Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
FASEB J ; 37(1): e22680, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36468710

RESUMO

Spermatid production is a complex regulatory process in which coordination between hormonal control and apoptosis plays a pivotal role in maintaining a balanced number of sperm cells. Apoptosis in spermatogenesis is controlled by pro-apoptotic and anti-apoptotic molecules. Hormones involved in the apoptotic process during spermatogenesis include gonadotrophins, sex hormones, and glucocorticoid (GC). GC acts broadly as an apoptosis inducer by binding to its receptor (glucocorticoid receptor: GR) during organ development processes, such as spermatogenesis. However, the downstream pathway induced in GC-GR signaling and the apoptotic process during spermatogenesis remains poorly understood. We reported previously that GC induces full-length glucocorticoid-induced transcript 1 (GLCCI1-long), which functions as an anti-apoptotic mediator in thymic T cell development. Here, we demonstrate that mature murine testis expresses a novel isoform of GLCCI1 protein (GLCCI1-short) in addition to GLCCI1-long. We demonstrate that GLCCI1-long is expressed in spermatocytes along with GR. In contrast, GLCCI1-short is primarily expressed in spermatids where GR is absent; instead, the estrogen receptor is expressed. GLCCI1-short also binds to LC8, which is a known mediator of the anti-apoptotic effect of GLCCI1-long. A luciferase reporter assay revealed that ß-estradiol treatment synergistically increased Glcci1-short promotor-driven luciferase activity in Erα-overexpressing cells. Together with the evidence that the conversion of testosterone to estrogen is preceded by aromatase expression in spermatids, we hypothesize that estrogen induces GLCCI1-short, which, in turn, may function as a novel anti-apoptotic mediator in mature murine testis.


Assuntos
Glucocorticoides , Sêmen , Masculino , Camundongos , Animais , Espermatogênese , Espermátides , Estrogênios
2.
J Am Soc Nephrol ; 33(11): 2008-2025, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35985815

RESUMO

BACKGROUND: The cause of podocyte injury in idiopathic nephrotic syndrome (INS) remains unknown. Although recent evidence points to the role of B cells and autoimmunity, the lack of animal models mediated by autoimmunity limits further research. We aimed to establish a mouse model mimicking human INS by immunizing mice with Crb2, a transmembrane protein expressed at the podocyte foot process. METHODS: C3H/HeN mice were immunized with the recombinant extracellular domain of mouse Crb2. Serum anti-Crb2 antibody, urine protein-to-creatinine ratio, and kidney histology were studied. For signaling studies, a Crb2-expressing mouse podocyte line was incubated with anti-Crb2 antibody. RESULTS: Serum anti-Crb2 autoantibodies and significant proteinuria were detected 4 weeks after the first immunization. The proteinuria reached nephrotic range at 9-13 weeks and persisted up to 29 weeks. Initial kidney histology resembled minimal change disease in humans, and immunofluorescence staining showed delicate punctate IgG staining in the glomerulus, which colocalized with Crb2 at the podocyte foot process. A subset of mice developed features resembling FSGS after 18 weeks. In glomeruli of immunized mice and in Crb2-expressing podocytes incubated with anti-Crb2 antibody, phosphorylation of ezrin, which connects Crb2 to the cytoskeleton, increased, accompanied by altered Crb2 localization and actin distribution. CONCLUSION: The results highlight the causative role of anti-Crb2 autoantibody in podocyte injury in mice. Crb2 immunization could be a useful model to study the immunologic pathogenesis of human INS, and may support the role of autoimmunity against podocyte proteins in INS.


