Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cell Endocrinol ; 576: 112008, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37422125

RESUMO

We established a methodology using machine learning algorithms for determining the pathogenic factors for premenstrual dysphoric disorder (PMDD). PMDD is a disease characterized by emotional and physical symptoms that occurs before menstruation in women of childbearing age. Owing to the diverse manifestations and various pathogenic factors associated with this disease, the diagnosis of PMDD is time-consuming and challenging. In the present study, we aimed to establish a methodology for diagnosing PMDD. Using an unsupervised machine-learning algorithm, we divided pseudopregnant rats into three clusters (C1 to C3), depending on the level of anxiety- and depression-like behaviors. From the results of RNA-seq and subsequent qPCR of the hippocampus in each cluster, we identified 17 key genes for building a PMDD diagnostic model using our original two-step feature selection with supervised machine learning. By inputting the expression levels of these 17 genes into the machine learning classifier, the PMDD symptoms of another group of rats were successfully classified as C1-C3 with an accuracy of 96%, corresponding to the classification by behavior. The present methodology would be applicable for the clinical diagnosis of PMDD using blood samples instead of samples from the hippocampus in the future.


Assuntos
Transtorno Disfórico Pré-Menstrual , Síndrome Pré-Menstrual , Humanos , Feminino , Animais , Ratos , Transtorno Disfórico Pré-Menstrual/diagnóstico , Transtorno Disfórico Pré-Menstrual/metabolismo , Transtorno Disfórico Pré-Menstrual/psicologia , Síndrome Pré-Menstrual/diagnóstico , Síndrome Pré-Menstrual/psicologia , Emoções , Aprendizado de Máquina , Algoritmos
2.
J Vet Med Sci ; 83(9): 1416-1424, 2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34334511

RESUMO

Rhabdomyosarcoma (RMS) is an aggressive type of soft tissue sarcoma, and pleomorphic RMS is a rare subtype of RMS found in adult. p16 is a tumor suppressor which inhibits cell cycle. In human RMS, p16 gene is frequently deleted, but p16-null mice do not develop RMS. We reported that genetic ablation of p16 by the crossbreeding of p16 knock-out rats (p16-KO rats) improved the dystrophic phenotype of a rat model of Duchenne muscular dystrophy (Dmd-KO rats). However, p16/Dmd double knock-out rats (dKO rats) unexpectedly developed sarcoma. In the present study, we raised p16-KO, Dmd-KO, and dKO rats until 11 months of age. Twelve out of 22 dKO rats developed pleomorphic RMS after 9 months of age, while none of p16-KO rats and Dmd-KO rats developed tumor. The neoplasms were connected to skeletal muscle tissue with indistinct borders and characterized by diffuse proliferation of pleomorphic cells which had eosinophilic cytoplasm and atypical nuclei with anisokaryosis. For almost all cases, the tumor cells immunohistochemically expressed myogenic markers including desmin, MyoD, and myogenin. The single cell cloning from tumor primary cells gained 20 individual Pax7-negative MyoD-positive RMS cell clones. Our results demonstrated that double knock-out of p16 and dystrophin in rats leads to the development of pleomorphic RMS, providing an animal model that may be useful to study the developmental mechanism of pleomorphic RMS.


Assuntos
Distrofia Muscular de Duchenne , Rabdomiossarcoma , Doenças dos Roedores , Sarcoma , Animais , Inibidor p16 de Quinase Dependente de Ciclina , Distrofina/genética , Camundongos , Músculo Esquelético , Ratos , Rabdomiossarcoma/genética
3.
Anim Sci J ; 92(1): e13573, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34231933

