Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
1.
Mol Ther ; 32(9): 3177-3193, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-38910328

RESUMO

Transforming growth factor (TGF)-ß signaling is a well-established pathogenic mediator of diabetic kidney disease (DKD). However, owing to its pleiotropic actions, its systemic blockade is not therapeutically optimal. The expression of TGF-ß signaling regulators can substantially influence TGF-ß's effects in a cell- or context-specific manner. Among these, leucine-rich α2-glycoprotein 1 (LRG1) is significantly increased in glomerular endothelial cells (GECs) in DKD. As LRG1 is a secreted molecule that can exert autocrine and paracrine effects, we examined the effects of LRG1 loss in kidney cells in diabetic OVE26 mice by single-cell transcriptomic analysis. Gene expression analysis confirmed a predominant expression of Lrg1 in GECs, which further increased in diabetic kidneys. Loss of Lrg1 led to the reversal of angiogenic and TGF-ß-induced gene expression in GECs, which were associated with DKD attenuation. Notably, Lrg1 loss also mitigated the increased TGF-ß-mediated gene expression in both podocytes and mesangial cells in diabetic mice, indicating that GEC-derived LRG1 potentiates TGF-ß signaling in glomerular cells in an autocrine and paracrine manner. Indeed, a significant reduction in phospho-Smad proteins was observed in the glomerular cells of OVE26 mice with LRG1 loss. These results indicate that specific antagonisms of LRG1 may be an effective approach to curb the hyperactive glomerular TGF-ß signaling to attenuate DKD.


Assuntos
Nefropatias Diabéticas , Células Endoteliais , Glicoproteínas , Glomérulos Renais , Transdução de Sinais , Fator de Crescimento Transformador beta , Animais , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/patologia , Camundongos , Fator de Crescimento Transformador beta/metabolismo , Glicoproteínas/metabolismo , Glicoproteínas/genética , Células Endoteliais/metabolismo , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Diabetes Mellitus Experimental/metabolismo , Humanos , Podócitos/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica
2.
J Bioenerg Biomembr ; 56(5): 543-552, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39120858

RESUMO

Diabetic nephropathy (DN) is one of microvascular complication associated with diabetes. Circular RNAs (circRNAs) have been shown to be involved in DN pathogenesis. Hence, this work aimed to explore the role and mechanism of circ_Arf3 in DN. Mouse mesangial cells (MCs) cultured in high glucose (HG) condition were used for functional analysis. Cell proliferation was determined using 5-ethynyl-2'-deoxyuridine (EdU) and cell counting kit-8 assays. Western blotting was used to measure the levels of proliferation indicator PCNA and fibrosis-related proteins α-smooth muscle actin (α-SMA), collagen I (Col I), fibronectin (FN), and collagen IV (Col IV). The binding interaction between miR-107-3p and circ_Arf3 or Tmbim6 (transmembrane BAX inhibitor motif containing 6) was confirmed using dual-luciferase reporter and pull-down assays. Circ_Arf3 is a stable circRNA, and the expression of circ_Arf3 was decreased after HG treatment in MCs. Functionally, ectopic overexpression of circ_Arf3 protected against HG-induced proliferation and elevation of fibrosis-related proteins in MCs. Mechanistically, circ_Arf3 directly bound to miR-107-3p, and Tmbim6 was a target of miR-107-3p. Further rescue assay showed miR-107-3p reversed the protective action of circ_Arf3 on MCs function under HG condition. Moreover, inhibition of miR-107-3p suppressed HG-induced proliferation and fibrosis, which were attenuated by Tmbim6 knockdown in MCs. CircRNA Arf3 could suppress HG-evoked mesangial cell proliferation and fibrosis via miR-107-3p/Tmbim6 axis, indicating the potential involvement of this axis in DN progression.


Assuntos
Proteínas Reguladoras de Apoptose , Proliferação de Células , Nefropatias Diabéticas , Fibrose , Proteínas de Membrana , Células Mesangiais , MicroRNAs , RNA Circular , Animais , Humanos , Camundongos , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Células Mesangiais/metabolismo , Células Mesangiais/patologia , MicroRNAs/metabolismo , MicroRNAs/genética , RNA Circular/metabolismo , RNA Circular/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo
3.
FASEB J ; 37(5): e22926, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37052733