Assuntos
Nefrose Lipoide , Síndrome Nefrótica , Podócitos , Camundongos , Humanos , Animais , Podócitos/metabolismo , Síndrome Nefrótica/metabolismo , Nefrose Lipoide/patologia , Camundongos Endogâmicos C3H , Proteinúria/metabolismo , Modelos Animais de Doenças , Imunização , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo
3.
Biochem Biophys Res Commun ; 614: 198-206, 2022 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-35605301

RESUMO

Podocyte damage is a major pathological lesion leading to focal segmental glomerulosclerosis (FSGS). Podocytes damaged by cellular stress undergo hypertrophy to compensate for podocytopenia. It is known that cyclin-dependent kinase inhibitors induced by p53 ensure podocytes hypertrophy; however, its precise mechanism remains to be further investigated. In this study, we found that ubiquitin specific protease 40 (USP40) is a novel regulator of p53. Although USP40 knockout mice established in the present study revealed no abnormal kidney phenotype, intermediate filament Nestin was upregulated in the glomeruli, and was bound to and colocalized with USP40. We also found that USP40 deubiquitinated histidine triad nucleotide-binding protein 1 (HINT1), an inducer of p53. Gene knockdown experiments of USP40 in cultured podocytes revealed the reduction of HINT1 and p53 protein expression. Finally, in glomerular podocytes of mouse FSGS, upregulation of HINT1 occurred in advance of the proteinuria, which was followed by upregulation of USP40, p53 and Nestin. In conclusion, USP40 bound to Nestin deubiquitinates HINT1, and in consequence upregulates p53. These results provide additional insight into the pathological mechanism of podocyte hypertrophy in FSGS.


Assuntos
Glomerulosclerose Segmentar e Focal , Proteínas do Tecido Nervoso , Nestina , Podócitos , Proteína Supressora de Tumor p53 , Proteases Específicas de Ubiquitina , Animais , Enzimas Desubiquitinantes/genética , Enzimas Desubiquitinantes/metabolismo , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Hipertrofia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina/genética , Nestina/metabolismo , Podócitos/metabolismo , Podócitos/patologia , Podócitos/fisiologia , Proteína Quinase C/antagonistas & inibidores , Estresse Fisiológico/genética , Estresse Fisiológico/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteases Específicas de Ubiquitina/genética , Proteases Específicas de Ubiquitina/metabolismo , Ubiquitinação , Regulação para Cima
4.
FASEB J ; 33(6): 7387-7402, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30860871

RESUMO

Glucocorticoids (GCs) potently induce T-cell apoptosis in a GC receptor (GR)-dependent manner and are used to control lymphocyte function in clinical practice. However, its downstream pathways remain controversial. Here, we showed that GC-induced transcript 1 (GLCCI1) is a novel downstream molecule of the GC-GR cascade that acts as an antiapoptotic mediator in thymic T cells. GLCCI1 was highly phosphorylated and colocalized with microtubules in GLCCI1-transfected human embryonic kidney QBI293A cells. GR-dependent up-regulation of GLCCI1 was associated with GC-induced proapoptotic events in a cultured thymocyte cell line. However, GLCCI1 knockdown in a thymocyte cell line led to apoptosis. Consistently, transgenic mice overexpressing human GLCCI1 displayed enlarged thymi that consisted of larger numbers of thymocytes. Further molecular characterization showed that GLCCI1 bound to both dynein light chain LC8-type 1 (LC8) and its functional kinase, p21-protein activated kinase 1 (PAK1), thereby inhibiting the kinase activity of PAK1 toward LC8 phosphorylation, a crucial event in apoptotic signaling. GLCCI1 induction facilitated LC8 dimer formation and reduced Bim expression. Thus, GLCCI1 is a candidate factor involved in apoptosis regulation of thymic T cells.-Kiuchi, Z., Nishibori, Y., Kutsuna, S., Kotani, M., Hada, I., Kimura, T., Fukutomi, T., Fukuhara, D., Ito-Nitta, N., Kudo, A., Takata, T., Ishigaki, Y., Tomosugi, N., Tanaka, H., Matsushima, S., Ogasawara, S., Hirayama, Y., Takematsu, H., Yan, K. GLCCI1 is a novel protector against glucocorticoid-induced apoptosis in T cells.