RESUMO

The degree of intramuscular adipose tissue accumulation is one of the factors affecting meat quality. Accumulation of adipocytes is also observed under the pathological condition of skeletal muscle such as muscular dystrophy and sarcopenia. The origin of adipocytes seen in skeletal muscle is mesenchymal progenitor cells that can give rise to both adipocytes and fibroblasts. In the present study, we demonstrated that siRNA-mediated suppression of MyoD expression in rat skeletal muscle progenitor cell culture, which comprises both myogenic satellite cells and mesenchymal progenitor cells, resulted in diminished myotube formation and an unexpected spontaneous appearance of white adipocytes. Suppressing myomaker expression also resulted in complete absence of myotube formation without reducing MyoD expression, but no adipogenesis was seen in this scenario, indicating that decline in MyoD expression rather than decreased myotube formation is necessary to induce adipogenesis. In addition, spontaneous adipogenesis induced by suppressing MyoD expression in culture was inhibited by the conditioned medium from control culture, indicating that anti-adipogenic factor(s) are secreted from MyoD-positive myogenic cells. These results indicate the presence of regulatory mechanism on adipogenesis by myogenic cells.


Assuntos
Adipogenia , Células Satélites de Músculo Esquelético , Adipogenia/genética , Animais , Fibras Musculares Esqueléticas , Músculo Esquelético , Ratos , Células-Tronco
4.
Sci Rep ; 10(1): 16385, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-33046751

RESUMO

Duchenne muscular dystrophy (DMD) is a progressive disease characterised by chronic muscle degeneration and inflammation. Our previously established DMD model rats (DMD rats) have a more severe disease phenotype than the broadly used mouse model. We aimed to investigate the role of senescence in DMD using DMD rats and patients. Senescence was induced in satellite cells and mesenchymal progenitor cells, owing to the increased expression of CDKN2A, p16- and p19-encoding gene. Genetic ablation of p16 in DMD rats dramatically restored body weight and muscle strength. Histological analysis showed a reduction of fibrotic and adipose tissues invading skeletal muscle, with increased muscle regeneration. Senolytic drug ABT263 prevented loss of body weight and muscle strength, and increased muscle regeneration in rats even at 8 months-the late stage of DMD. Moreover, senescence markers were highly expressed in the skeletal muscle of DMD patients. In situ hybridization of CDKN2A confirmed the expression of it in satellite cells and mesenchymal progenitor cells in patients with DMD. Collectively, these data provide new insights into the integral role of senescence in DMD progression.


Assuntos
Senescência Celular/genética , Modelos Animais de Doenças , Distrofina/genética , Distrofia Muscular de Duchenne/genética , Mutação , Animais , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Distrofina/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/metabolismo , Ratos , Regeneração/genética , Células Satélites de Músculo Esquelético/metabolismo
5.
Dis Model Mech ; 13(9)2020 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-32859695

RESUMO

Dystrophin, encoded by the DMD gene on the X chromosome, stabilizes the sarcolemma by linking the actin cytoskeleton with the dystrophin-glycoprotein complex (DGC). In-frame mutations in DMD cause a milder form of X-linked muscular dystrophy, called Becker muscular dystrophy (BMD), characterized by the reduced expression of truncated dystrophin. So far, no animal model with in-frame mutations in Dmd has been established. As a result, the effect of in-frame mutations on the dystrophin expression profile and disease progression of BMD remains unclear. In this study, we established a novel rat model carrying in-frame Dmd gene mutations (IF rats) and evaluated the pathology. We found that IF rats exhibited reduced expression of truncated dystrophin in a proteasome-independent manner. This abnormal dystrophin expression caused dystrophic changes in muscle tissues but did not lead to functional deficiency. We also found that the expression of additional dystrophin named dpX, which forms the DGC in the sarcolemma, was associated with the appearance of truncated dystrophin. In conclusion, the outcomes of this study contribute to the further understanding of BMD pathology and help elucidate the efficiency of dystrophin recovery treatments in Duchenne muscular dystrophy, a more severe form of X-linked muscular dystrophy.


