Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Lupus Sci Med ; 11(1)2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38906550

RESUMO

OBJECTIVE: Systemic lupus erythematosus (SLE) is a type of autoimmune disease that involves multiple organs involved as well as cytokine dysregulation. The treatment of SLE is still challenging due to the side effects of the different drugs used. Receptor-interacting protein kinase 1 (RIPK1) is a kinase involved in T cell homeostasis and autoinflammation. Although clinical trials have shown that RIPK1 inhibition exhibits significant efficacy in different autoimmune diseases, its role in SLE remains unclear. METHODS: MRL/lpr lupus-prone mice received RIPK1 inhibitor ZJU37 or vehicle intraperitoneally for 10 weeks. A BM12-induced chronic graft-versus-host-disease (cGVHD) lupus-like model was introduced in RIPK1 D138N mice or C57BL/6 mice. Nephritis, serum autoantibody levels, dysregulation of adaptive immune response and cytokines were compared in treated and untreated mice. RESULTS: ZJU37 alleviated the clinical features of the MRL/lpr mice including nephritis and anti-dsDNA antibody production. In addition, ZJU37 treatment reduced the proportion of double-negative T cells in the spleen and the cytokines of TNFα, IFN-γ, IL-6, IL-17 and IL-1ß in the serum. Moreover, RIPK1 D138N mice were able to prevent the cGVHD lupus-like model from SLE attack, manifesting as anti-dsDNA antibody production, the proliferation of germinal centre B cells, plasma cells, and T follicular helper cells as well as IgG and C3 deposits in kidneys. CONCLUSION: RIPK1 inhibition has a protective effect in the mouse model of SLE and can potentially become a new therapeutic target for SLE in humans.


Assuntos
Citocinas , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro , Lúpus Eritematoso Sistêmico , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Proteína Serina-Treonina Quinases de Interação com Receptores , Animais , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Camundongos , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/imunologia , Citocinas/metabolismo , Feminino , Anticorpos Antinucleares/sangue , Nefrite Lúpica/tratamento farmacológico , Nefrite Lúpica/imunologia , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Baço/imunologia , Baço/efeitos dos fármacos
2.
Biomed Pharmacother ; 167: 115617, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37801905

RESUMO

Podocyte injury is the common initiating event in focal segmental glomerulosclerosis (FSGS). Oxidative stress and inflammation mediate podocyte injury in FSGS. NRF2 pathway regulates the constitutive and inducible transcription of various genes that encode antioxidant proteins and anti-inflammatory proteins and have pivotal roles in the defense against cellular oxidative stress. In this study, we used adriamycin-induced nephropathy (ADR) in mice as a model of FSGS to confirm that CDDO-Me treatment ameliorated adriamycin-induced kidney damage by improving renal function and kidney histology. CDDO-Me inhibited the level of oxidative stress, inflammation, and apoptosis in adriamycin-induced podocyte injury by activating NRF2 pathway in vivo and in vitro. Furthermore, CDDO-Me stabled the cytoskeleton by regulating NRF2/srGAP2a pathway. Together, these findings show that by activating NRF2 pathway, CDDO-Me could be a therapeutic strategy to prevent the adverse effects of adriamycin-induced podocyte injury.


Assuntos
Glomerulosclerose Segmentar e Focal , Nefropatias , Podócitos , Camundongos , Animais , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Doxorrubicina/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Nefropatias/induzido quimicamente , Nefropatias/tratamento farmacológico , Nefropatias/metabolismo , Citoesqueleto de Actina/metabolismo , Estresse Oxidativo , Inflamação/metabolismo
3.
Commun Biol ; 6(1): 912, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37670124