RESUMO

Glomerulosclerosis is one of the major histopathologic changes in diabetic kidney diseases (DKD), which is characterized by excessive deposition of extracellular matrix (ECM) in the glomerulus mainly produced by mesangial cells in response to transforming growth factor-ß (TGF-ß) stimuli under diabetic conditions. Despite TGF-ß has been implicated as a major pathogenic factor in the development of diabetic glomerulosclerosis, clinical trials of monoclonal antibodies against TGF-ß failed to demonstrate therapeutic benefits. Thus, developing alternative therapeutic strategies to effectively block the TGF-ß/Smad signaling could be of paramount importance for DKD treatment. Emerging evidence indicates that dysregulation of certain lncRNAs can lead to aberrant activation of TGF-ß/Smad signaling. Herein, we identified a novel lncRNA, named DANCR, which could efficiently function as a negative regulator of TGF-ß/Smad signaling in mesangial cells. Ectopic expression of DANCR could specifically block the activation of TGF-ß/Smad signaling induced by high-glucose or TGF-ß in human renal mesangial cells (HRMCs). Mechanistically, DANCR functions to stabilize nemo-like kinase (NLK) mRNA through interaction with insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2), resulting in enhanced phosphorylating on the linker region of activated Smad2/3 in the nucleus. Taken together, our data have uncovered an lncRNA-based regulatory modality of the TGF-ß/Smad signaling and identified DANCR as an endogenous blocker of TGF-ß/Smad signaling in HRMCs, which may represent a potential therapeutic target against the diabetic glomerulosclerosis.


Assuntos
Nefropatias Diabéticas , RNA Longo não Codificante , Humanos , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Matriz Extracelular/metabolismo , Mesângio Glomerular/metabolismo , Glucose/farmacologia , Glucose/metabolismo , Células Mesangiais/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Smad/metabolismo
4.
Adv Exp Med Biol ; 1445: 101-117, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38967753

RESUMO

The urinary system comprises kidneys, ureters, bladder, and urethra with its primary function being excretion, referring to the physiological process of transporting substances that are harmful or surplus out of the body. The male reproductive system consists of gonads (testis), vas deferens, and accessory glands such as the prostate. According to classical immunology theory, the tissues and organs mentioned above are not thought to produce immunoglobulins (Igs), and any Ig present in the relevant tissues under physiological and pathological conditions is believed to be derived from B cells. For instance, most renal diseases are associated with uncontrolled inflammation caused by pathogenic Ig deposited in the kidney. Generally, these pathological Igs are presumed to be produced by B cells. Recent studies have demonstrated that renal parenchymal cells can produce and secrete Igs, including IgA and IgG. Glomerular mesangial cells can express and secrete IgA, which is associated with cell survival and adhesion. Likewise, human podocytes demonstrate the ability to produce and secrete IgG, which is related to cell survival and adhesion. Furthermore, renal tubular epithelial cells also express IgG, potentially involved in the epithelial-mesenchymal transition (EMT). More significantly, renal cell carcinoma, bladder cancer, and prostate cancer have been revealed to express high levels of IgG, which promotes tumour progression. Given the widespread Ig expression in the urinary and male reproductive systems, continued efforts to elucidate the roles of Igs in renal physiological and pathological processes are necessary.


Assuntos
Imunoglobulinas , Humanos , Masculino , Imunoglobulinas/metabolismo , Imunoglobulinas/genética , Imunoglobulinas/imunologia , Sistema Urinário/imunologia , Sistema Urinário/metabolismo , Sistema Urinário/patologia , Genitália Masculina/imunologia , Genitália Masculina/metabolismo , Genitália Masculina/patologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Imunoglobulina G/imunologia , Relevância Clínica
5.
Am J Physiol Renal Physiol ; 324(1): F124-F134, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36417276

RESUMO

Although mesangial cell-glomerular basement membrane (GBM) connections play a key role in maintaining the glomerular capillary loop structure, information remains limited about how these connections are formed during glomerulogenesis. We have previously shown that weakened podocyte-GBM interactions owing to tensin 2 (Tns2) deficiency lead to abnormal GBM maturation during postnatal glomerulogenesis. Here, we investigated whether abnormal GBM maturation affected mesangial cell-GBM connections and mesangial cell differentiation. Histological analysis of the outer cortical glomeruli in Tns2-deficient mice revealed that GBM materials overproduced by stressed immature podocytes accumulated in the mesangium and interrupted the formation of mesangial cell-GBM connections, resulting in fewer capillary loops compared with that of normal glomeruli. In addition, expression of α-smooth muscle actin, an immature mesangial cell marker, persisted in mesangial cells of Tns2-deficient outer cortical glomeruli even after glomerulogenesis was completed, resulting in mesangial expansion. Furthermore, analysis of mouse primary mesangial cells revealed that mesangial cell differentiation depended on the type of extracellular matrix components to which the cells adhered, suggesting the participation of mesangial cell-GBM connections in mesangial cell differentiation. These findings suggest that abnormal GBM maturation affects mesangial cell differentiation by impairing mesangial cell-GBM connections.NEW & NOTEWORTHY Mesangial cell-glomerular basement membrane (GBM) connections play an important role in maintaining the structural integrity of the glomerular tuft. However, information remains scarce about how GBM maturation affects the formation of these connections during glomerular development. Here, we show that abnormal GBM maturation due to tensin 2 deficiency affects mesangial cell differentiation by impairing mesangial cell-GBM connections during postnatal glomerulogenesis.