Assuntos
Distrofina/genética , Distrofia Muscular de Duchenne/genética , Mutação/genética , Fases de Leitura Aberta/genética , Animais , Sequência de Bases , Membrana Celular/metabolismo , Modelos Animais de Doenças , Distroglicanas/metabolismo , Músculo Esquelético/patologia , Miocárdio/patologia , Fenótipo , Isoformas de Proteínas/metabolismo , Ratos , Sarcolema/metabolismo
6.
Anim Sci J ; 91(1): e13368, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32285501

RESUMO

Accumulation of intramuscular adipose tissue (IMAT) and development of fibrous tissues due to accumulation of collagen both affect meat quality such as tenderness, texture, and flavor. Thus, it is important for the production of high-quality meat to regulate the amount of adipose and fibrous tissues in skeletal muscle. IMAT is comprised of adipocytes, while collagens included in fibrous tissues are mainly produced by activated fibroblasts. Both adipocytes and fibroblasts are differentiated from their common ancestors, called mesenchymal progenitor cells (MPC). We previously established rat MPC clone, 2G11 cells. As several reports implicated the plasticity of fibroblast differentiation, in the present study, using 2G11 cells, we asked whether myofibroblasts differentiated from MPC are capable of re-gaining adipogenic potential in vitro. By treating with bFGF, their αSMA expression was reduced and adipogenic potential was restored partially. Furthermore, by lowering cell density together with bFGF treatment, 2G11 cell-derived myofibroblasts lost αSMA expression and showed the highest adipogenic potential, and this was along with their morphological change from flattened- to spindle-like shape, which is typically observed with MPC. These results indicated that MPC-derived myofibroblasts could re-acquire adipogenic potential, possibly mediated through returning to an undifferentiated MPC-like state.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/fisiologia , Músculo Esquelético/citologia , Miofibroblastos , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Colágeno/metabolismo , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/metabolismo , Miofibroblastos/metabolismo
7.
Neuroreport ; 31(6): 478-483, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32168097

RESUMO

Progranulin (PGRN) is a glycoprotein that is widely expressed among organs, including the central nervous system. PGRN insufficiency is involved in various neurodegenerative disorders such as frontotemporal dementia, Alzheimer's disease, and neuronal ceroid lipofuscinosis. One of the major causes of neuronal damage is hyperactivation of the cerebrum triggered by upregulation of excitatory systems. In the present study, we examined the possible involvement of PGRN in modulating excitability of the cerebrum using wild type and PGRN-deficient mice. First, we treated wild type and PGRN-deficient mice with seizure-inducible drugs, bicuculline or N-methyl-D-aspartate (NMDA), which provoke hyperexcitement of neurons. PGRN-deficient mice showed higher intensity of seizure and longer duration of convulsive behavior when treated with either bicuculline or NMDA. Next, we quantified the expression of NMDA receptor subunits in the hippocampus and cerebral cortex. The expression level of NR2A subunit protein was significantly higher in the hippocampus of PGRN-deficient mice, while no difference was observed in the cerebral cortex. On the other hand, mRNA levels of NMDA receptor subunits in the hippocampus were comparable or even lower in PGRN-deficient mice. These results suggest that PGRN modulates the excitability of the cerebrum by regulating at least partially the protein level of NMDA receptors in the hippocampus.


Assuntos
Bicuculina/efeitos adversos , Convulsivantes/efeitos adversos , N-Metilaspartato/efeitos adversos , Progranulinas/metabolismo , Convulsões/genética , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Progranulinas/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsões/induzido quimicamente , Convulsões/metabolismo
8.
J Vet Med Sci ; 81(11): 1649-1654, 2019 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-31582603

RESUMO

We have previously shown that secreted protein acidic and rich in cysteine (SPARC) promotes myogenic differentiation of rat skeletal muscle progenitor cells in vitro, and in vivo small interfering RNA (siRNA)-mediated transient suppression of SPARC expression in skeletal muscle of mice causes atrophic changes of myofibers, suggesting that SPARC plays a role in the maintenance of skeletal muscle function. In order to know the effect of long-term deficiency of SPARC on skeletal muscle, we performed phenotypic analyses of skeletal muscle of SPARC-null mice. Age-associated changes of myofiber diameters were comparable between wild type (WT) and SPARC-null mice at all ages examined, indicating that the growth of myofibers is unaffected by the absence of SPARC. On the other hand, accumulation of fibrillar collagen was significantly reduced in SPARC-null mice compared to WT mice after 5 months of age without significant changes of collagen I gene expression. The results obtained in the present study suggest that SPARC plays a role to maintain the stiffness of skeletal muscle by regulating collagen accumulation.


