metabolismo , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Ubiquitinas/farmacologia , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas Culina/química , Desenho de Fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Proteínas F-Box/antagonistas & inibidores , Proteínas F-Box/química , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Variação Genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Biblioteca de Peptídeos , Engenharia de Proteínas , Domínios e Motivos de Interação entre Proteínas , Proteínas Ligases SKP Culina F-Box/química , Proteínas Ligases SKP Culina F-Box/genética , Homologia de Sequência de Aminoácidos , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética , Ubiquitinas/química , Ubiquitinas/genética , Proteínas Contendo Repetições de beta-Transducina/antagonistas & inibidores , Proteínas Contendo Repetições de beta-Transducina/química , Proteínas Contendo Repetições de beta-Transducina/genética
20.
PLoS One ; 10(8): e0135591, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26295339

RESUMO

Alpha-1 antitrypsin deficiency is the leading cause of childhood liver failure and one of the most common lethal genetic diseases. The disease-causing mutant A1AT-Z fails to fold correctly and accumulates in the endoplasmic reticulum (ER) of the liver, resulting in hepatic fibrosis and hepatocellular carcinoma in a subset of patients. Furthermore, A1AT-Z sequestration in hepatocytes leads to a reduction in A1AT secretion into the serum, causing panacinar emphysema in adults. The purpose of this work was to elucidate the details by which A1AT-Z is degraded in hepatic cell lines. We identified the ubiquitin ligase FBG1, which has been previously shown to degrade proteins by both the ubiquitin proteasome pathway and autophagy, as being key to A1AT-Z degradation. Using chemical and genetic approaches we show that FBG1 degrades A1AT-Z through both the ubiquitin proteasome system and autophagy. Overexpression of FBG1 decreases the half-life of A1AT-Z and knocking down FBG1 in a hepatic cell line, and in mice results in an increase in ATAT. Finally, we show that FBG1 degrades A1AT-Z through a Beclin1-dependent arm of autophagy. In our model, FBG1 acts as a safety ubiquitin ligase, whose function is to re-ubiquitinate ER proteins that have previously undergone de-ubiquitination to ensure they are degraded.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Autofagia/genética , Proteínas de Membrana/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Deficiência de alfa 1-Antitripsina/genética , alfa 1-Antitripsina/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Linhagem Celular Tumoral , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Feminino , Regulação da Expressão Gênica , Meia-Vida , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Ligação Proteica , Proteólise , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais , Ubiquitinação , alfa 1-Antitripsina/metabolismo , Deficiência de alfa 1-Antitripsina/metabolismo , Deficiência de alfa 1-Antitripsina/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...