Assuntos
Membrana Basal Glomerular , Podócitos , Camundongos , Animais , Membrana Basal/metabolismo , Tensinas , Mesângio Glomerular , Podócitos/metabolismo , Diferenciação Celular
6.
Clin Immunol ; 257: 109840, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37939913

RESUMO

IgA nephropathy (IgAN) is an essential cause of kidney failure and end-stage kidney disease worldwide. Mesangial hypercellularity is an important characteristic of IgAN, but the underlying mechanism remains unclear. Endoplasmic reticulum (ER) stress is a series of stress responses to restore the function of endoplasmic reticulum. We aimed to explore how ER stress functioned in kidneys of IgAN. We first examined ER stress in IgAN kidneys in vivo and in vitro, by testing the levels of ER stress associated proteins (BIP, p-eIF2α and ATF4). Our results showed that ER stress was activated in IgAN patients, mice and cell model. ER stress activation was related to the distribution of IgA deposition and the degree of mesangial proliferation. To determine the role of ER stress in mesangial cell (MC) proliferation of IgAN, we then tested the levels of ER stress and MC proliferation (cyclin D1, cell viability and cell cycle) through inhibiting ER stress associated proteins. After inhibiting ER stress associated proteins, ER stress was inactivated and cell proliferation was inhibited in MCs. We also explored the correlation between ER stress in the glomerulus and the clinical outcomes of IgAN patients in a prospective study. Patients with lower expression of p-eIF2α or ATF4 had higher rates of hematuria remission, proteinuria remission and clinical remission. In summary, our work outlines that in IgAN, ER stress mediated by eIF2α/ATF4 pathway promotes MC proliferation via up-regulating the expression of cyclin D1. Furthermore, p-eIF2α and ATF4 in the glomerulus negatively correlate with the clinical remission of IgAN patients.


Assuntos
Glomerulonefrite por IGA , Células Mesangiais , Animais , Humanos , Camundongos , Fator 4 Ativador da Transcrição/metabolismo , Proliferação de Células , Ciclina D1/metabolismo , Estresse do Retículo Endoplasmático , Glomerulonefrite por IGA/metabolismo , Células Mesangiais/metabolismo , Estudos Prospectivos , Transdução de Sinais
7.
Cell Commun Signal ; 21(1): 10, 2023 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-36639674

RESUMO

BACKGROUND: Diabetic nephropathy (DN) is an increasing threat to human health and regarded to be the leading cause of end-stage renal disease worldwide. Exosomes delivery may play a key role in cross-talk among kidney cells and the progression of DN. However, the mechanisms underlying exosomes in DN remain unclear. METHODS: The cross-disciplinary study, including in vivo, in vitro, and human studies was conducted to explore the cross-talk between proximal tubular epithelial cells (PTECs) and mesangial cells (MCs) in DN. We purified exosome from PTECs treated with high glucose and db/db mice and assessed their influences in the pathologic change of MCs and downstream signal pathway. Healthy individuals and type 2 diabetic patients were enrolled to examine the role of exosomes in clinical applications. RESULTS: High glucose stimulated PTECs to secrete exosomal miR-92a-1-5p, which was taken-up by glomerular MCs, inducing myofibroblast transdifferentiation (MFT) in vitro and in vivo. PTEC-released exosomal 92a-1-5p decreased reticulocalbin-3 expression, leading to endoplasmic reticulum (ER) stress by downregulating genes essential for ER homeostasis including calreticulin and mesencephalic astrocyte-derived neurotrophic factor. Treatment with miR-92a-1-5p inhibitor ameliorated kidney damage in db/db mice with DN. Urinary miR-92a-1-5p could predict kidney injury in type 2 diabetic patients. CONCLUSIONS: PTEC-derived exosomal miR-92a-1-5p modulated the kidney microenvironment in vivo and in vitro models, which altered ER stress and MFT in MCs resulting in DN progression. Further blocking miR-92a-1-5p epigenetic regulatory network could be a potential therapeutic strategy to prevent the progression of DN. Video Abstract.


Diabetic nephropathy (DN) has been the leading cause of end-stage renal disease worldwide. Exosomes play a principle role in cross-talk of kidney cells and further affect the onset or progression of DN. This study firstly demonstrated the communication between proximal tubular epithelial cells (PTECs) and mesangial cells (MCs) through exosome transmission. PTEC-released exosomal 92a-1-5p induced endoplasmic reticulum stress and epithelial-mesenchymal transition in MCs through reticulocalbin-3 modulation. Kidney damage was rescued in DN mice after treatment with miR-92a-1-5p inhibitor. Moreover, urinary exosomal miR-92a-1-5p could predict DN progression in type 2 diabetic patients. These findings prove the impact of exosomal miR-92a-1-5p on pathophysiologic mechanisms and its potential use in clinical care and prediction of DN.