Assuntos
Apoptose/fisiologia , Glucocorticoides/fisiologia , Receptores de Glucocorticoides/fisiologia , Linfócitos T/citologia , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2/biossíntese , Proteína 11 Semelhante a Bcl-2/genética , Linhagem Celular , Dineínas do Citoplasma/metabolismo , Dimerização , Regulação para Baixo , Técnicas de Silenciamento de Genes , Glucocorticoides/farmacologia , Humanos , Hipertrofia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microtúbulos/metabolismo , Fosforilação , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Receptores de Glucocorticoides/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais/fisiologia , Timo/patologia , Quinases Ativadas por p21/metabolismo
5.
Am J Physiol Renal Physiol ; 312(6): F1184-F1199, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28228401

RESUMO

Altered expression of nephrin underlies the pathophysiology of proteinuria in both congenital and acquired nephrotic syndrome. However, the epigenetic mechanisms of nephrin gene regulation remain elusive. Here, we show that Wolf-Hirschhorn syndrome candidate 1-like 1 long form (WHSC1L1-L) is a novel epigenetic modifier of nephrin gene regulation. WHSC1L1-L was associated with histone H3K4 and H3K36 in human embryonic kidney cells. WHSC1L1-L gene was expressed in the podocytes, and functional protein product was detected in these cells. WHSC1L1-L was found to bind nephrin but not other podocyte-specific gene promoters, leading to its inhibition/suppression, abrogating the stimulatory effect of WT1 and NF-κB. Gene knockdown of WHSC1L1-L in primary cultured podocytes accelerated the transcription of nephrin but not CD2AP. An in vivo zebrafish study involving the injection of Whsc1l1 mRNA into embryos demonstrated an apparent reduction of nephrin mRNA but not podocin and CD2AP mRNA. Immunohistochemistry showed that both WHSC1L1-L and nephrin emerged at the S-shaped body stage in glomeruli. Immunofluorescence and confocal microscopy displayed WHSC1L1 to colocalize with trimethylated H3K4 in the glomerular podocytes. Chromatin immunoprecipitation assay revealed the reduction of the association of trimethylated H3K4 at the nephrin promoter regions. Finally, nephrin mRNA was upregulated in the glomerulus at the early proteinuric stage of mouse nephrosis, which was associated with the reduction of WHSC1L1. In conclusion, our results demonstrate that WHSC1L1-L acts as a histone methyltransferase in podocytes and regulates nephrin gene expression, which may in turn contribute to the integrity of the slit diaphragm of the glomerular filtration barrier.


Assuntos
Epigênese Genética , Histona-Lisina N-Metiltransferase/genética , Proteínas de Membrana/genética , Síndrome Nefrótica/genética , Proteínas Nucleares/genética , Podócitos/enzimologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Sítios de Ligação , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Doxorrubicina , Regulação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Células HEK293 , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Metilação , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Síndrome Nefrótica/induzido quimicamente , Síndrome Nefrótica/enzimologia , Síndrome Nefrótica/patologia , Proteínas Nucleares/metabolismo , Podócitos/patologia , Regiões Promotoras Genéticas , Interferência de RNA , Transfecção , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
6.
Am J Physiol Renal Physiol ; 312(4): F702-F715, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28148530