Assuntos
Colágenos Fibrilares/metabolismo , Músculo Esquelético/metabolismo , Osteonectina/metabolismo , Envelhecimento/metabolismo , Animais , Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Miofibrilas , Osteonectina/genética
9.
Aging (Albany NY) ; 10(4): 747-763, 2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29695641

RESUMO

Sarcopenia is the age-related loss of skeletal muscle mass and function. Skeletal muscle comprises diverse progenitor cells, including mesenchymal progenitor cells (MPCs), which normally support myogenic cell function but cause a decline in skeletal muscle function after differentiating into fibrous/adipose tissue. Cellular senescence is a form of persistent cell cycle arrest caused by cellular stress, including oxidative stress, and is accompanied by the acquisition of senescence-associated secretory phenotype (SASP). Here, we found γH2AX+ senescent cells appeared in the interstitium in skeletal muscle, corresponding in position to that of MPCs. H2O2 mediated oxidative stress in 2G11 cells, a rat MPC clone previously established in our laboratory, successfully induced senescence, as shown by the upregulation of p21 and SASP factors, including IL-6. The senescent 2G11 cells lost their fibro/adipogenic potential, but, intriguingly, coculture of myoblasts with senescent 2G11 cells abrogated the myotube formation, which coincided with the downregulation of myomaker, a muscle-specific protein involved in myogenic cell fusion; however, forced expression of myomaker could not rescue this abrogation. These results suggest that senescent MPCs in aged rat skeletal muscle lose their fibro/adipogenic potential, but differ completely from undifferentiated progenitor cells in that senescent MPCs suppress myoblast fusion and thereby potentially accelerate sarcopenia.


Assuntos
Diferenciação Celular/fisiologia , Senescência Celular/fisiologia , Células-Tronco Mesenquimais/patologia , Mioblastos/citologia , Estresse Oxidativo/fisiologia , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/fisiologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Fibroblastos/citologia , Fibroblastos/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Desenvolvimento Muscular/fisiologia , Mioblastos/metabolismo , Ratos , Ratos Wistar , Sarcopenia/metabolismo , Sarcopenia/patologia
10.
Cytokine ; 108: 17-23, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29558694

RESUMO

Skeletal muscle is an endocrine organ that secretes several proteins, which are collectively termed myokines. Although many studies suggest that exercise regulates myokine secretion, the underlying mechanisms remain unclear and all the exercise-dependent myokines have not yet been identified. Therefore, in this study, we attempted to identify novel exercise-dependent myokines by using our recently developed in vitro contractile model. Differentiated C2C12 myotubes were cultured with or without electrical pulse stimulation (EPS) for 24 h to induce cell contraction, and the myokines secreted in conditioned medium were analyzed using a cytokine array. Although most myokine secretions were not affected by EPS, the secretion of Chemokine (C-C motif) ligand 5 (CCL5) (regulated on activation, normal T cell expressed and secreted (RANTES)) was significantly reduced by EPS. This was further confirmed by ELISA and quantitative PCR. Contraction-dependent calcium transients and activation of 5'-AMP activating protein kinase (AMPK) appears to be involved in this decrease, as the chelating Ca2+ by EGTA blocked contraction-dependent CCL5 reduction, whereas the pharmacological activation of AMPK significantly reduced it. However, Ccl5 gene expression was increased by AMPK activation, suggesting that AMPK-dependent CCL5 decrease occurred via post-transcriptional regulation. Finally, mouse experiments revealed that voluntary wheel-running exercise reduced serum CCL5 levels and Ccl5 gene expression in the fast-twitch muscles. Overall, our study provides the first evidence of an exercise-reducible myokine, CCL5, in the mouse skeletal muscle. Although further studies are required to understand the precise roles of the skeletal muscle cell contraction-induced decrease in CCL5, this decrease may explain some exercise-dependent physiological changes such as those in immune responses.