Assuntos
Diabetes Mellitus Tipo 2 , Nefropatias Diabéticas , Exossomos , MicroRNAs , Animais , Humanos , Camundongos , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Nefropatias Diabéticas/metabolismo , Exossomos/metabolismo , Glucose/metabolismo , Células Mesangiais/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo
8.
Nephrol Dial Transplant ; 38(Supplement_2): ii3-ii10, 2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-37218714

RESUMO

Glomerulonephritis (GN) is a diverse group of immune-mediated disorders. Currently, GN is classified largely by histological patterns that are difficult to understand and teach, and most importantly, do not indicate treatment choices. Indeed, altered systemic immunity is the primary pathogenic process and the key therapeutic target in GN. Here, we apply a conceptual framework of immune-mediated disorders to GN guided by immunopathogenesis and hence immunophenotyping: (i) infection-related GN require pathogen identification and control; (ii) autoimmunity-related GN, defined by presence of autoantibodies and (iii) alloimmunity-related GN in transplant recipients both require the suppression of adaptive immunity in lymphoid organs and bone marrow; (iv) autoinflammation-related GN, e.g. inborn errors of immunity diagnosed by genetic testing, requires suppression of single cytokine or complement pathways; and (v) Monoclonal gammopathy-related GN requires B or plasma cell clone-directed therapy. A new GN classification should include disease category, immunological activity to tailor the use of the increasing number of immunomodulatory drugs, and chronicity to trigger standard chronic kidney disease care including the evolving spectrum of cardio-renoprotective drugs. Certain biomarkers allow diagnosis and the assessment of immunological activity and disease chronicity without kidney biopsy. The use of these five GN categories and a therapy-focused GN classification is likely to overcome some of the existing hurdles in GN research, management and teaching by reflecting disease pathogenesis and guiding the therapeutic approach.


Assuntos
Glomerulonefrite , Insuficiência Renal Crônica , Humanos , Glomerulonefrite/diagnóstico , Glomerulonefrite/etiologia , Glomerulonefrite/terapia , Biomarcadores , Autoanticorpos , Nefrectomia
9.
Int J Mol Sci ; 24(22)2023 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-38003679

RESUMO

Lupus nephritis (LN) is a serious complication for many patients who develop systemic lupus erythematosus, which primarily afflicts women. Our studies to identify biomarkers and the pathogenic mechanisms underlying LN will provide a better understanding of disease progression and sex bias, and lead to identification of additional potential therapeutic targets. The glycosphingolipid lactosylceramide (LacCer) and N-linked glycosylated proteins (N-glycans) were measured in urine and serum collected from LN and healthy control (HC) subjects (10 females and 10 males in each group). The sera from the LN and HC subjects were used to stimulate cytokine secretion and intracellular Ca2+ flux in female- and male-derived primary human renal mesangial cells (hRMCs). Significant differences were observed in the urine of LN patients compared to HCs. All major LacCers species were significantly elevated and differences between LN and HC were more pronounced in males. 72 individual N-glycans were altered in LN compared to HC and three N-glycans were significantly different between the sexes. In hRMCs, Ca2+ flux, but not cytokine secretion, was higher in response to LN sera compared to HC sera. Ca2+ flux, cytokine secretion, and glycosphingolipid levels were significantly higher in female-derived compared to male-derived hRMCs. Relative abundance of some LacCers and hexosylceramides were higher in female-derived compared to male-derived hRMCs. Urine LacCers and N-glycome could serve as definitive LN biomarkers and likely reflect renal disease activity. Despite higher sensitivity of female hRMCs, males may experience greater increases in LacCers, which may underscore worse disease in males. Elevated glycosphingolipid metabolism may poise renal cells to be more sensitive to external stimuli.


Assuntos
Lúpus Eritematoso Sistêmico , Nefrite Lúpica , Humanos , Feminino , Masculino , Nefrite Lúpica/patologia , Biomarcadores , Citocinas , Glicoesfingolipídeos , Polissacarídeos
10.
Cell Tissue Res ; 390(1): 93-112, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35821438

RESUMO

Glomerular mesangial cell proliferation and extracellular matrix accumulation contribute to the progression of diabetic nephropathy (DN). As a conserved stress-inducible protein, sestrin2 (Sesn2) plays critical role in the regulation of oxidative stress, inflammation, autophagy, metabolism, and endoplasmic reticulum stress. In this study, we investigated the role of Sesn2 on renal damage in diabetic kidney using transgenic mice overexpressing Sesn2 and the effect of Sesn2 on mesangial cell proliferation and extracellular matrix accumulation in diabetic conditions and the possible molecular mechanisms involved. Sesn2 overexpression improved renal function and decreased glomerular hypertrophy, albuminuria, mesangial expansion, extracellular matrix accumulation, and TGF-ß1 expression, as well as oxidative stress in diabetic mice. In vitro experiments, using human mesangial cells (HMCs), revealed that Sesn2 overexpression inhibited high glucose (HG)-induced proliferation, fibronectin and collagen IV production, and ROS generation. Meanwhile, Sesn2 overexpression restored phosphorylation levels of Lats1 and YAP and inhibited TEAD1 expression. Inhibition of Lats1 accelerated HG-induced proliferation and expression of fibronectin and collagen IV. Verteporfin, an inhibitor of YAP, suppressed HG-induced proliferation and expression of fibronectin and collagen IV. However, Sesn2 overexpression reversed Lats1 deficiency-induced Lats1 and YAP phosphorylation, nuclear expression levels of YAP and TEAD1, and proliferation and fibronectin and collagen IV expressions in HMCs exposed to HG. In addition, antioxidant NAC or tempol treatment promoted phosphorylation of Lats1 and YAP and inhibited TEAD1 expression, proliferation, and fibronectin and collagen IV accumulation in HG-treated HMCs. Taken together, Sesn2 overexpression inhibited mesangial cell proliferation and fibrosis via regulating Hippo pathway in diabetic nephropathy. Induction of Sesn2 may be a potential therapeutic target in diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Animais , Antioxidantes/farmacologia , Colágeno Tipo IV/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Glucose/metabolismo , Via de Sinalização Hippo , Humanos , Rim/metabolismo , Camundongos , Proteínas Nucleares , Proteínas Serina-Treonina Quinases , Espécies Reativas de Oxigênio/metabolismo , Sestrinas , Fator de Crescimento Transformador beta1/metabolismo , Verteporfina/metabolismo , Verteporfina/farmacologia , Verteporfina/uso terapêutico
11.
FASEB J ; 35(7): e21729, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34143493

RESUMO

Glomerular mesangial cell (GMC)-derived pleiotropic cytokine, interleukin-1 (IL-1), contributes to hypercellularity in human and experimental proliferative glomerulonephritis. IL-1 promotes mesangial proliferation and may stimulate extracellular matrix accumulation, mechanisms of which are unclear. The present study shows that the beta isoform of IL-1 (IL-1ß) is a potent inducer of IL-1 type I receptor-dependent Ca2+ entry in mouse GMCs. We also demonstrate that the transient receptor potential ankyrin 1 (TRPA1) is an intracellular store-independent diacylglycerol-sensitive Ca2+ channel in the cells. IL-1ß-induced Ca2+ and Ba2+ influxes in the cells were negated by pharmacological inhibition and siRNA-mediated knockdown of TRPA1 channels. IL-1ß did not stimulate fibronectin production in cultured mouse GMCs and glomerular explants but promoted Ca2+ -dependent cell proliferation. IL-1ß also stimulated TRPA1-dependent ERK mitogen-activated protein kinase (MAPK) phosphorylation in the cells. Concomitantly, IL-1ß-induced GMC proliferation was attenuated by TRPA1 and RAF1/ MEK/ERK inhibitors. These findings suggest that IL-1ß-induced Ca2+ entry via TRPA1 channels engenders MAPK-dependent mesangial cell proliferation. Hence, TRPA1-mediated Ca2+ signaling could be of pathological significance in proliferative glomerulonephritis.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Proliferação de Células/fisiologia , Interleucina-1beta/metabolismo , Células Mesangiais/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Canal de Cátion TRPA1/metabolismo , Animais , Células Cultivadas , Camundongos , Fosforilação/fisiologia
12.
BMC Nephrol ; 23(1): 211, 2022 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-35710406

RESUMO

BACKGROUND: T-type calcium channels (TTCC) are low voltage activated channels that are widely expressed in the heart, smooth muscle and neurons. They are known to impact on cell cycle progression in cancer and smooth muscle cells and more recently, have been implicated in rat and human mesangial cell proliferation. The aim of this study was to investigate the roles of the different isoforms of TTCC in mouse mesangial cells to establish which may be the best therapeutic target for treating mesangioproliferative kidney diseases.  METHODS: In this study, we generated single and double knockout (SKO and DKO) clones of the TTCC isoforms CaV3.1 and CaV3.2 in mouse mesangial cells using CRISPR-cas9 gene editing. The downstream signals linked to this channel activity were studied by ERK1/2 phosphorylation assays in serum, PDGF and TGF-ß1 stimulated cells. We also examined their proliferative responses in the presence of the TTCC inhibitors mibefradil and TH1177. RESULTS: We demonstrate a complete loss of ERK1/2 phosphorylation in response to multiple stimuli (serum, PDGF, TGF-ß1) in CaV3.1 SKO clone, whereas the CaV3.2 SKO clone retained these phospho-ERK1/2 responses. Stimulated cell proliferation was not profoundly impacted in either SKO clone and both clones remained sensitive to non-selective TTCC blockers, suggesting a role for more than one TTCC isoform in cell cycle progression. Deletion of both the isoforms resulted in cell death. CONCLUSION: This study confirms that TTCC are expressed in mouse mesangial cells and that they play a role in cell proliferation. Whereas the CaV3.1 isoform is required for stimulated phosphorylation of ERK1/2, the Ca V3.2 isoform is not. Our data also suggest that neither isoform is necessary for cell proliferation and that the anti-proliferative effects of mibefradil and TH1177 are not isoform-specific. These findings are consistent with data from in vivo rat mesangial proliferation Thy1 models and support the future use of genetic mouse models to test the therapeutic actions of TTCC inhibitors.