RESUMO

Unbiased transcriptome profiling and functional genomics approaches have identified ubiquitin-specific protease 40 (USP40) as a highly specific glomerular transcript. This gene product remains uncharacterized, and its biological function is completely unknown. Here, we showed that mouse and rat glomeruli exhibit specific expression of the USP40 protein, which migrated at 150 kDa and was exclusively localized in the podocyte cytoplasm of the adult kidney. Double-labeling immunofluorescence staining and confocal microscopy analysis of fetal and neonate kidney samples revealed that USP40 was also expressed in the vasculature, including in glomerular endothelial cells at the premature stage. USP40 in cultured glomerular endothelial cells and podocytes was specifically localized to the intermediate filament protein nestin. In glomerular endothelial cells, immunoprecipitation confirmed actual protein-protein binding of USP40 with nestin, and USP40-small-interfering RNA transfection revealed significant reduction of nestin. In a rat model of minimal-change nephrotic syndrome, USP40 expression was apparently reduced, which was also associated with the reduction of nestin. Zebrafish morphants lacking Usp40 exhibited disorganized glomeruli with the reduction of the cell junction in the endothelium and foot process effacement in the podocytes. Permeability studies in these zebrafish morphants demonstrated a disruption of the selective glomerular permeability filter. These data indicate that USP40/Usp40 is a novel protein that might play a crucial role in glomerulogenesis and the glomerular integrity after birth through the modulation of intermediate filament protein homeostasis.


Assuntos
Taxa de Filtração Glomerular , Glomérulos Renais/enzimologia , Ubiquitina Tiolesterase/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Regulação para Baixo , Células Endoteliais/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genótipo , Células HEK293 , Humanos , Glomérulos Renais/embriologia , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Camundongos , Nefrose Lipoide/enzimologia , Nefrose Lipoide/genética , Nefrose Lipoide/fisiopatologia , Nestina/metabolismo , Permeabilidade , Fenótipo , Podócitos/enzimologia , Interferência de RNA , Ratos , Transfecção , Ubiquitina Tiolesterase/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
7.
J Am Soc Nephrol ; 22(11): 2037-46, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21949092

RESUMO

Unbiased transcriptome profiling and functional genomics approaches identified glucocorticoid-induced transcript 1 (GLCCI1) as being a transcript highly specific for the glomerulus, but its role in glomerular development and disease is unknown. Here, we report that mouse glomeruli express far greater amounts of Glcci1 protein compared with the rest of the kidney. RT-PCR and Western blotting demonstrated that mouse glomerular Glcci1 is approximately 60 kD and localizes to the cytoplasm of podocytes in mature glomeruli. In the fetal kidney, intense Glcci1 expression occurs at the capillary-loop stage of glomerular development. Using gene knockdown in zebrafish with morpholinos, morphants lacking Glcci1 function had collapsed glomeruli with foot-process effacement. Permeability studies of the glomerular filtration barrier in these zebrafish morphants demonstrated a disruption of the selective glomerular permeability filter. Taken together, these data suggest that Glcci1 promotes the normal development and maintenance of podocyte structure and function.


Assuntos
Glomérulos Renais/fisiopatologia , Podócitos/fisiologia , Pronefro/fisiopatologia , Proteinúria/metabolismo , Proteinúria/fisiopatologia , Receptores de Glucocorticoides/deficiência , Animais , Citoplasma/metabolismo , Dexametasona/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glucocorticoides/farmacologia , Glomérulos Renais/anormalidades , Masculino , Camundongos , Camundongos Endogâmicos ICR , Oligonucleotídeos Antissenso/farmacologia , Pronefro/anormalidades , Proteinúria/patologia , Coelhos , Receptores de Glucocorticoides/genética , Peixe-Zebra , Proteínas de Peixe-Zebra
8.
Lab Invest ; 91(7): 992-1006, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21403644

RESUMO

Molecular mechanisms and signaling pathways leading to cellular proliferation and lesion formation in the crescentic glomerulonephritis (CGN) remain elusive. In the present study we have explored a potential role of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway and amino acid transporter (LAT) in the pathogenesis of CGN. Immunohistochemistry and western blot analysis of glomeruli isolated from a rat model of CGN revealed that activation of mTORC1 preceded crescent formation in glomerular parietal epithelial cells (PECs) and podocytes. Daily treatment of rats with the mTOR inhibitor everolimus just after induction of CGN was not beneficial and instead led to increased cellular necrosis of PECs. However, daily treatment starting 7 days after the onset of CGN was beneficial and maintained intact glomeruli. Out of three forms of L-type neutral amino acid transporters (LAT1-LAT3) studied here, only LAT2 was found to be upregulated in the PECs and podocytes in advance of the crescent formation as well as in the crescent lesion itself. Cell culture study revealed that plasma membrane expression of LAT2 markedly stimulated mTORC1 signaling pathway, which was significantly abrogated by coexistence of LAT inhibitor. Finally, LAT inhibitor significantly abrogated development of crescent formation of CGN on day 7. Our data suggest that LAT2 may have a pivotal role in the pathogenesis of CGN by activating the mTORC1 pathway in the glomerular epithelial cells.