Assuntos
Quimiocina CCL5/fisiologia , Contração Muscular , Músculo Esquelético/fisiologia , Condicionamento Físico Animal , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Células Cultivadas , Quimiocina CCL5/genética , Citocinas/genética , Citocinas/metabolismo , Estimulação Elétrica , Expressão Gênica , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real
11.
Biosci Biotechnol Biochem ; 82(1): 97-105, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29235416

RESUMO

Accumulating evidence indicates that skeletal muscle secrets proteins referred to as myokines and that exercise contributes to their regulation. In this study, we propose that chemokine (C-X-C motif) ligand 10 (CXCL10) functions as a novel myokine. Initially, we stimulated differentiated C2C12 myotubes with or without electrical pulse stimulation (EPS) to identify novel myokines. Cytokine array analysis revealed that CXCL10 secretion was significantly reduced by EPS, which was further confirmed by enzyme-linked immunosorbent assay and quantitative polymerase chain reaction analysis. Treadmill experiments in mice identified significant reduction of Cxcl10 gene expression in the soleus muscle. Additionally, contraction-dependent p38 MAPK activation appeared to be involved in this reduction. Furthermore, C2C12 conditioned medium obtained after applying EPS could induce survival of MSS31, a vascular endothelial cell model, which was partially attenuated by the addition of recombinant CXCL10. Overall, our findings suggest CXCL10 as a novel exercise-reducible myokine, to control endothelial cell viability.


Assuntos
Quimiocina CXCL10/fisiologia , Teste de Esforço , Fibras Musculares Esqueléticas/fisiologia , Indutores da Angiogênese , Animais , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Sistema de Sinalização das MAP Quinases , Camundongos , Contração Muscular , Condicionamento Físico Animal , Reação em Cadeia da Polimerase
12.
J Vet Med Sci ; 80(2): 346-353, 2018 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-29249750

RESUMO

Skeletal muscle has an ability to regenerate in response to injury due to the presence of satellite cells. Injury in skeletal muscle causes infiltration of pro-inflammatory macrophages (M1 macrophages) to remove necrotic myofibers, followed by their differentiation into anti-inflammatory macrophages (M2 macrophages) to terminate the inflammation. Since both M1 and M2 macrophages play important roles, coordinated regulation of their kinetics is important to complete muscle regeneration successfully. Progranulin (PGRN) is a pluripotent growth factor, having a protective role against the inflamed tissue. In the central nervous system, PGRN regulates inflammation by inhibiting the activation of microglia. Here we used muscle injury model of PGRN-knockout (PGRN-KO) mice to elucidate whether it has a role in the kinetics of macrophages during muscle regeneration. We found the prolonged persistence of macrophages at the late phase of regeneration in PGRN-KO mice, and these macrophages were suggested to be M2 macrophages since this was accompanied with an increased CD206 expression. We also observed muscle hypertrophy in PGRN-KO mice at the late stage of muscle regeneration. Since M2 macrophages are known to have a role in maturation of myofibers, this muscle hypertrophy may be due to the presence of increased number of M2 macrophages. Our results suggest that PGRN plays a role in the regulation of kinetics of macrophages for the systemic progress of muscle regeneration.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Macrófagos/fisiologia , Músculo Esquelético/fisiologia , Regeneração , Animais , Proteínas Cardiotóxicas de Elapídeos/farmacologia , Feminino , Granulinas , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Musculares Esqueléticas/fisiologia , Progranulinas
13.
Sci Rep ; 7(1): 16812, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29196732

RESUMO

Autism spectrum disorders (ASD) are more common among boys than girls. The mechanisms responsible for ASD symptoms and their sex differences remain mostly unclear. We previously identified collapsin response mediator protein 4 (CRMP4) as a protein exhibiting sex-different expression during sexual differentiation of the hypothalamic sexually dimorphic nucleus. This study investigated the relationship between the sex-different development of autistic features and CRMP4 deficiency. Whole-exome sequencing detected a de novo variant (S541Y) of CRMP4 in a male ASD patient. The expression of mutated mouse CRMP4 S540Y, which is homologous to human CRMP4 S541Y, in cultured hippocampal neurons derived from Crmp4-knockout (KO) mice had increased dendritic branching, compared to those transfected with wild-type (WT) Crmp4, indicating that this mutation results in altered CRMP4 function in neurons. Crmp4-KO mice showed decreased social interaction and several alterations of sensory responses. Most of these changes were more severe in male Crmp4-KO mice than in females. The mRNA expression levels of some genes related to neurotransmission and cell adhesion were altered in the brain of Crmp4-KO mice, mostly in a gender-dependent manner. These results indicate a functional link between a case-specific, rare variant of one gene, Crmp4, and several characteristics of ASD, including sexual differences.