Assuntos
Canais de Cálcio Tipo T , Células Mesangiais , Animais , Humanos , Células Mesangiais/metabolismo , Mibefradil/metabolismo , Mibefradil/farmacologia , Camundongos , Fosforilação , Ratos , Fator de Crescimento Transformador beta1/metabolismo
13.
J Enzyme Inhib Med Chem ; 37(1): 178-188, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34894983

RESUMO

Diabetic nephropathy (DN) is one of the severe microvascular complications of diabetes mellitus. Oxidative stress resulting from aberrant metabolism of glucose mediates renal inflammation and fibrosis in the progression of DN. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor regulating the expression of antioxidant enzymes. Activating Nrf2 will give a promising therapy for DN. To discover novel Nrf2 activators, we have investigated caffeoylisocitric acid using mesangial cells under high glucose. The results showed at 10 µM, caffeoylisocitric acid significantly inhibited the self-limited proliferation of mesangial cells induced by high glucose. Further assessments have disclosed caffeoylisocitric acid mitigated oxidative stress, inflammation and accumulation of extracellular matrix resulting from high glucose via inactivating MAPK signalling. Meanwhile activation of Nrf2 was observed and involved in these effects through the interaction between Keap1 and caffeoylisocitric acid to disrupt Keap1-Nrf2 complex. Therefore, caffeoylisocitric acid is a promising Nrf2 activator targeting DN.


Assuntos
Ácidos Cafeicos/farmacologia , Nefropatias Diabéticas/tratamento farmacológico , Descoberta de Drogas , Glucose/antagonistas & inibidores , Hipoglicemiantes/farmacologia , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Células Mesangiais/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Ácidos Cafeicos/química , Células Cultivadas , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Relação Dose-Resposta a Droga , Matriz Extracelular/efeitos dos fármacos , Glucose/metabolismo , Humanos , Hipoglicemiantes/química , Células Mesangiais/metabolismo , Estrutura Molecular , Estresse Oxidativo/efeitos dos fármacos , Relação Estrutura-Atividade
14.
Mar Drugs ; 20(6)2022 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-35736158

RESUMO

Advanced glycation end-products (AGEs) play a vital role in the pathogenesis of diabetic complications. Methylglyoxal (MGO), one of the major precursors of AGEs, is a highly reactive dicarbonyl compound that plays an important role in the pathogenesis of diabetic nephropathy. This study was designed to evaluate the therapeutic potential of phlorotannin-rich Ecklonia cava extract (ECE) on MGO-induced diabetic nephropathy in in vitro models using mouse glomerular mesangial cells. ECE showed anti-glycation activity via breaking of AGEs-collagen cross-links and inhibition of AGEs formation and AGE-collagen cross-linking formation. The renoprotective effects were determined by assessing intracellular reactive oxygen species (ROS) and MGO accumulation, cell apoptosis, and the Nrf-2/ARE signaling pathway. MGO-induced renal damage, intracellular ROS production level, and MGO-protein adduct accumulation were significantly decreased by pretreating ECE. Moreover, ECE pretreatment exhibited preventive properties against MGO-induced dicarbonyl stress via activation of the Nrf2/ARE signaling pathway and reduction of RAGE protein expression in mouse glomerular mesangial cells. Collectively, these results indicated potential anti-glycation properties and prominent preventive effects of ECE against MGO-induced renal damage. Additionally, ECE may be utilized for the management of AGE-related diabetic nephropathy.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Animais , Nefropatias Diabéticas/induzido quimicamente , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Óxido de Magnésio , Camundongos , Aldeído Pirúvico/toxicidade , Espécies Reativas de Oxigênio/metabolismo
15.
J Cell Mol Med ; 25(12): 5691-5706, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33949114