RESUMO

The occurrence of diabetic nephropathy (DN) and diabetic retinopathy (DR) are closely associated in patients with diabetes. However, the cellular and molecular linkage of DN and DR has not been elucidated, and further revelations are needed to improve mutual prognostic decisions and management. Here, we generate and integrate single-cell RNA sequencing profiles of kidney and retina to explore the cellular and molecular association of kidney and retina in both physiological and pathological conditions. We find renal mesangial cells and retinal pericytes share molecular features and undergo similar molecular transitions under diabetes. Furthermore, we uncover that chemokine regulation shared by the two cell types is critical for the co-occurrence of DN and DR, and the chemokine score can be used for the prognosis of DN complicated with DR. These findings shed light on the mechanism of the co-occurrence of DN and DR and could improve the prevention and treatments of diabetic microvascular complications.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Retinopatia Diabética , Humanos , Transcriptoma , Rim , Células Mesangiais
4.
Adv Sci (Weinh) ; 10(25): e2301492, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37395441

RESUMO

T cells play an important role in the development of focal segmental glomerulosclerosis (FSGS). The mechanism underlying such T cell-based kidney disease, however, remains elusive. Here the authors report that activated CD8 T cells elicit renal inflammation and tissue injury via releasing miR-186-5p-enriched exosomes. Continuing the cohort study identifying the correlation of plasma level of miR-186-5p with proteinuria in FSGS patients, it is demonstrated that circulating miR-186-5p is mainly derived from activated CD8 T cell exosomes. Renal miR-186-5p, which is markedly increased in FSGS patients and mice with adriamycin-induced renal injury, is mainly delivered by CD8 T cell exosomes. Depleting miR-186-5p strongly attenuates adriamycin-induced mouse renal injury. Supporting the function of exosomal miR-186-5p as a key circulating pathogenic factor, intravenous injection of miR-186-5p or miR-186-5p-containing T cell exosomes results in mouse renal inflammation and tissue injury. Tracing the injected T cell exosomes shows their preferential distribution in mouse renal tubules, not glomerulus. Mechanistically, miR-186-5p directly activates renal tubular TLR7/8 signal and initiates tubular cell apoptosis. Mutating the TLR7-binding sequence on miR-186-5p or deleting mouse TLR7 largely abolishes renal tubular injuries induced by miR-186-5p or adriamycin. These findings reveal a causative role of exosomal miR-186-5p in T cell-mediated renal dysfunction.


Assuntos
Linfócitos T CD8-Positivos , Exossomos , Inflamação , Nefropatias , Túbulos Renais , MicroRNAs , Transdução de Sinais , Receptor 7 Toll-Like , Receptor 8 Toll-Like , Animais , Humanos , Masculino , Camundongos , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Exossomos/genética , Exossomos/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Nefropatias/metabolismo , Nefropatias/patologia , Túbulos Renais/metabolismo , Receptor 7 Toll-Like/metabolismo , MicroRNAs/metabolismo , Receptor 8 Toll-Like/metabolismo
5.
J Proteome Res ; 22(6): 1779-1789, 2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37191251

RESUMO

Although diabetic nephropathy (DN) is the leading cause of the end-stage renal disease, the exact regulation mechanisms remain unknown. In this study, we integrated the transcriptomics and proteomics profiles of glomeruli isolated from 50 biopsy-proven DN patients and 25 controls to investigate the latest findings about DN pathogenesis. First, 1152 genes exhibited differential expression at the mRNA or protein level, and 364 showed significant association. These strong correlated genes were divided into four different functional modules. Moreover, a regulatory network of the transcription factors (TFs)-target genes (TGs) was constructed, with 30 TFs upregulated at the protein levels and 265 downstream TGs differentially expressed at the mRNA levels. These TFs are the integration centers of several signal transduction pathways and have tremendous therapeutic potential for regulating the aberrant production of TGs and the pathological process of DN. Furthermore, 29 new DN-specific splice-junction peptides were discovered with high confidence; these peptides may play novel functions in the pathological course of DN. So, our in-depth integrative transcriptomics-proteomics analysis provided deeper insights into the pathogenesis of DN and opened the potential avenue for finding new therapeutic interventions. MS raw files were deposited into the proteomeXchange with the dataset identifier PXD040617.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Humanos , Nefropatias Diabéticas/genética , Multiômica , Perfilação da Expressão Gênica , Fatores de Transcrição/genética , RNA Mensageiro
6.
Arch Biochem Biophys ; 739: 109578, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36948351