Assuntos
Sistema y+ de Transporte de Aminoácidos/fisiologia , Cadeias Leves da Proteína-1 Reguladora de Fusão/fisiologia , Glomerulonefrite/fisiopatologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Feminino , Glomerulonefrite/etiologia , Imuno-Histoquímica , Ratos , Ratos Endogâmicos WKY , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
9.
Lab Invest ; 91(11): 1584-95, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21876538

RESUMO

Although podocyte damage is known to be responsible for the development of minimal-change disease (MCD), the underlying mechanism remains to be elucidated. Previously, using a rat MCD model, we showed that endoplasmic reticulum (ER) stress in the podocytes was associated with the heavy proteinuric state and another group reported that a mammalian target of rapamycin complex 1 (mTORC1) inhibitor protected against proteinuria. In this study, which utilized a rat MCD model, a combination of immunohistochemistry, dual immunofluorescence and confocal microscopy, western blot analysis, and quantitative real-time RT-PCR revealed co-activation of the unfolded protein response (UPR), which was induced by ER stress, and mTORC1 in glomerular podocytes before the onset of proteinuria and downregulation of nephrin at the post-translational level at the onset of proteinuria. Podocyte culture experiments revealed that mTORC1 activation preceded the UPR that was associated with a marked decrease in the energy charge. The mTORC1 inhibitor everolimus completely inhibited proteinuria through a reduction in both mTORC1 and UPR activity and preserved nephrin expression in the glomerular podocytes. In conclusion, mTORC1 activation may perturb the regulatory system of energy metabolism primarily by promoting energy consumption and inducing the UPR, which underlie proteinuria in MCD.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Nefrose Lipoide/metabolismo , Podócitos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Animais , Western Blotting , Células Cultivadas , Everolimo , Imunofluorescência , Imuno-Histoquímica , Microscopia Confocal , Nefrose Lipoide/etiologia , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirolimo/análogos & derivados , Estatísticas não Paramétricas , Serina-Treonina Quinases TOR/antagonistas & inibidores
10.
J Am Soc Nephrol ; 20(7): 1586-96, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19443642

RESUMO

LAT3 is a Na+-independent neutral l-amino acid transporter recently isolated from a human hepatocellular carcinoma cell line. Although liver, skeletal muscle, and pancreas are known to express LAT3, the tissue distribution and physiologic function of this transporter are not completely understood. Here, we observed that glomeruli express LAT3. Immunofluorescence, confocal microscopy, and immunoelectron microscopy revealed that LAT3 localizes to the apical plasma membrane of podocyte foot processes. In mice, starvation upregulated glomerular LAT3, phosphorylated AKT1, reconstituted the actin network, and elongated foot processes. In the fetal kidney, we observed intense LAT3 expression at the capillary loops stage of renal development. Finally, zebrafish morphants lacking lat3 function showed collapsed glomeruli with thickened glomerular basement membranes. Permeability studies of the glomerular filtration barrier in these zebrafish morphants demonstrated a disruption of selective glomerular permeability. Our data suggest that LAT3 may play a crucial role in the development and maintenance of podocyte structure and function by regulating protein synthesis and the actin cytoskeleton.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Diferenciação Celular/fisiologia , Glomérulos Renais/metabolismo , Podócitos/metabolismo , Actinas/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/genética , Animais , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Feminino , Membrana Basal Glomerular/metabolismo , Taxa de Filtração Glomerular/fisiologia , Humanos , Glomérulos Renais/citologia , Glomérulos Renais/embriologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fosforilação , Podócitos/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
11.
PLoS One ; 13(8): e0202400, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30125302

RESUMO

The evidence that gene mutations in the polarity determinant Crumbs homologs-2 (CRB2) cause congenital nephrotic syndrome suggests the functional importance of this gene product in podocyte development. Because another isoform, CRB3, was reported to repress the mechanistic/mammalian target of the rapamycin complex 1 (mTORC1) pathway, we examined the role of CRB2 function in developing podocytes in relation to mTORC1. In HEK-293 and MDCK cells constitutively expressing CRB2, we found that the protein localized to the apicolateral side of the cell plasma membrane and that this plasma membrane assembly required N-glycosylation. Confocal microscopy of the neonate mouse kidney revealed that both the tyrosine-phosphorylated form and non-phosphorylated form of CRB2 commence at the S-shaped body stage at the apicolateral side of podocyte precursor cells and move to foot processes in a capillary tuft pattern. The pattern of phosphorylated mTOR in developing podocytes was similar to that of CRB2 tyrosine phosphorylation. Additionally, the lack of a tyrosine phosphorylation site on CRB2 led to the reduced sensitivity of mTORC1 activation in response to energy starvation. CRB2 may play an important role in the mechanistic pathway of developing podocytes through tyrosine phosphorylation by associating with mTORC1 activation.


Assuntos
Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Membrana/metabolismo , Podócitos/metabolismo , Células-Tronco/metabolismo , Animais , Proteínas de Transporte/genética , Membrana Celular/genética , Cães , Glicosilação , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Proteínas de Membrana/genética , Camundongos , Fosforilação/genética , Podócitos/citologia , Células-Tronco/citologia
12.
Cell Cycle ; 11(8): 1504-11, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22433955

RESUMO

Podocytes are terminally differentiated post-mitotic cells similar to neurons, and their damage leads to nephrotic syndrome, which is characterized by massive proteinuria associated with generalized edema. A recent functional genetic approach has identified the pathological relevance of several mutated proteins in glomerular podocytes to the mechanism of proteinuria in hereditary nephrotic syndrome. In contrast, the pathophysiology of acquired primary nephrotic syndrome, including minimal change disease, is still largely unknown. We recently demonstrated the possible linkage of an energy-consuming process in glomerular podocytes to the mechanism of proteinuria. Puromycin aminonucleoside nephrosis, a rat model of minimal change disease, revealed the activation of the unfolded protein response (UPR) in glomerular podocytes to be a cause of proteinuria. The pretreatment of puromycin aminonucleoside rat podocytes and cultured podocytes with the mammalian target of rapamycin (mTOR) inhibitor everolimus further revealed that mTOR complex 1 consumed energy, which was followed by UPR activation. In this paper, we will review nutritional transporters to summarize the energy uptake process in podocytes and review the involvement of the UPR in the pathogenesis of glomerular diseases. We will also present additional data that reveal how mTOR complex 1 acts upstream of the UPR. Finally, we will discuss the potential significance of targeting the energy metabolism of podocytes to develop new therapeutic interventions for acquired nephrotic syndrome.


Assuntos
Síndrome Nefrótica/metabolismo , Podócitos/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Metabolismo Energético , Everolimo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Imunossupressores/farmacologia , Proteínas de Membrana Transportadoras/metabolismo , Síndrome Nefrótica/patologia , Podócitos/efeitos dos fármacos , Proteinúria/patologia , Ratos , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Resposta a Proteínas não Dobradas
13.
J Pharmacol Sci ; 97(2): 234-41, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15684566

RESUMO

Decreased expression levels of the glomerular slit membrane proteins, nephrin and podocin, have been reported after the onset of puromycin aminonucleoside (PA) nephrosis. We examined nephrin and podocin expressions prior to the onset of proteinuria of PA nephrosis to elucidate the proteinuria induction mechanism of PA. PA nephrosis was induced by a subcutaneous single injection of 120 mg kg(-1) PA. The mRNA levels of nephrin and podocin in whole kidney total RNA were quantified by the TaqMan real time PCR quantification system. The localization and levels of nephrin and podocin molecules were analyzed by immunofluorescence and Western blotting, respectively. Albuminuria and proteinuria were significant on days 3 and 4 in PA nephrosis rats. The protein levels of nephrin and podocin decreased significantly at day 3. The protein localization of nephrin and podocin changed at day 2 and day 1, respectively. The mRNA level of nephrin increased at day 2 and subsequently decreased at day 4. The podocin mRNA level did not change significantly. In conclusions, the protein level of nephrin and podocin decreased at the onset of albuminuria in the PA nephrosis. However, the first change induced by PA was the change of podocin localization from a linear pattern to a dot-like one prior to the onset of albuminuria.


Assuntos
Proteínas de Membrana/biossíntese , Nefrose/metabolismo , Puromicina Aminonucleosídeo/toxicidade , Animais , Peptídeos e Proteínas de Sinalização Intracelular , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Nefrose/induzido quimicamente , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley
14.
Kidney Int ; 66(5): 1755-65, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15496146

RESUMO

BACKGROUND: Podocin is a membrane-integrated protein that is located at the glomerular slit diaphragm and directly interacts with nephrin. The gene encoding podocin, NPHS2, is mutated in patients with autosomal-recessive steroid-resistant nephrotic syndrome (SRN). In order to study a potential pathomechanism of massive proteinuria in patients with SRN, we have investigated the trafficking and subcellular localization of five common disease-causing missense mutants of human podocin. METHODS: Site-directed mutagenesis was applied to generate cDNA constructs encoding five different missense mutations of human podocin (P20L, G92C, R138Q, V180M, and R291W). To identify the subcellular localization of each mutant in transfected human embryonic kidney (HEK)293 cells, we have generated and characterized a rabbit polyclonal antibody against the human podocin. Specificity of the antibody was determined by light and immunoelectron microscopy, as well as immunoblot analysis using human glomeruli. Confocal microscopy was applied to determine subcellular localization of the wild-type and the mutated podocin molecules, as well as wild-type nephrin in transfected cells. Immunoprecipitation and pull-down studies were carried out to investigate the molecular interaction of podocin mutants and wild-type nephrin. RESULTS: Immunofluorescence and confocal microscopy showed that wild-type podocin located to the plasma membrane when expressed in HEK293 cells. Two missense mutations, P20L and G92C, located at the N-terminus part of the molecule, were also present at the plasma membrane, indicating that these mutations did not affect the subcellular localization of the mutated podocin molecules. In contrast, subcellular localization of three other missense mutants located in the proximal C-terminus part of the protein was drastically altered, in which R138Q was retained in the endoplasmic reticulum (ER), V180M formed inclusion bodies in the cytoplasm, and the R291W mutant was trapped both in the ER and in small intracellular vesicles. Interestingly, this abnormal subcellular localization of podocin missense mutants also resulted in alteration in protein trafficking of wild-type nephrin in cotransfected cells through the strong protein binding between both molecules. CONCLUSION: In patients with SRN, some missense mutations in the NPHS2 gene not only lead to misfolding and mislocalization of the mutated podocin, but they can also interfere with slit diaphragm structure and function by altering the proper trafficking of nephrin to the plasma membrane.


Assuntos
Resistência a Medicamentos/genética , Rim/metabolismo , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Síndrome Nefrótica/genética , Proteínas/metabolismo , Esteroides/uso terapêutico , Linhagem Celular , Membrana Celular/metabolismo , Genes Recessivos , Humanos , Técnicas Imunológicas , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/metabolismo , Microscopia Imunoeletrônica , Síndrome Nefrótica/tratamento farmacológico , Transporte Proteico , Frações Subcelulares/metabolismo , Distribuição Tecidual , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA