Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Oral Biosci ; 65(4): 334-346, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37716425

RESUMO

OBJECTIVE: We previously reported that dickkopf WNT signaling pathway inhibitor 3 (DKK3) expression is correlated with poorer prognosis in head and neck squamous cell carcinoma (HNSCC). Here we investigated DKK3 expression by using The Cancer Genome Atlas (TCGA) public database and bioinformatic analyses. METHODS: We used the RNA sequence data and divided the tumor samples into "DKK3-high" and "DKK3-low" groups according to median DKK3 expression. The correlations between DKK3 expression and the clinical data were investigated. Differentially expressed genes (DEGs) were detected using DESEq2 and analyzed by ShinyGO 0.77. A gene set enrichment analysis (GSEA) was also performed using GSEA software. The DEGs were also analyzed with TargetMine to establish the protein-protein interaction (PPI) network. RESULTS: DKK3 expression was significantly increased in cancer samples, and a high DKK3 expression was significantly associated with shorter overall survival. We identified 854 DEGs, including 284 up-regulated and 570 down-regulated. Functional enrichment analyses revealed several Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways associated with extracellular matrix remodeling. The PPI network identified COL8A1, AGTR1, FN1, P4HA3, PDGFRB, and CEP126 as the key genes. CONCLUSIONS: These results suggested the cancer-promoting ability of DKK3, the expression of which is a promising prognostic marker and therapeutic target for HNSCC.


Assuntos
Neoplasias de Cabeça e Pescoço , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/genética , Regulação Neoplásica da Expressão Gênica/genética , Prognóstico , Biologia Computacional/métodos , Bases de Dados Genéticas , Pró-Colágeno-Prolina Dioxigenase/genética , Proteínas Adaptadoras de Transdução de Sinal/genética
2.
Am J Pathol ; 193(5): 591-607, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36773783

RESUMO

α-Klotho is a longevity-related protein. Its deficiency shortens lifespan with prominent senescent phenotypes, including muscle atrophy and weakness in mice. α-Klotho has two forms: membrane α-Klotho and circulating α-Klotho (c-α-Klotho). Loss of membrane α-Klotho impairs a phosphaturic effect, thereby accelerating phosphate-induced aging. However, the mechanisms of senescence on c-α-Klotho loss remain largely unknown. Herein, with the aging of wild-type mice, c-α-Klotho declined, whereas Smad2, an intracellular transforming growth factor (TGF)-ß effector, became activated in skeletal muscle. Moreover, c-α-Klotho suppressed muscle-wasting TGF-ß molecules, including myostatin, growth and differentiation factor 11, activin, and TGF-ß1, through binding to ligands as well as type I and type II serine/threonine kinase receptors. Indeed, c-α-Klotho reversed impaired in vitro myogenesis caused by these TGF-ßs. Oral administration of Ki26894, a small-molecule inhibitor of type I receptors for these TGF-ßs, restored muscle atrophy and weakness in α-Klotho (-/-) mice and in elderly wild-type mice by suppression of activated Smad2 and up-regulated Cdkn1a (p21) transcript, a target of phosphorylated Smad2. Ki26894 also induced the slow to fast myofiber switch. These findings show c-α-Klotho's potential as a circulating inhibitor counteracting TGF-ß-induced sarcopenia. These data highlight the potential of a novel therapy involving TGF-ß blockade to prevent sarcopenia.


Assuntos
Sarcopenia , Fator de Crescimento Transformador beta , Camundongos , Animais , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Sarcopenia/prevenção & controle , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fatores de Crescimento Transformadores
3.
Oral Dis ; 29(8): 3193-3204, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35708905