RESUMO

Podocyte injury plays a critical role in diabetic kidney disease (DKD). Our previous work demonstrated a protective role of tyrosine-protein kinase receptor TYRO3 in glomerular disease; However, the downstream signaling of TYRO3 remains unclear. Our data showed that genetic ablation of tyro3 in zebrafish recapitulated a nephrotic syndrome phenotype. TYRO3 expression was suppressed by high glucose and TGF-ß, which may contribute to the decreased TYRO3 expression in progressive DKD. Moreover, knockdown of TYRO3 expression with siRNA induced podocytes apoptosis and cytoskeleton rearrangement. Further study revealed that TYRO3 conferred antiapoptotic effects through the activation of JNK/c-jun-P53 in podocytes. Our results revealed a novel signaling module of TYRO3 in podocyte homeostasis, which provides a new molecular insight of TYRO3 effect in podocyte protection.


Assuntos
Nefropatias Diabéticas , Podócitos , Animais , Podócitos/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra/metabolismo , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Transdução de Sinais , Apoptose
7.
J Transl Med ; 21(1): 86, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36747266

RESUMO

BACKGROUND: Diabetic nephropathy (DN) is a complex disease involving the upregulation of many inflammation-related proteins. Alternative polyadenylation (APA), a crucial post-transcriptional regulatory mechanism, has been proven to play vital roles in many inflammatory diseases. However, it is largely unknown whether and how APA exerts function in DN. METHODS: We performed transcriptomics and proteomics analysis of glomeruli samples isolated from 50 biopsy-proven DN patients and 25 control subjects. DaPars and QAPA algorithms were adopted to identify APA events from RNA-seq data. The qRT-PCR analysis was conducted to verify 3'UTR length alteration. Short and long 3'UTRs isoforms were also overexpressed in podocytes under hyperglycemia condition for examining protein expression. RESULTS: We detected transcriptome-wide 3'UTR APA events in DN, and found that APA-mediated 3'UTR lengthening of genes (APA genes) increased their expression at protein but not mRNA level. Increased protein level of 3'UTR lengthening gene was validated in podocytes under hyperglycemia condition. Pathway enrichment analysis showed that APA genes were enriched in inflammation-related biological processes including endoplasmic reticulum stress pathways, NF-κB signaling and autophagy. Further bioinformatics analysis demonstrated that 3'UTR APA of genes probably altered the binding sites for RNA-binding proteins, thus enhancing protein translation. CONCLUSION: This study revealed for the first time that 3'UTR lengthening of APA genes contributed to the progression of DN by elevating the translation of corresponding proteins, providing new insight and a rich resource for investigating DN mechanisms.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Humanos , Poliadenilação , Transcriptoma/genética , Regiões 3' não Traduzidas/genética , Nefropatias Diabéticas/genética , Proteômica , Inflamação/genética , Biossíntese de Proteínas
8.
Kidney Dis (Basel) ; 8(4): 308-318, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36157258

RESUMO

Introduction: Membranous nephropathy (MN) is a common chronic kidney disease in adults and a major challenge of clinical practice for its treatment. Despite major advances, since the discovery of the phospholipase A2 receptor as the major autoantigen of podocytes in MN, the mechanisms leading to glomerular damage remain elusive. Pyroptosis, a newly discovered type of programed necrotic cell death mainly mediated by gasdermin, was found to be responsible for podocyte injury in MN in our recent work. Objectives: The aim of this study was to explore the therapeutic effect of an FDA-approved drug, disulfiram (DSF), in the treatment of MN by inhibiting pyroptosis. Methods and Results: DSF significantly alleviated C3a/C5a-induced podocyte injury in vitro and renal lesions in passive Heymann nephritis (PHN) rats, as reflected by the decreased percentage of propidium iodide staining podocytes, decreased lactate dehydrogenase release from cultured podocytes and improvement in 24-h urine protein, serum albumin, serum creatinine, abnormal alterations of podocyte injury markers Desmin and WT-1 and podocyte foot process fusion in PHN rats. The protective effect of DSF on podocyte injury in vitro and in vivo can be ascribed to its inhibition of the activation and membrane translocation of the pyroptosis executor gasdermin D (GSDMD) in podocytes. DSF also inhibited the increase and activation of the pyroptosis signaling pathway NLRP3-ASC-Caspase-1/IL-18/GSDMD in C3a/C5a-treated podocytes and renal tissue of PHN rats. Conclusion: DSF is a potential drug for MN treatment, and its clinical application needs to be further investigated.

9.
Am J Transl Res ; 13(11): 12249-12263, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34956451

RESUMO

Glomerular diseases are the leading cause of chronic kidney diseases with the pathomechanisms largely unclear. ANGPT2 is known to regulate endothelial cell homeostasis through TEK/Tie2 and its dysregulation causes endothelial damage. Here, we found that ANGPT2 is upregulated in glomerular diseases and wondered whether it also acts on the other two glomerular cell types, podocytes and mesangial cells. We treated podocytes and mesangial cells in culture with ANGPT2 but didn't find changes in morphology and survival. RNA-seq analysis revealed that gene expression was altered in both podocytes and mesangial cells and that the differentially expressed genes in the two cell types were fundamentally different and enriched in distinct cellular processes and pathways according to GO and KEGG analyses. Mechanistically, the Ingenuity Pathway Analysis (IPA) analysis revealed that ERK and AKT were the most connected nodes in the networks of the regulated genes of both podocytes and mesangial cells, suggesting that ANGPT2 affected ERK and AKT in both cell types. Interestingly, immunoblotting showed that phosphorylated ERK and AKT were both increased in podocytes while decreased in mesangial cells by ANGPT2. We found that mesangial cells, but not podocytes, expressed TEK and ANGPT1, suggesting that ANGPT2 could antagonize ANGPT1-TEK-ERK axis in mesangial cells similarly to endothelial cells. We searched databases and found that integrin alpha(v) (ITGAV) is an ANGPT2 interacting protein and expressed in podocytes, suggesting that ITGAV mediates ANGPT2 effect on podocytes. In conclusion, increased ANGPT2 may be involved in glomerular injury by affecting podocytes and mesangial cells in addition to endothelial cells. The complexity of the effect of ANGPT2 in glomeruli may apply to other factors.

10.
Front Physiol ; 12: 649801, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149444

RESUMO

C3 glomerulopathy (C3GP) is a disease entity caused by abnormality of the complement alternative pathway (AP) and characterized by C3 deposition in glomeruli. Many variations or mutations of complement factors are believed to underlie the susceptibility to C3GP, but there is a lack of experimental evidence. We have recently reported a patient with C3 glomerulonephritis (C3GN) and compound heterozygosity of two novel variations in the complement factor (CFI). Here, we generated a mouse model to mimic the CFI variations for studying pathogenicity of CFI variations in C3GN development. We used the CRISPR/Cas9 system to make mutant mouse lines that carried D288G and P467S mutations in CFI, respectively, and crossed them to generate mice with compound heterozygosity of CFI D288G and P467S. The mice were all normal in either SPF (specific pathogen free) or regular environment. When treated with lipopolysaccharides (LPS), a bacterial endotoxin that mimics infection and sepsis, the mice developed albuminuria, kidney function impairment, and C3 glomerular deposition at levels comparable with the wild-type mice. The mice with other genotypes concerning CFI D288G and P467S were also tested in parallel. Unexpectedly, we found that the D288G homozygotes all developed severe mesangial deposition of C3 in the LPS model, indicating that CFI D288G variation was involved in the C3 deposition, a key feature of C3GN. The mouse lines generated in the present study can be used to further study the role of CFI variations in C3GN development; in addition, they may be used to screen and test infections and environmental factors capable of triggering C3GN.