Assuntos
Transtorno do Espectro Autista/genética , Hipocampo/citologia , Proteínas Musculares/deficiência , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Estudos de Associação Genética , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Camundongos , Proteínas Musculares/genética , Neurônios/citologia , Neurônios/metabolismo , Neurônios/patologia , Caracteres Sexuais
14.
Exp Gerontol ; 95: 1-8, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28479389

RESUMO

It is well established that adult neurogenesis in the hippocampus declines with age. Our previous studies have suggested that progranulin (PGRN) has a facilitative effect on hippocampal neurogenesis. We have also shown that PGRN plays a role in suppressing excessive neuroinflammatory responses in the cortex and thalamus after brain injury and aging, respectively. However, the roles of PGRN in modulating neurogenesis and neuroinflammatory responses in the hippocampus of aged animals are not yet understood. In the present study, we investigated neurogenesis and neuroinflammation-related responses in the hippocampus of young (15-week-old) and old (135-week-old) wild-type and PGRN-deficient male mice. Neurogenesis in the dentate gyrus of the hippocampus markedly declined with age, and there was no significant difference between the genotype. The number of CD68-positive activated microglia and the expression of lysosomal genes in the hippocampus were significantly increased with age, and PGRN deficiency further increased them. The expression of pro-inflammatory genes was also increased with age, and PGRN deficiency significantly enhanced some of them. These results suggest that PGRN deficiency exacerbates neuroinflammatory responses related to activated microglia in aged animals, while PGRN may not counteract the decline of hippocampal neurogenesis with age.


Assuntos
Envelhecimento/metabolismo , Encefalite/metabolismo , Hipocampo/metabolismo , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neurogênese , Fatores Etários , Envelhecimento/genética , Envelhecimento/patologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Giro Denteado/metabolismo , Giro Denteado/patologia , Encefalite/genética , Encefalite/patologia , Granulinas , Hipocampo/patologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Lisossomos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , Progranulinas
15.
FEBS Open Bio ; 7(2): 149-159, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28174682

RESUMO

Progranulin (PGRN) is a growth factor implicated in several neurodegenerative diseases, such as frontotemporal lobar degeneration. Despite its important role in the central nervous system (CNS), the mechanisms controlling PGRN expression in the CNS are largely unknown. Recent evidence, however, suggested that several stressors, such as hypoxia, acidosis, or oxidative stress, induce PGRN expression. The present study was mainly aimed at determining whether and, if so, how glucose deprivation affects PGRN expression in PC12 cells. Initially, it was found that glucose deprivation gradually induced PGRN gene expression in PC12 cells. To elucidate the underlying molecular mechanisms, several intracellular signalings that were modified in response to glucose deprivation were examined. Both adenosine monophosphate kinase (AMPK) activation and changes in osmotic pressure, which are modified by extracellular glucose concentration, had no effect on PGRN gene expression; on the other hand, p38 activation in response to glucose deprivation played an important role in inducing PGRN gene expression. It was also found that expression of sortilin, a PGRN receptor implicated in PGRN endocytosis, was dramatically reduced by glucose deprivation. In contrast to glucose-dependent regulation of PGRN gene expression, AMPK activation played a central role in reducing sortilin expression. Overall, the present study suggests that the PGRN-sortilin axis is modulated by glucose deprivation via two distinct mechanisms. As PGRN is neuroprotective, this system may represent a new neuroprotective mechanism activated by glucose deprivation in the CNS.

16.
Hum Mol Genet ; 26(5): 969-988, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28073925

RESUMO

Progranulin (PGRN) haploinsufficiency resulting from loss-of-function mutations in the PGRN gene causes frontotemporal lobar degeneration accompanied by TDP-43 accumulation, and patients with homozygous mutations in the PGRN gene present with neuronal ceroid lipofuscinosis. Although it remains unknown why PGRN deficiency causes neurodegenerative diseases, there is increasing evidence that PGRN is implicated in lysosomal functions. Here, we show PGRN is a secretory lysosomal protein that regulates lysosomal function and biogenesis by controlling the acidification of lysosomes. PGRN gene expression and protein levels increased concomitantly with the increase of lysosomal biogenesis induced by lysosome alkalizers or serum starvation. Down-regulation or insufficiency of PGRN led to the increased lysosomal gene expression and protein levels, while PGRN overexpression led to the decreased lysosomal gene expression and protein levels. In particular, the level of mature cathepsin D (CTSDmat) dramatically changed depending upon PGRN levels. The acidification of lysosomes was facilitated in cells transfected with PGRN. Then, this caused degradation of CTSDmat by cathepsin B. Secreted PGRN is incorporated into cells via sortilin or cation-independent mannose 6-phosphate receptor, and facilitated the acidification of lysosomes and degradation of CTSDmat. Moreover, the change of PGRN levels led to a cell-type-specific increase of insoluble TDP-43. In the brain tissue of FTLD-TDP patients with PGRN deficiency, CTSD and phosphorylated TDP-43 accumulated in neurons. Our study provides new insights into the physiological function of PGRN and the role of PGRN insufficiency in the pathogenesis of neurodegenerative diseases.


Assuntos
Catepsina D/genética , Proteínas de Ligação a DNA/genética , Degeneração Lobar Frontotemporal/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Neurônios/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Catepsina D/metabolismo , Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/metabolismo , Degeneração Lobar Frontotemporal/patologia , Regulação da Expressão Gênica , Haploinsuficiência/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Lisossomos/genética , Lisossomos/patologia , Camundongos , Mutação , Neuroblastoma/metabolismo , Neurônios/patologia , Cultura Primária de Células , Progranulinas , Proteínas/genética
17.
Mol Neurobiol ; 54(3): 1953-1966, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-26910812

RESUMO

Stress and glucocorticoids suppress adult neurogenesis in the hippocampus. However, the molecular mechanisms underlying stress-induced impairment of adult neurogenesis are poorly understood. We previously suggested that cyclooxygenase (COX)-2 is a common mediator of stresses in the brain. Here, using a lipopolysaccharide (LPS)-induced acute infectious stress model, we evaluated the roles of COX-2 and its major downstream product prostaglandin E2 (PGE2) in adult neurogenesis and the influence of glucocorticoids on COX-2-related signaling. Treatment of rats with LPS significantly decreased neurogenesis in the dentate gyrus (DG) of the hippocampus, and this inhibitory effect of LPS on neurogenesis was reversed by the glucocorticoid receptor antagonist RU486. Moreover, RU486 significantly enhanced the increase in messenger RNA (mRNA) levels of COX-2 and microsomal prostaglandin E synthase (mPGES)-1 in the hippocampus following LPS stimulation. Administration of AH6809, a selective antagonist of the PGE2 EP2 receptor, as well as NS398, a COX-2 selective inhibitor, exacerbated the suppression of proliferation of neural progenitor cells (NPCs) in the DG. Gene expression of EP1, EP2, and EP3, but not EP4, receptors was also increased following LPS stimulation. Immunohistochemical studies indicated that NPCs expressed EP2 receptor, whereas the majority of cells expressing COX-2 and mPGES-1 were mature neurons in the DG. These results suggest that acute infectious stress upregulates COX-2-related signaling in neurons in the DG, which plays a protective role in neurogenesis through EP2 receptor at least partially. In addition, LPS-induced glucocorticoids suppress this COX-2-related signaling, resulting in decreased neurogenesis.


Assuntos
Doenças Transmissíveis/metabolismo , Ciclo-Oxigenase 2/biossíntese , Glucocorticoides/metabolismo , Hipocampo/metabolismo , Imunidade Celular/fisiologia , Neurogênese/fisiologia , Doença Aguda , Animais , Doenças Transmissíveis/induzido quimicamente , Doenças Transmissíveis/imunologia , Glucocorticoides/antagonistas & inibidores , Glucocorticoides/imunologia , Hipocampo/efeitos dos fármacos , Hipocampo/imunologia , Imunidade Celular/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Masculino , Mifepristona/farmacologia , Neurogênese/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/imunologia , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Xantonas/farmacologia
18.
Mol Neurobiol ; 54(5): 3717-3728, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27215202

RESUMO

Immune stress is well known to suppress adult neurogenesis in the hippocampus. We have demonstrated that progranulin (PGRN) has a mitogenic effect on neurogenesis under several experimental conditions. We have also shown that PGRN suppresses excessive neuroinflammatory responses after traumatic brain injury. However, the role of PGRN in modulating neurogenesis under acute immune stress is yet to be elucidated. In the present study, we evaluated the involvement of PGRN in neurogenesis and inflammatory responses in the hippocampus using a lipopolysaccharide (LPS)-induced immune stress model. Treatment of mice with LPS significantly increased the expression of PGRN in activated microglia and decreased neurogenesis in the dentate gyrus of the hippocampus. PGRN deficiency increased CD68-immunoreactive area and exacerbated suppression of neurogenesis following LPS treatment. The expression levels of lysosomal genes including lysozyme M, macrophage expressed gene 1, and cathepsin Z were higher in PGRN-deficient than in wild-type mice, while PGRN deficiency decreased mammalian target of rapamycin (mTOR) mRNA levels, suggesting that PGRN suppresses excessive lysosomal biogenesis by promoting mTOR signaling. LPS treatment also increased the expression of proinflammatory genes such as interleukin (IL)-1ß, tumor necrosis factor-α, and microsomal prostaglandin E synthase-1 (mPGES-1) in the hippocampus, and PGRN deficiency further enhanced gene expression of IL-6 and mPGES-1. These results suggest that PGRN plays a protecting role in hippocampal neurogenesis at least partially by attenuating neuroinflammatory responses during LPS-induced acute immune stress.


Assuntos
Hipocampo/patologia , Inflamação/imunologia , Neurogênese , Estresse Fisiológico/imunologia , Animais , Granulinas , Hipocampo/metabolismo , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lipopolissacarídeos , Lisossomos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Progranulinas , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
19.
Exp Cell Res ; 347(2): 367-77, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27582000

RESUMO

Intramuscular adipose tissue and fibrous tissue are observed in some skeletal muscle pathologies such as Duchenne muscular dystrophy and sarcopenia, and affect muscle strength and myogenesis. They originate from common fibrogenic/adipogenic cells in the skeletal muscle. Thus, elucidating the regulatory mechanisms underlying fibrogenic/adipogenic cell differentiation is an important step toward the mediation of these disorders. Previously, we established a highly adipogenic progenitor clone, 2G11, from rat skeletal muscle and showed that basic fibroblast growth factor (bFGF) is pro-adipogenic in these cells. Here, we demonstrated that 2G11 cells give rise to fibroblasts upon transforming growth factor (TGF)-ß1 stimulation, indicating that they possess mesenchymal progenitor cells (MPC)-like characteristics. The previously reported MPC marker PDGFRα is expressed in other cell populations. Accordingly, we produced monoclonal antibodies that specifically bind to 2G11 cell surface antigens and identified chondroitin sulfate proteoglycan 4 (CSPG4) as a potential MPC marker. Based on an RNA interference analysis, we found that CSPG4 is involved in both the pro-adipogenic effect of bFGF and in TGF-ß-induced alpha smooth muscle actin expression and stress fiber formation. By establishing an additional marker for MPC detection and characterizing its role in fibrogenic/adipogenic differentiation, these results will facilitate the development of effective treatments for skeletal muscle pathologies.


Assuntos
Adipogenia , Antígenos/metabolismo , Diferenciação Celular , Fibroblastos/citologia , Músculo Esquelético/citologia , Proteoglicanas/metabolismo , Adipogenia/efeitos dos fármacos , Animais , Anticorpos Monoclonais/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/efeitos dos fármacos , Camundongos Endogâmicos BALB C , RNA Interferente Pequeno/metabolismo , Ratos , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...