RESUMO

OBJECTIVE: We previously reported that dickkopf WNT signaling inhibitor 3 (DKK3) would modulate malignant potential of oral squamous cell carcinoma (OSCC) via activating Akt. Recently, cytoskeleton associated protein 4 (CKAP4) functions as receptor of DKK3, which activates Akt in esophageal squamous cell carcinoma, but its expression and function in OSCC were unclear. METHODS: We studied DKK3 and CKAP4 protein expression in OSCC tissue and investigated the correlation between protein expression and clinical data. We also investigated whether antibodies (Ab) for DKK3 or CKAP4 could suppress malignant potential of the cancer cells. RESULTS: DKK3/CKAP4 protein expression was observed in majority of OSCC cases and was associated with significantly higher T-stage and TNM stage. Multivariate analysis revealed that DKK3 and CKAP4 were independent prognostic biomarkers for overall survival (OS) and disease-free survival (DFS), respectively. Survival analyses revealed that DKK3-positive cases and CKAP4-positive cases showed significantly shorter OS and DFS, respectively, and that DKK3/CKAP4 double-negative cases showed significantly favorable prognosis. Both anti-DKK3Ab and anti-CKAP4Ab could suppress cancer cell proliferation, migration, and invasion. CONCLUSION: DKK3/CKAP4 axis is thought to be important in OSCC, and it would be a promising therapeutic target.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Carcinoma de Células Escamosas/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço , Prognóstico , Proliferação de Células , Linhagem Celular Tumoral , Proteínas de Membrana/metabolismo , Proteínas Adaptadoras de Transdução de Sinal
4.
Cancer Cell Int ; 22(1): 352, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36376957

RESUMO

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is the most common malignant tumor of the head and neck. We identified cancer-specific genes in HNSCC and focused on DKK3 expression. DKK3 gene codes two isoforms of proteins (secreted and non-secreted) with two distinct cysteine rich domains (CRDs). It is reported that DKK3 functions as a negative regulator of oncogenic Wnt signaling and, is therefore, considered to be a tumor suppressor gene. However, our series of studies have demonstrated that DKK3 expression is specifically high in HNSCC tissues and cells, and that DKK3 might determine the malignant potentials of HNSCC cells via the activation of Akt. Further analyses strongly suggested that both secreted DKK3 and non-secreted DKK3 could activate Akt signaling in discrete ways, and consequently exert tumor promoting effects. We hypothesized that DKK3 might be a specific druggable target, and it is necessary to establish a DKK3 inhibitor that can inhibit both secreted and non-secreted isoforms of DKK3. METHODS: Using inverse polymerase chain reaction, we generated mutant expression plasmids that express DKK3 without CRD1, CRD2, or both CRD1 and CRD2 (DKK3ΔC1, DKK3ΔC2, and DKK3ΔC1ΔC2, respectively). These plasmids were then transfected into HNSCC-derived cells to determine the domain responsible for DKK3-mediated Akt activation. We designed antisense peptides using the MIMETEC program, targeting DKK3-specific amino acid sequences within CRD1 and CRD2. The structural models for peptides and DKK3 were generated using Raptor X, and then a docking simulation was performed using CluPro2. Afterward, the best set of the peptides was applied into HNSCC-derived cells, and the effects on Akt phosphorylation, cellular proliferation, invasion, and migration were assessed. We also investigated the therapeutic effects of the peptides in the xenograft models. RESULTS: Transfection of mutant expression plasmids and subsequent functional analyses revealed that it is necessary to delete both CRD1 and CRD2 to inhibit Akt activation and inhibition of proliferation, migration, and invasion. The inhibitory peptides for CRD1 and CRD2 of DKK3 significantly reduced the phosphorylation of Akt, and consequently suppressed cellular proliferation, migration, invasion and in vivo tumor growth at very low doses. CONCLUSIONS: This inhibitory peptide represents a promising new therapeutic strategy for HNSCC treatment.

5.
Biochem Biophys Res Commun ; 628: 84-90, 2022 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-36084555

RESUMO

Mutations of the caveolin 3 gene cause autosomal dominant limb-girdle muscular dystrophy (LGMD)1C. In mice, overexpression of mutant caveolin 3 leads to loss of caveolin 3 and results in myofiber hypotrophy in association with activation of neuronal nitric oxide synthase (nNOS) at the sarcolemma. Here, we show that caveolin 3 directly bound to nNOS and suppressed its phosphorylation-dependent activation at a specific residue, Ser1412 in the nicotinamide adenine dinucleotide phosphate (NADPH)-flavin adenine dinucleotide (FAD) module near the C-terminus of the reduction domain, in vitro. Constitutively active nNOS enhanced myoblast fusion, but not myogenesis, in vitro. Phosphorylation-dependent activation of nNOS occurred in muscles from caveolin 3-mutant mice and LGMD1C patients. Mating with nNOS-mutant mice exacerbated myofiber hypotrophy in the caveolin 3-mutant mice. In nNOS-mutant mice, regenerating myofibers after cardiotoxin injury became hypotrophic with reduced myoblast fusion. Administration of NO donor increased myofiber size and the number of myonuclei in the caveolin 3-mutant mice. Exercise also increased myofiber size accompanied by phosphorylation-dependent activation of nNOS in wild-type and caveolin 3-mutant mice. These data indicate that caveolin 3 inhibits phosphorylation-dependent activation of nNOS, which leads to myofiber hypertrophy via enhancing myoblast fusion. Hypertrophic signaling by nNOS phosphorylation could act in a compensatory manner in caveolin 3-deficient muscles.


Assuntos
Caveolina 3 , Flavina-Adenina Dinucleotídeo , Óxido Nítrico Sintase Tipo I , Animais , Cardiotoxinas , Caveolina 3/genética , Caveolina 3/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Camundongos , NADP/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Fosforilação , Sarcolema/metabolismo
6.
Intern Med ; 61(8): 1241-1245, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-34615823

RESUMO

Muscle phosphorylase b kinase (PHK) deficiency is a rare mild metabolic disorder caused by mutations of the PHKA1 gene encoding the αM subunit of PHK. A 16-year-old boy experienced myalgia during the maximal multistage 20-m shuttle run test targeting the maximal oxygen consumption. Although an ischemic forearm exercise test was normal, a muscle biopsy revealed subsarcolemmal glycogen accumulation. He harbored a novel insertion mutation in the PHKA1 gene that resulted in premature termination of the αM subunit close to the C-terminus. Compared with previously reported cases, his reduction in PHK activity was relatively mild.


Assuntos
Mialgia , Fosforilase Quinase , Adolescente , Doenças Genéticas Ligadas ao Cromossomo X , Doença de Depósito de Glicogênio , Humanos , Masculino , Músculos , Mialgia/etiologia , Fosforilase Quinase/genética , Fosforilase Quinase/metabolismo
7.
J Neurol Neurosurg Psychiatry ; 90(5): 529-536, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-29666206

RESUMO

OBJECTIVE: The aim of this study was to evaluate the efficacy and safety of high-dose taurine supplementation for prevention of stroke-like episodes of MELAS (mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes), a rare genetic disorder caused by point mutations in the mitochondrial DNA that lead to a taurine modification defect at the first anticodon nucleotide of mitochondrial tRNALeu(UUR), resulting in failure to decode codons accurately. METHODS: After the nationwide survey of MELAS, we conducted a multicentre, open-label, phase III trial in which 10 patients with recurrent stroke-like episodes received high-dose taurine (9 g or 12 g per day) for 52 weeks. The primary endpoint was the complete prevention of stroke-like episodes during the evaluation period. The taurine modification rate of mitochondrial tRNALeu(UUR) was measured before and after the trial. RESULTS: The proportion of patients who reached the primary endpoint (100% responder rate) was 60% (95% CI 26.2% to 87.8%). The 50% responder rate, that is, the number of patients achieving a 50% or greater reduction in frequency of stroke-like episodes, was 80% (95% CI 44.4% to 97.5%). Taurine reduced the annual relapse rate of stroke-like episodes from 2.22 to 0.72 (P=0.001). Five patients showed a significant increase in the taurine modification of mitochondrial tRNALeu(UUR) from peripheral blood leukocytes (P<0.05). No severe adverse events were associated with taurine. CONCLUSIONS: The current study demonstrates that oral taurine supplementation can effectively reduce the recurrence of stroke-like episodes and increase taurine modification in mitochondrial tRNALeu(UUR) in MELAS. TRIAL REGISTRATION NUMBER: UMIN000011908.


Assuntos
Suplementos Nutricionais , Síndrome MELAS/complicações , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/prevenção & controle , Taurina/uso terapêutico , Administração Oral , Adolescente , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
8.
Rejuvenation Res ; 22(1): 51-59, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29962294

RESUMO

OBJECTIVE: Pulp regeneration by stem cell transplantation declines due to age-related reduction. We hypothesized that administration of a cytokine together with the cell transplantation may improve the stem cell niche microenvironment and promote regeneration. CCL11 is implicated as a factor in aging. This investigation was performed to investigate the changes in the quality of the regenerated pulp by administration of CCL11 antibody in the aged mice and elucidate the underlying mechanisms. MATERIALS AND METHODS: Mobilized dental pulp stem cell (MDPSC) transplants were characterized in an ectopic tooth root transplantation model in both the aged and young mice. The amount of regenerated pulp tissue was analyzed in the transplants with continuous administration of CCL11 antibody compared with those without the antibody administration. Blood CCL11 levels were assessed at the onset of the experiment. Furthermore, immunostaining of CD68 together with CD11c or CD206 for M1 and M2 macrophage, respectively, were performed. Each double-positive cell count of M1 and M2 macrophages and M1/M2 ratio in the transplants with administration were compared with those without administration both in the aged and young mice. RESULTS: The administration of CCL11 antibody enhanced pulp regeneration and significantly reduced the blood CCL11 level in the aged mice. As the number of M1 macrophages decreased, the M1/M2 ratio in the treated aged mouse was less than that in the untreated aged mouse. There was, however, significant difference between the treated aged mouse and the untreated young mouse. CONCLUSION: CCL11 antibody has the potential to enhance and stimulate pulp regeneration in the aged mice.


Assuntos
Envelhecimento , Anticorpos Neutralizantes/administração & dosagem , Quimiocina CCL11/antagonistas & inibidores , Polpa Dentária/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Reimplante Dentário , Animais , Diferenciação Celular , Quimiocina CCL11/imunologia , Polpa Dentária/imunologia , Polpa Dentária/metabolismo , Camundongos , Camundongos SCID , Regeneração , Células-Tronco/imunologia , Células-Tronco/metabolismo , Suínos
9.
Int J Oncol ; 54(3): 1021-1032, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30569110

RESUMO

Dickkopf­related protein 3 (DKK3), which is a member of the Dickkopf WNT signaling pathway inhibitor family, is considered to be a tumor suppressor, due to its reduced expression in cancer cells and its ability to induce apoptosis when overexpressed by adenovirus. However, our previous study demonstrated alternative functions for DKK3 in head and neck squamous cell carcinoma (HNSCC). Our study reported that DKK3 expression was predominantly upregulated in HNSCC cell lines and tissue samples, and its expression was significantly correlated with poor prognosis. Furthermore, DKK3 overexpression in HNSCC cells significantly increased cancer cell proliferation, migration, invasion and in vivo tumor growth. These data have led to the hypothesis that DKK3 may exert oncogenic functions and may increase the malignant properties of HNSCC. The present study established a stable DKK3 knockdown cell line (HSC­3 shDKK3) using lentivirus­mediated short hairpin RNA, and assessed its effects on cancer cell behavior using MTT, migration and invasion assays. In addition, its effects on in vivo tumor growth were assessed using a xenograft model. Furthermore, the molecular mechanisms underlying the effects of DKK3 knockdown were investigated by microarray analysis, pathway analysis and western blotting. Compared with control cells, HSC­3 shDKK3 cells exhibited significantly reduced proliferation, migration and invasion, and formed significantly smaller tumor masses when subcutaneously transplanted into nude mice. In addition, in HSC­3 shDKK3 cells, the expression levels of phosphorylated (p)­protein kinase B (Akt) (Ser473), p­phosphoinositide 3­kinase (PI3K) p85 (Tyr467), p­PI3K p55 (Try199), p­3­phosphoinositide­dependent protein kinase­1 (PDK1) (Ser241) and total p38 mitogen­activated protein kinase (MAPK) were reduced. Furthermore, phosphorylation of mechanistic target of rapamycin (mTOR) (Ser2448) was slightly decreased in HSC­3 shDKK3 cells, which may be due to the increased expression of DEP domain­containing mTOR­interacting protein. Conversely, DKK3 overexpression in HSC­3 shDKK3 cells rescued cellular proliferation, migration and invasion. With regards to expression levels, p­PI3K and p­PDK1 expression was not altered, whereas mTOR and p­p38 MAPK expression was elevated. These data supported the hypothesis and indicated that DKK3 may contribute to the malignant phenotype of HNSCC cells via the PI3K/Akt/mTOR and MAPK signaling pathways.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Sistema de Sinalização das MAP Quinases , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Quimiocinas , Biologia Computacional , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Sistema de Sinalização das MAP Quinases/genética , Camundongos Nus , Análise em Microsséries , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/genética , Prognóstico , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Serina-Treonina Quinases TOR/metabolismo
10.
Oncol Res ; 26(1): 45-58, 2018 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-28470144

RESUMO

DKK3, a member of the dickkopf Wnt signaling pathway inhibitor family, is believed to be a tumor suppressor because of its reduced expression in cancer cells. However, our previous studies have revealed that DKK3 expression is predominantly observed in head and neck/oral squamous cell carcinoma (HNSCC/OSCC). Interestingly, HNSCC/OSCC patients with DKK3 expression showed a high rate of metastasis and poorer survival, and siRNA-mediated knockdown of DKK3 in HNSCC-derived cancer cell lines resulted in reduced cellular migration and invasion. From these data, it was hypothesized that DKK3 might exert an oncogenic function specific to HNSCC. In the present research, the DKK3 overexpression model was established, and its influences were investigated, together with molecular mechanism studies. The DKK3 expression profile in cancer cell lines was investigated, including HNSCC/OSCC, esophageal, gastric, colorectal, pancreatic, prostatic, and lung cancers. DKK3 overexpression was performed in HNSCC-derived cells by transfection of expression plasmid. The effects of DKK3 overexpression were assessed on cellular proliferation, migration, invasion, and in vivo tumor growth. The molecular mechanism of DKK3 overexpression was investigated by Western blotting and microarray analysis. DKK3 overexpression significantly elevated cellular proliferation, migration, and invasion, as well as increased mRNA expression of cyclin D1 and c-myc. However, reporter assays did not show TCF/LEF activation, suggesting that the increased malignant property of cancer cells was not driven by the Wnt/ß-catenin pathway. For the investigation of the pathways/molecules in DKK3-mediated signals, the Western blot analyses revealed that phosphorylation of Akt (S473) and c-Jun (Ser63) was elevated. The application of a PI3K kinase inhibitor, LY294002, on HSC-3 DKK3 cells significantly decreased tumor cell proliferation, migration, and invasion. From these results, we demonstrated that DKK3 might contribute to cellular proliferation, invasion, migration, and tumor cell survival in HNSCC cells through a mechanism other than the canonical Wnt signaling pathway, which might be attributed to PI3K-Akt signaling.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias de Cabeça e Pescoço/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Quimiocinas , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço , Regulação para Cima
11.
Biochem Biophys Res Commun ; 492(2): 199-205, 2017 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-28821434

RESUMO

BACKGROUND: The dystroglycan complex consists of two subunits: extracellular α-dystroglycan and membrane-spanning ß-dystroglycan, which provide a tight link between the extracellular matrix and the intracellular cytoskeleton. Previous studies showed that 43 kDa ß-dystroglycan is proteolytically cleaved into the 30 kDa fragment by matrix metalloproteinases (MMPs) in various non-muscle tissues, whereas it is protected from cleavage in muscles by the sarcoglycan complex which resides close to the dystroglycan complex. It is noteworthy that cleaved ß-dystroglycan is detected in muscles from patients with sarcoglycanopathy, sarcoglycan-deficient muscular dystrophy. In vitro assays using protease inhibitors suggest that both MMP-2 and MMP-9 contribute to the cleavage of ß-dystroglycan. However, this has remained uninvestigated in vivo. METHODS: We generated triple-knockout (TKO) mice targeting MMP-2, MMP-9 and γ-sarcoglycan to examine the status of ß-dystroglycan cleavage in the absence of the candidate matrix metalloproteinases in sarcoglycan-deficient muscles. RESULTS: Unexpectedly, ß-dystroglycan was cleaved in muscles from TKO mice. Muscle pathology was not ameliorated but worsened in TKO mice compared with γ-sarcoglycan single-knockout mice. The gene expression of MMP-14 was up-regulated in TKO mice as well as in γ-sarcoglycan knockout mice. In vitro assay showed MMP-14 is capable to cleave ß-dystroglycan. CONCLUSIONS: Double-targeting of MMP-2 and MMP-9 cannot prevent cleavage of ß-dystroglycan in sarcoglycanopathy. Thus, matrix metalloproteinases contributing to ß-dystroglycan cleavage are redundant, and MMP-14 could participate in the pathogenesis of sarcoglycanopathy.


Assuntos
Distroglicanas/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Músculo Esquelético/metabolismo , Sarcoglicanopatias/genética , Sarcoglicanas/genética , Animais , Deleção de Genes , Humanos , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Knockout , Músculo Esquelético/patologia , Proteólise , Sarcoglicanopatias/metabolismo , Sarcoglicanopatias/patologia , Sarcoglicanas/metabolismo , Regulação para Cima
12.
Sci Rep ; 7(1): 4486, 2017 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-28667270

RESUMO

Fetal cardiomyocytes actively proliferate to form the primitive heart in utero in mammals, but they stop dividing shortly after birth. The identification of essential molecules maintaining this active cardiomyocyte proliferation is indispensable for potential adult heart regeneration. A recent study has shown that this proliferation depends on a low fetal oxygen condition before the onset of breathing at birth. We have established an isolation protocol for mouse fetal cardiomyocytes, performed under strict low oxygen conditions to mimic the intrauterine environment, that gives the highest proliferative activities thus far reported. Oxygen exposure during isolation/culture markedly inhibited cell division and repressed cell cycle-promoting genes, and subsequent genome-wide analysis identified Fam64a as a novel regulatory molecule. Fam64a was abundantly expressed in hypoxic fetal cardiomyocyte nuclei, but this expression was drastically repressed by oxygen exposure, and in postnatal cardiomyocytes following the onset of breathing and the resulting elevation of oxygen tension. Fam64a knockdown inhibited and its overexpression enhanced cardiomyocyte proliferation. Expression of a non-degradable Fam64a mutant suggested that optimum Fam64a expression and subsequent degradation by anaphase-promoting complex/cyclosome (APC/C) during the metaphase-to-anaphase transition are required for fetal cardiomyocyte division. We propose that Fam64a is a novel cell cycle promoter of hypoxic fetal cardiomyocytes in mice.


Assuntos
Proteínas de Transporte/genética , Ciclo Celular/genética , Hipóxia/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Biomarcadores , Proteínas de Transporte/metabolismo , Divisão Celular/genética , Proliferação de Células , Células Cultivadas , Desenvolvimento Embrionário/genética , Feto , Expressão Gênica , Camundongos , Miócitos Cardíacos/citologia , Consumo de Oxigênio , Ligação Proteica
13.
Stem Cells ; 35(7): 1815-1834, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28480592

RESUMO

Recently, we found that resident myogenic stem satellite cells upregulate a multi-functional secreted protein, semaphorin 3A (Sema3A), exclusively at the early-differentiation phase in response to muscle injury; however, its physiological significance is still unknown. Here we show that Sema3A impacts slow-twitch fiber generation through a signaling pathway, cell-membrane receptor (neuropilin2-plexinA3) → myogenin-myocyte enhancer factor 2D → slow myosin heavy chain. This novel axis was found by small interfering RNA-transfection experiments in myoblast cultures, which also revealed an additional element that Sema3A-neuropilin1/plexinA1, A2 may enhance slow-fiber formation by activating signals that inhibit fast-myosin expression. Importantly, satellite cell-specific Sema3A conditional-knockout adult mice (Pax7CreERT2 -Sema3Afl °x activated by tamoxifen-i.p. injection) provided direct in vivo evidence for the Sema3A-driven program, by showing that slow-fiber generation and muscle endurance were diminished after repair from cardiotoxin-injury of gastrocnemius muscle. Overall, the findings highlight an active role for satellite cell-secreted Sema3A ligand as a key "commitment factor" for the slow-fiber population during muscle regeneration. Results extend our understanding of the myogenic stem-cell strategy that regulates fiber-type differentiation and is responsible for skeletal muscle contractility, energy metabolism, fatigue resistance, and its susceptibility to aging and disease. Stem Cells 2017;35:1815-1834.


Assuntos
Fibras Musculares de Contração Lenta/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Regeneração/genética , Células Satélites de Músculo Esquelético/metabolismo , Semaforina-3A/genética , Animais , Cardiotoxinas/administração & dosagem , Diferenciação Celular , Regulação da Expressão Gênica , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Musculares de Contração Lenta/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Miogenina/genética , Miogenina/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuropilina-2/genética , Neuropilina-2/metabolismo , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Regeneração/efeitos dos fármacos , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Semaforina-3A/antagonistas & inibidores , Semaforina-3A/metabolismo , Transdução de Sinais , Tamoxifeno/farmacologia
14.
Parasitol Int ; 66(2): 116-118, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28027968

RESUMO

Methods to maintain the life cycle of pathogenic organisms become powerful tools for studying molecular and cellular bases of infectious diseases. Spirometra erinaceieuropaei is a parasitic tapeworm that causes sparganosis in humans. Because S. erinaceieuropaei has a complex life cycle with different stages and host species requirements, there have been no reports to establish the complete life cycle in the laboratory. In this study, using Cyclops as the first intermediate host, mouse as the experimental second intermediate host, and dog as the final host, we succeeded in maintaining S. erinaceieuropaei in the laboratory. By repeating the established life cycle five times, we obtained a clonal population of S. erinaceieuropaei from a single adult worm. A karyotype study showed that the chromosome of this clone is triploid (3n=27), indicating that a genetically uniform strain is established by apomictic reproduction. The strain was named Kawasaki triploid (Kt). A partial sequence of mitochondrial cytochrome c oxidase subunit 1 gene of the strain Kt showed more than 98% similarity with those of S. erinaceieuropaei isolates from Australia, China, and South Korea, and the resultant phylogeny indicated that the strain Kt is a member of a distinctive clade from East Asia and Oceania. Our system will be particularly useful for studies of S. erinaceieuropaei infection and human sparganosis.


Assuntos
Estágios do Ciclo de Vida , Spirometra/crescimento & desenvolvimento , Spirometra/genética , Triploidia , Animais , Austrália , Infecções por Cestoides/parasitologia , China , DNA Mitocondrial , Cães , Ásia Oriental , Genes Mitocondriais , Variação Genética , Interações Hospedeiro-Parasita , Humanos , Cariótipo , Camundongos , Filogenia , República da Coreia , Esparganose/parasitologia
16.
PLoS One ; 10(7): e0133713, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26226340

RESUMO

Myostatin, a muscle-specific transforming growth factor-ß (TGF-ß), negatively regulates skeletal muscle mass. The N-terminal prodomain of myostatin noncovalently binds to and suppresses the C-terminal mature domain (ligand) as an inactive circulating complex. However, which region of the myostatin prodomain is required to inhibit the biological activity of myostatin has remained unknown. We identified a 29-amino acid region that inhibited myostatin-induced transcriptional activity by 79% compared with the full-length prodomain. This inhibitory core resides near the N-terminus of the prodomain and includes an α-helix that is evolutionarily conserved among other TGF-ß family members, but suppresses activation of myostatin and growth and differentiation factor 11 (GDF11) that share identical membrane receptors. Interestingly, the inhibitory core co-localized and co-immunoprecipitated with not only the ligand, but also its type I and type II membrane receptors. Deletion of the inhibitory core in the full-length prodomain removed all capacity for suppression of myostatin. A synthetic peptide corresponding to the inhibitory core (p29) ameliorates impaired myoblast differentiation induced by myostatin and GDF11, but not activin or TGF-ß1. Moreover, intramuscular injection of p29 alleviated muscle atrophy and decreased the absolute force in caveolin 3-deficient limb-girdle muscular dystrophy 1C model mice. The injection suppressed activation of myostatin signaling and restored the decreased numbers of muscle precursor cells caused by caveolin 3 deficiency. Our findings indicate a novel concept for this newly identified inhibitory core of the prodomain of myostatin: that it not only suppresses the ligand, but also prevents two distinct membrane receptors from binding to the ligand. This study provides a strong rationale for the use of p29 in the amelioration of skeletal muscle atrophy in various clinical settings.


Assuntos
Atrofia Muscular/metabolismo , Miostatina/metabolismo , Receptores de Superfície Celular/metabolismo , Ativinas/metabolismo , Animais , Células COS , Caveolina 3/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Estruturas da Membrana Celular/metabolismo , Chlorocebus aethiops , Fatores de Diferenciação de Crescimento/metabolismo , Células HEK293 , Humanos , Ligantes , Masculino , Camundongos , Músculo Esquelético/metabolismo , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Mioblastos/metabolismo , Estrutura Terciária de Proteína/fisiologia , Fator de Crescimento Transformador beta1/metabolismo
17.
BMC Cell Biol ; 16: 13, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25925429

RESUMO

BACKGROUND: Skeletal muscle differentiation is a multistep, complex pathway in which several important signaling molecules are involved. Recently, microRNAs (miRNAs), endogenous non-coding small RNAs that regulate mRNAs, have been proposed to be involved in skeletal muscle differentiation. In this study, we identified skeletal muscle differentiation-associated miRNAs by comparing miRNA expression profiles between C2C12 cells and Wnt4 over-expressing C2C12 cells (W4-08), which can spontaneously differentiate into myotubes. RESULTS: We identified miR-206, miR-133a, and miR-133b as up-regulated miRNAs and miR-487b, miR-3963 and miR-6412 as down-regulated miRNAs in differentiating cells. We focused on the down-regulated miRNAs because their functions were largely unknown. Transfection of mimics of these miRNAs into C2C12 cells resulted in significantly reduced expression of myogenic differentiation markers, including troponin T and myosin heavy chain fast type and slow type, but did not affect the expression of the myogenic transcription factors, MyoD and myogenin. CONCLUSIONS: These miRNAs were characterized as new myogenic differentiation-associated miRNAs which may delay late myogenic differentiation or maturation.


Assuntos
MicroRNAs/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Regulação para Baixo , Camundongos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Proteína MyoD/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Miogenina/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Oligonucleotídeos Antissenso/metabolismo , Transfecção , Troponina T/metabolismo , Regulação para Cima , Proteína Wnt4/genética , Proteína Wnt4/metabolismo
18.
J Med Chem ; 58(3): 1544-9, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25569186

RESUMO

Myostatin, an endogenous negative regulator of skeletal muscle mass, is a therapeutic target for muscle atrophic disorders. Here, we identified minimum peptides 2 and 7 to effectively inhibit myostatin activity, which consist of 24 and 23 amino acids, respectively, derived from mouse myostatin prodomain. These peptides, which had the propensity to form α-helix structure, interacted to myostatin with KD values of 30-36 nM. Moreover, peptide 2 significantly increased muscle mass in Duchenne muscular dystrophy model mice.


Assuntos
Músculo Esquelético/química , Miostatina/antagonistas & inibidores , Peptídeos/farmacologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Hep G2 , Humanos , Camundongos , Camundongos Endogâmicos mdx , Estrutura Molecular , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Peptídeos/administração & dosagem , Peptídeos/química , Relação Estrutura-Atividade
19.
Dev Growth Differ ; 56(9): 640-52, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25330272

RESUMO

Wnt proteins are thought to bind to their receptors on the cell surfaces of neighboring cells. Wnt8 likely substitutes for the dorsal determinants in Xenopus embryos to dorsalize early embryos via the Wnt/ß-catenin pathway. Here, we show that Wnt8 can dorsalize Xenopus embryos working cell autonomously. Wnt8 mRNA was injected into a cleavage-stage blastomere, and the subcellular distribution of Wnt8 protein was analyzed. Wnt8 protein was predominantly found in the endoplasmic reticulum (ER) and resided at the periphery of the cells; however, this protein was restricted to the mRNA-injected cellular region as shown by lineage tracing. A mutant Wnt8 that contained an ER retention signal (Wnt8-KDEL) could dorsalize Xenopus embryos. Finally, Wnt8-induced dorsalization occurred only in cells injected with Wnt8 mRNA. These experiments suggest that the Wnt8 protein acts within the cell, likely in the ER or on the cell surface in an autocrine manner for dorsalization.


Assuntos
Comunicação Autócrina/fisiologia , Embrião não Mamífero/metabolismo , Oócitos/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Embrião não Mamífero/citologia , Oócitos/citologia , Sinais Direcionadores de Proteínas/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Wnt/biossíntese , Proteínas Wnt/genética , Proteínas de Xenopus/biossíntese , Proteínas de Xenopus/genética , Xenopus laevis , beta Catenina/genética
20.
Int J Cell Biol ; 2013: 616294, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23864860

RESUMO

Background. Wnt signaling is involved in muscle formation through ß-catenin-dependent or -independent pathways, but interactions with other signaling pathways including transforming growth factor ß/Smad have not been precisely elucidated. Results. As Wnt4 stimulates myogenic differentiation by antagonizing myostatin (GDF8) activity, we examined the role of Wnt4 signaling during muscle differentiation in the C2C12 myoblast cell line. Among several extrinsic signaling molecules examined in a microarray analysis of C2C12 cells during the transition from cell proliferation to differentiation after mitogen deprivation, bone morphogenetic protein 4 (BMP4) expression was prominently increased. Wnt4 overexpression had similar effects on BMP4 expression. BMP4 was able to inhibit muscle differentiation when added to the culture medium. BMP4 and noggin had no effects on the cellular localization of ß-catenin induced by Wnt3a; however, the BMP4-induced phosphorylation of Smad1/5/8 was enhanced by Wnt4, but not by Wnt3a. The BMP antagonist noggin effectively stimulated muscle differentiation through binding to endogenous BMPs, and the effect of noggin was enhanced by the presence of Wnt3a and Wnt4. Conclusion. These results suggest that BMP/Smad pathways are modified through Wnt signaling during the transition from progenitor cell proliferation to myogenic differentiation, although Wnt/ß-catenin signaling is not modified with BMP/Smad signaling.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...