11.
J Cancer ; 12(3): 849-859, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33403042

RESUMO

Platinum-based adjuvant chemotherapy is very common for gastric cancer (GC) patients, but the chemotherapy sensitivity is very heterogeneous. The genomic variants and the gene-gene interactions involved in Fas-mediated apoptosis pathway including Fas (FAS 1377 G > A and 670 A > G), FasL (FASL 844 C > T) and caspase-8 (CASP8 -652 6N ins > del or I > D), may paly vital roles in the response to platinum-based treatment. In our investigation, 662 stage II-III postoperative GC patients were enrolled between 1998 and 2006. 261 patients accepted platinum-based regimens and the remaining 401 were not. The log rank tests, Kaplan Meier plots, Pearson chi-square tests, Student t-tests and Cox regression analyses were performed. For the chemotherapy cohort, FAS 1377 G > A or FAS 670 A > G variants alone was related with inferior survival, and a greater than additive effect was identified when patients simultaneously carrying FAS 1377 GA and FAS 670 GA genotypes. But the poor response was neutralized when patients simultaneously carrying FASL 844 C > T or CASP8 -652 6N ins > del mutations. Our study suggested that FAS 1377 G > A and FAS 670 A > G variants may serve as potential biomarkers to predict the response to platinum-based adjuvant chemotherapy, and the gene-gene interactions involved in Fas-mediated apoptosis pathway may enhance or neutralize the chemosensitivity.

12.
J Exp Med ; 218(3)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33346797

RESUMO

Podocyte injury is a common hallmark in various glomerular diseases. The level of LRRC55 was increased in podocytes of patients with focal segmental glomerulosclerosis (FSGS), diabetic nephropathy (DN), and membranous nephropathy (MN). Upregulated LRRC55 and increased intracellular Ca2+ led to BK channel activation and the loss of intracellular potassium, resulting in apoptosome formation and caspase-3 activation in angiotensin II (Ang II)-treated podocytes. Knockout of Lrrc55 or the BK channel prevented the BK current and ameliorated podocyte injury in Ang II-treated mice. Upstream, NFATc3 regulated the expression of LRRC55. Increased LRRC55 expression in podocytes was also evident in animal models of FSGS, DN, and MN. Treatment with losartan or LRRC55 siRNA suppressed LRRC55 expression, prevented BK channel activation, and attenuated podocyte injury in animal models of FSGS, DN, and MN. In conclusion, upregulated LRRC55 promotes BK channel activation and aggravates cell injury in podocytes in FSGS, DN, and MN. LRRC55 inhibition may represent a new therapeutic approach for podocyte injury.


Assuntos
Ativação do Canal Iônico , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Proteínas de Membrana/genética , Podócitos/metabolismo , Podócitos/patologia , Regulação para Cima , Angiotensina II , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Humanos , Espaço Intracelular/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Losartan/farmacologia , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Fatores de Transcrição NFATC/metabolismo , Podócitos/ultraestrutura , Potássio/metabolismo , Regiões Promotoras Genéticas/genética , Transporte Proteico/efeitos dos fármacos , Canal de Cátion TRPC6/metabolismo , Regulação para Cima/efeitos dos fármacos
13.
Medicine (Baltimore) ; 99(5): e18960, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32000422

RESUMO

To investigate the changes in blood coagulability as measured by thromboelastography (TEG) in patients with nephrotic syndrome of different etiologies as well as in patients with venous thromboembolic events (VTE).From January 2013 to October 2017, patients who were diagnosed as idiopathic membranous nephropathy (IMN), minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS) were enrolled into this retrospective study in which their clinical characteristics, including TEG variables, were investigated. According to the presence or absence of VTE, the patients with IMN were divided into 2 groups of VTE and non-VTE. The risk factors of VTE were analyzed with logistic regression.Significant differences in TEG parameters were found among the 3 groups of patients with R and K values lower, while the α-angle, maximum amplitude (MA) and confidence interval (CI) values higher, in the IMN group than those in the MCD and FSGS groups (P < .01). Multiple linear regression analysis indicated that the histologic subtype was an independent relevant factor of K time, angle, MA, and CI values. Multivariate logistic regression analysis revealed that serum albumin and CI value were independent risk factors of VTE (P < .05).The results showed that IMN patients may have higher whole blood coagulability than MCD and FSGS patients. The hypercoagulability in IMN patients may be attributed to platelet hyperactivity and the accelerated fibrin-platelet interaction. Hypoproteinemia and increased CI value were independent risk factors of VTE in IMN.


Assuntos
Síndrome Nefrótica/complicações , Tromboelastografia/métodos , Trombofilia/diagnóstico , Trombofilia/etiologia , Adulto , Feminino , Glomerulosclerose Segmentar e Focal/complicações , Humanos , Masculino , Pessoa de Meia-Idade , Nefrose Lipoide/complicações , Estudos Retrospectivos , Fatores de Risco
14.
Arch Biochem Biophys ; 671: 210-217, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31330131

RESUMO

Podocytes are important to glomerular filtration barrier integrity and maintenance of size selectivity in protein filtration in the kidney. Although there is evidence to suggest that triptolide has direct protective effects on podocyte injuries, the mechanism mediating this process remains largely unexplored. In this study, we found triptolide suppresses podocyte p53 and GADD45B expression in vivo and in vitro. We used our previously developed in vivo zebrafish model of inducible podocyte-targeted injury and found that triptolide or the inhibition of p53 and gadd45ba with morpholino (MO) alleviated metronidazole (MTZ) induced edema in zebrafish, while the overexpression of gadd45ba in podocytes blocked the protective effect of triptolide and p53 MO on podocyte injury in zebrafish. Further study showed that p53 directly transactivated GADD45B. Triptolide inhibited p53 binding to the GADD45B promoter and subsequent GADD45B transcription. We further demonstrated that p53 may indirectly regulate GADD45B expression via NF-κB signaling. Taken together, our findings demonstrated that triptolide maintained glomerular barrier function via the inhibition of p53-NF-κB-GADD45B signaling, which provides a new understanding of the antiproteinuric effects of triptolide in glomerular diseases.


Assuntos
Antígenos de Diferenciação/metabolismo , Diterpenos/farmacologia , Barreira de Filtração Glomerular/efeitos dos fármacos , Fenantrenos/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Edema/induzido quimicamente , Compostos de Epóxi/farmacologia , Metronidazol , NF-kappa B/metabolismo , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Ligação Proteica/efeitos dos fármacos , Puromicina Aminonucleosídeo , Transdução de Sinais/efeitos dos fármacos , Saco Vitelino/patologia , Peixe-Zebra
16.
J Clin Pathol ; 72(10): 705-711, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31243053

RESUMO

AIMS: Phospholipase A2 receptor (PLA2R) and thrombospondin type-1 domain-containing 7A (THSD7A) were identified as pathogenic antigens in patients with membranous nephropathy (MN). Notably, PLA2R is detected in few patients with malignancy-associated MN, and a high incidence of cancer is reported in patients with THSD7A-associated MN. Therefore, the roles of PLA2R and THSD7A in malignancy-associated MN must be clarified. METHODS: Serum anti-PLA2R antibodies and glomerular PLA2R staining were assessed in 36 patients with malignancy-associated MN, followed by examination of serum anti-THSD7A antibodies and glomerular THSD7A. THSD7A staining in cancer tissues was also assessed in 9 of the 36 patients. RESULTS: Twelve (33%) of 36 patients were positive for both glomerular PLA2R and serum anti-PLA2R antibodies, one of whom had enhanced glomerular THSD7A staining. Two patients were positive for either glomerular PLA2R or serum anti-PLA2R antibody. All these patients had IgG4-dominant deposits in glomeruli. Among the 22 (61%) patients who were double negative for glomerular PLA2R and serum anti-PLA2R antibodies, 17 of 20 (85%) had IgG1-dominant deposits in glomeruli, and 2 (9.1%) were positive for glomerular THSD7A staining. Serum anti-THSD7A antibody was not detected in any of the 36 patients. Among the nine patients with available cancer tissues, positive staining of THSD7A in the cancer tissues was observed in five (56%) patients, and one showed enhanced glomerular staining of THSD7A. CONCLUSIONS: Screening of glomerular PLA2R antigen and serum anti-PLA2R antibodies is necessary in patients with malignancy-associated MN, whereas the incidence of glomerular THSD7A antigen or circulating anti-THSD7A antibodies is uncommon.


Assuntos
Glomerulonefrite Membranosa/enzimologia , Receptores da Fosfolipase A2/metabolismo , Trombospondinas/metabolismo , Feminino , Glomerulonefrite Membranosa/patologia , Humanos , Imunoglobulina G/sangue , Masculino , Neoplasias
17.
JCI Insight ; 4(7)2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30944246

RESUMO

Chronic tubulointerstitial injury impacts the prognosis of focal segmental glomerulosclerosis (FSGS). We found that the level of versican V1 was increased in tubular cells of FSGS patients. Tubular cell-derived versican V1 induced proliferation and collagen synthesis by activating the CD44/Smad3 pathway in fibroblasts. Both urine C3a and suPAR were increased and bound to the tubular cells in FSGS patients. C3a promoted the transcription of versican by activating the AKT/ß-catenin pathway. C3aR knockout decreased the expression of versican in Adriamycin-treated (ADR-treated) mice. On the other hand, suPAR bound to integrin ß6 and activated Rac1, which bound to SRp40 at the 5' end of exon 7 in versican pre-mRNA. This binding inhibited the 3'-end splicing of intron 6 and the base-pair interactions between intron 6 and intron 8, leading to the formation of versican V1. Cotreatment with ADR and suPAR specifically increased the level of versican V1 in tubulointerstitial tissues and caused more obvious interstitial fibrosis in mice than treatment with only ADR. Altogether, our results show that C3a and suPAR drive versican V1 expression in tubular cells by promoting transcription and splicing, respectively, and the increases in tubular cell-derived versican V1 induce interstitial fibrosis by activating fibroblasts in FSGS.


Assuntos
Complemento C3a/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Túbulos Renais/patologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Versicanas/metabolismo , Adulto , Animais , Biópsia , Estudos de Casos e Controles , Linhagem Celular , Complemento C3a/urina , Modelos Animais de Doenças , Doxorrubicina/toxicidade , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Fibrose , Perfilação da Expressão Gênica , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/urina , Humanos , Túbulos Renais/citologia , Túbulos Renais/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/urina , Receptores de Complemento/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/análise , Versicanas/urina , Adulto Jovem
18.
Sci Rep ; 9(1): 716, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30679767

RESUMO

Tubulointerstitial fibrosis impacts renal prognosis of focal segmental glomerulosclerosis (FSGS). Based on transcriptomic analysis, we found that the level of LOC105375913 was increased in tubular cells of FSGS patients. C3a induced the expression of LOC105375913, which promoted the expression of fibronectin and collagen I in tubular cells. Silence of snail reversed the level of fibronectin and collagen I in cells overexpressing LOC105375913. MiR-27b was predicted and confirmed to regulate the expression of snail in tubular cells, and LOC105375913 contained the response element of miR-27b. The competitive binding between LOC105375913 and miR-27b increased the level of snail and promoted fibrogenesis in tubular cells. Upstream, p38 and XBP-1s regulated the expression of LOC105375913. Inhibition of p38 or silence of XBP-1s decreased the level of LOC105375913, and suppressed the expression of snail, fibronectin and collagen I in tubular cells treated with C3a. Overexpression of LOC105375913 decreased the level of miR-27b, increased the level of snail and caused tubulointerstitial fibrosis in mice. In conclusion, the activation of C3a/p38/XBP-1s pathway induces the expression of LOC105375913 in tubular cells, and LOC105375913 increases the level of snail and induces tubulointerstitial fibrosis through competitive binding of miR-27b in tubular cells of FSGS patients.


Assuntos
Biomarcadores/análise , Fibrose/diagnóstico , Regulação da Expressão Gênica , Glomerulosclerose Segmentar e Focal/complicações , Nefropatias/diagnóstico , Túbulos Renais/patologia , RNA Longo não Codificante/genética , Adulto , Animais , Estudos de Casos e Controles , Complemento C3a/genética , Complemento C3a/metabolismo , Feminino , Fibrose/etiologia , Fibrose/patologia , Glomerulosclerose Segmentar e Focal/genética , Humanos , Nefropatias/etiologia , Nefropatias/patologia , Túbulos Renais/metabolismo , Masculino , Camundongos , MicroRNAs/genética , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
J Biol Chem ; 293(52): 20227-20239, 2018 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-30389788

RESUMO

Focal segmental glomerulosclerosis (FSGS) is a common kidney disease that results in nephrotic syndrome. FSGS arises from dysfunction and apoptosis of podocytes in the glomerulus of the kidney, leading to podocytopathy. The molecular mechanisms underlying podocyte apoptosis remain incompletely understood. Using an array of gene expression profiling, PCR, and in situ hybridization assay, we found here that the levels of the long noncoding RNA LOC105374325 were elevated in the renal podocytes of individuals with FSGS. We also observed that the microRNAs miR-34c and miR-196a/b down-regulated the expression of the apoptosis regulators BCL2-associated X, apoptosis regulator (Bax), and BCL2 antagonist/killer 1 (Bak) in podocytes. Competitive binding between LOC105374325 and miR-34c or miR-196a/b increased Bax and Bak levels and caused podocyte apoptosis. Of note, the mitogen-activated protein kinase P38 and the transcription factor CCAAT enhancer-binding protein ß (C/EBPß) up-regulated LOC105374325 expression. P38 inhibition or C/EBPß silencing decreased LOC105374325 levels and inhibited apoptosis in adriamycin-treated podocytes. LOC105374325 overexpression decreased miR-34c and miR-196a/b levels, increased Bax and Bak levels, and induced proteinuria and focal segmental lesions in mice. In conclusion, activation of the P38/C/EBPß pathway stimulates the expression of LOC105374325, which, in turn, increases Bax and Bak levels and causes apoptosis by competitively binding to miR-34c and miR-196a/b in the podocytes of individuals with FSGS.


Assuntos
Glomerulosclerose Segmentar e Focal/metabolismo , Podócitos/metabolismo , RNA Longo não Codificante/biossíntese , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Células Cultivadas , Doxorrubicina/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Masculino , MicroRNAs/biossíntese , Podócitos/patologia , Transdução de Sinais/efeitos dos fármacos , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
20.
Diabetes ; 67(4): 717-730, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29242313

RESUMO

Podocytes play a pivotal role in maintaining glomerular filtration function through their interdigitated foot processes. However, the mechanisms that govern the podocyte cytoskeletal rearrangement remain unclear. Through analyzing the transcriptional profile of renal biopsy specimens from patients with diabetic nephropathy (DN) and control donors, we identify SLIT-ROBO ρGTPase-activating protein 2a (SRGAP2a) as one of the main hub genes strongly associated with proteinuria and glomerular filtration in type 2 DN. Immunofluorescence staining and Western blot analysis revealed that human and mouse SRGAP2a is primarily localized at podocytes and largely colocalized with synaptopodin. Moreover, podocyte SRGAP2a is downregulated in patients with DN and db/db mice at both the mRNA and the protein level. SRGAP2a reduction is observed in cultured podocytes treated with tumor growth factor-ß or high concentrations of glucose. Functional and mechanistic studies show that SRGAP2a suppresses podocyte motility through inactivating RhoA/Cdc42 but not Rac1. The protective role of SRGAP2a in podocyte function also is confirmed in zebrafish, in which knockdown of SRGAP2a, a SRGAP2 ortholog in zebrafish, recapitulates podocyte foot process effacement. Finally, increasing podocyte SRGAP2a levels in db/db mice through administration of adenovirus-expressing SRGAP2a significantly mitigates podocyte injury and proteinuria. The results demonstrate that SRGAP2a protects podocytes by suppressing podocyte migration.


Assuntos
Proteínas de Transporte/genética , Diabetes Mellitus Tipo 2/metabolismo , Nefropatias Diabéticas/genética , Proteínas Ativadoras de GTPase/genética , Glomérulos Renais/metabolismo , Podócitos/ultraestrutura , Adulto , Animais , Proteínas de Transporte/metabolismo , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Modelos Animais de Doenças , Feminino , Proteínas Ativadoras de GTPase/metabolismo , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Peixe-Zebra , Proteínas de Peixe-Zebra , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...