RESUMO

Mesangial cell (MC) proliferation is a key pathological feature in a number of common human renal diseases, including mesangial proliferative nephritis and diabetic nephropathies. Knowledge of MC responses to pathological stimuli is crucial to the understanding of these disease processes. We previously determined that Krϋppel-like factor 15 (KLF15), a kidney-enriched zinc-finger transcription factor, was required for inhibition of MC proliferation. In the present study, we investigated the direct target gene and the underlying mechanism by which KLF15 regulated mesangial proliferation. First, we screened small ubiquitin-related modifier 1 (SUMO1) as the direct transcriptional target of KLF15 and validated this finding with ChIP-PCR and luciferase assays. Furthermore, we demonstrated that overexpressing KLF15 or SUMO1 enhanced the stability of P53, which blocked the cell cycle of human renal MCs (HRMCs) and therefore abolished cell proliferation. Conversely, knockdown of SUMO1 in HRMCs, even those overexpressed with KLF15, could not inhibit HRMC proliferation rates and increase SUMOylation of P53. Finally, the results showed that the levels of SUMOylated P53 in the kidney cortices of anti-Thy 1 model rats were decreased during proliferation periods. These findings reveal the critical mechanism by which KLF15 targets SUMO1 to mediate the proliferation of MCs.


Assuntos
Proliferação de Células , Glomerulonefrite/patologia , Glomérulos Renais/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Células Mesangiais/patologia , Proteína SUMO-1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica , Glomerulonefrite/metabolismo , Glomérulos Renais/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Masculino , Células Mesangiais/metabolismo , Ratos , Ratos Wistar , Proteína SUMO-1/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/genética
16.
Immunology ; 162(4): 418-433, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33314123

RESUMO

Previously, we demonstrated neuraminidase (NEU) activity or NEU1 expression, specifically, is increased in the kidneys of lupus mice and urine of human patients with nephritis. Additionally, NEU activity mediates IL-6 secretion from lupus-prone MRL/lpr primary mouse mesangial cells (MCs) in response to an IgG mimic. IL-6 mediates glomerular inflammation and promotes tissue damage in patients and mouse strains with lupus nephritis. This study further elucidates the mechanisms by which NEU activity and NEU1 specifically mediates the release of IL-6 and other cytokines from lupus-prone MCs. We demonstrate significantly increased release of multiple cytokines and NEU activity in MRL/lpr MCs in response to serum from MRL/lpr mice (lupus serum). Inhibiting NEU activity significantly reduced secretion of three of those cytokines: IL-6, GM-CSF and MIP1α. Message levels of Il-6 and Gm-csf were also increased in response to lupus serum and reduced when NEU activity was inhibited. Neutralizing antibodies to cell-surface receptors and MAPK inhibitors in lupus serum- or LPS-stimulated MCs indicate TLR4 and p38 or ERK MAP kinase signalling play key roles in the NEU-mediated secretion of IL-6. Significantly reduced IL-6 release was observed in C57BL/6 (B6) Neu1+/+ primary MCs compared with wild-type (Neu1+/+) B6 MCs in response to lupus serum. Additional results show inhibiting NEU activity significantly increases sialic acid-containing N-glycan levels. Together, our novel observations support a role for NEU activity, and specifically NEU1, in mediating release of IL-6 from lupus-prone MCs in response to lupus serum through a TLR4-p38/ERK MAPK signalling pathway that likely includes desialylation of glycoproteins.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Rim/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Nefrite Lúpica/metabolismo , Células Mesangiais/fisiologia , Neuraminidase/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Neuraminidase/genética , Soro/metabolismo , Transdução de Sinais
17.
Cell Immunol ; 361: 104279, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33422698

RESUMO

Mesangial cells (MCs), as resident cells of the kidneys, play an important role in maintaining glomerular function. MCs are located between the capillary loops of the glomeruli and mainly support the capillary plexus, constrict blood vessels, extracellular matrix components, produce cytokines, and perform phagocytosis and clearance of macromolecular substances. When the glomerular environment changes, MCs are often affected, which can lead to functional transformation. The immune response is involved in the occurrence and development of various kidney diseases, in these diseases, antigen-presenting cells (APCs) play an important role. APCs can present antigens to T lymphocytes, causing them to become activated and proliferate. Studies have shown that MCs have phagocytic function and express APC markers on the cell surface. Additionally, MCs are stimulated by or produce various inflammatory factors to participate in the renal inflammatory response. Therefore, MCs have potential antigen presentation function and participate in the pathological changes of various kidney diseases as APCs upon activation. In this paper, by reviewing MC phagocytic function, activated MC expression of APC surface markers, and MC participation in the inflammatory response and local renal immune response, we confirm that activated MCs can act as APCs in renal disease.


Assuntos
Células Mesangiais/imunologia , Células Mesangiais/metabolismo , Células Mesangiais/fisiologia , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Cultivadas , Citocinas/metabolismo , Humanos , Inflamação , Rim/imunologia , Nefropatias/metabolismo , Fagocitose/imunologia , Linfócitos T
18.
Cell Tissue Res ; 385(2): 371-392, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33433692

RESUMO

The lysosome represents an important regulatory platform within numerous vesicle trafficking pathways including the endocytic, phagocytic, and autophagic pathways. Its ability to fuse with endosomes, phagosomes, and autophagosomes enables the lysosome to break down a wide range of both endogenous and exogenous cargo, including macromolecules, certain pathogens, and old or damaged organelles. Due to its center position in an intricate network of trafficking events, the lysosome has emerged as a central signaling node for sensing and orchestrating the cells metabolism and immune response, for inter-organelle and inter-cellular signaling and in membrane repair. This review highlights the current knowledge of general lysosome function and discusses these findings in their implication for renal glomerular cell types in health and disease including the involvement of glomerular cells in lysosomal storage diseases and the role of lysosomes in nongenetic glomerular injuries.


Assuntos
Glomérulos Renais/patologia , Lisossomos/patologia , Animais , Humanos
19.
Mol Ther ; 28(3): 963-974, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-31991106

RESUMO

Diabetic nephropathy (DN) is an increasing threat to human health and is regarded as an important public issue. The pathophysiologic mechanisms of DN are complicated. The initiating molecular events triggering the loss function in mesangial cells (MCs) in DN are not well known. In this cross-disciplinary study, transcriptome analysis of high glucose (HG)-treated mouse MCs (MMCs) using next-generation sequencing and systematic bioinformatics analyses indicated that miR-15b-5p and its downstream target B cell lymphoma 2 (BCL-2) contribute to HG-induced apoptosis in MMCs. HG elevated miR-15b-5p expression, which in turn decreased the translation of BCL-2, leading to MMC apoptosis under HG. Apoptosis of MCs was enhanced in the presence of extracellular vesicles isolated from the urine of type 2 diabetic patients with high levels of miR-15b-5p. Furthermore, increased levels of urinary miR-15b-5p were found in db/db mice and type 2 diabetic patients, and such levels correlated with low baseline kidney function and rapid decline in kidney function during a mean of follow-up period of 2.4 ± 0.1 years. Therefore, miR-15b-5p induced mesangial cells apoptosis by targeting BCL-2 under HG. miR-15b-5p has the potential to predict kidney injury in DN. Blocking the miR-15b-5p epigenetic regulatory network could be a potential therapeutic strategy to prevent mesangial apoptosis in DN.


Assuntos
Apoptose/genética , Glicemia , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/metabolismo , Vesículas Extracelulares/metabolismo , Glucose/metabolismo , Células Mesangiais/metabolismo , MicroRNAs/genética , Animais , Transporte Biológico , Biomarcadores , Linhagem Celular , Nefropatias Diabéticas/patologia , Suscetibilidade a Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genes bcl-2 , Humanos , Imuno-Histoquímica , Imunofenotipagem , Células Mesangiais/patologia , Camundongos , Modelos Biológicos , Interferência de RNA
20.
Endocr J ; 68(7): 751-762, 2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-33790061

RESUMO

Rosiglitazone (RSG) is widely used to reduce the amount of sugar in the blood of patients with diabetes mellitus. Diabetic nephropathy is the most common microvascular complication of diabetes. The role of RSG in diabetic nephropathy is not fully understood. Diabetic nephropathy model was constructed in high glucose (HG)-treated mouse mesangial cells. The effects of RSG on cell viability and cell cycle were investigated using cell counting kit-8 (CCK-8) assay and flow cytometry assay. Oxidative stress was assessed according to ROS production and SOD activity in cells. Inflammatory responses were assessed according to the releases of inflammatory cytokines. Extracellular matrix (ECM) accumulation was determined by the levels of fibronectin and collagen IV using western blot. The expression of Gm26917 and microRNA-185-5p (miR-185-5p) was detected by quantitative real-time polymerase chain reaction (qPCR). The interaction between Gm26917 and miR-185-5p was validated by dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and pull-down assay. RSG significantly inhibited HG-induced proliferation, oxidative stress, inflammatory responses and ECM accumulation in mouse mesangial cells. The expression of Gm26917 was induced by HG but weakened by RSG. Gm26917 knockdown alleviated HG-induced proliferation, oxidative stress, inflammatory responses and ECM accumulation in mouse mesangial cells, and Gm26917 overexpression partly abolished the effects of RSG. Moreover, miR-185-5p was a target of Gm26917, and miR-185-5p inhibition recovered proliferation, oxidative stress, inflammatory responses and ECM accumulation in mouse mesangial cells that were alleviated by Gm26917 knockdown. RSG ameliorated HG-induced mouse mesangial cell proliferation, oxidative stress, inflammation and ECM accumulation partially by governing the Gm26917/miR-185-5p pathway.


Assuntos
Proliferação de Células/efeitos dos fármacos , Matriz Extracelular/metabolismo , Glucose/farmacologia , Hipoglicemiantes/farmacologia , Inflamação/metabolismo , Células Mesangiais/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Rosiglitazona/farmacologia , Animais , Células Mesangiais/metabolismo , Camundongos , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA