Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 11850, 2024 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-38782980

RESUMO

Natriuretic peptides (NPs) are cardio-derived hormones that have a crucial role in maintaining cardiovascular homeostasis. Physiological effects of NPs are mediated by binding to natriuretic peptide receptors 1 and 2 (NPR1/2), whereas natriuretic peptide receptor 3 (NPR3) acts as a clearance receptor that removes NPs from the circulation. Mouse studies have shown that local NP-signaling in the kidney glomerulus is important for the maintenance of renal homeostasis. In this study we examined the expression of NPR3 in kidney tissue and explored its involvement in renal physiology and disease by generating podocyte-specific knockout mice (NPR3podKO) as well as by using an NPR3 inhibitor (NPR3i) in rodent models of kidney disease. NPR3 was highly expressed by podocytes. NPR3podKO animals showed no renal abnormalities under healthy conditions and responded similarly to nephrotoxic serum (NTS) induced glomerular injury. However, NPR3i showed reno-protective effects in the NTS-induced model evidenced by decreased glomerulosclerosis and reduced podocyte loss. In a ZSF1 rat model of diabetic kidney injury, therapy alone with NPR3i did not have beneficial effects on renal function/histology, but when combined with losartan (angiotensin receptor blocker), NPR3i potentiated its ameliorative effects on albuminuria. In conclusion, these results suggest that NPR3 may contribute to kidney disease progression.


Assuntos
Camundongos Knockout , Podócitos , Receptores do Fator Natriurético Atrial , Animais , Receptores do Fator Natriurético Atrial/metabolismo , Receptores do Fator Natriurético Atrial/genética , Camundongos , Podócitos/metabolismo , Podócitos/patologia , Ratos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Modelos Animais de Doenças , Nefropatias/metabolismo , Nefropatias/patologia , Losartan/farmacologia , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia
2.
J Med Chem ; 67(6): 4442-4462, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38502780

RESUMO

Relaxin H2 is a clinically relevant peptide agonist for relaxin family peptide receptor 1 (RXFP1), but a combination of this hormone's short plasma half-life and the need for injectable delivery limits its therapeutic potential. We sought to overcome these limitations through the development of a potent small molecule (SM) RXFP1 agonist. Although two large SM HTS campaigns failed in identifying suitable hit series, we uncovered novel chemical space starting from the only known SM RXFP1 agonist series, represented by ML290. Following a design-make-test-analyze strategy based on improving early dose to man ranking, we discovered compound 42 (AZ7976), a highly selective RXFP1 agonist with sub-nanomolar potency. We used AZ7976, its 10 000-fold less potent enantiomer 43 and recombinant relaxin H2 to evaluate in vivo pharmacology and demonstrate that AZ7976-mediated heart rate increase in rats was a result of RXFP1 agonism. As a result, AZ7976 was selected as lead for continued optimization.


Assuntos
Relaxina , Humanos , Masculino , Ratos , Animais , Relaxina/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores de Peptídeos/agonistas
3.
J Med Chem ; 67(6): 4419-4441, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38502782

RESUMO

Optimization of the highly potent and selective, yet metabolically unstable and poorly soluble hRXFP1 agonist AZ7976 led to the identification of the clinical candidate, AZD5462. Assessment of RXFP1-dependent cell signaling demonstrated that AZD5462 activates a highly similar panel of downstream pathways as relaxin H2 but does not modulate relaxin H2-mediated cAMP second messenger responsiveness. The therapeutic potential of AZD5462 was assessed in a translatable cynomolgus monkey heart failure model. Following 8 weeks of treatment with AZD5462, robust improvements in functional cardiac parameters including LVEF were observed at weeks 9, 13, and 17 without changes in heart rate or mean arterial blood pressure. AZD5462 was well tolerated in both rat and cynomolgus monkey and has successfully completed phase I studies in healthy volunteers. In summary, AZD5462 is a small molecule pharmacological mimetic of relaxin H2 signaling at RXFP1 and holds promise as a potential therapeutic approach to treat heart failure patients.


Assuntos
Insuficiência Cardíaca , Relaxina , Humanos , Ratos , Animais , Relaxina/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Macaca fascicularis/metabolismo , Receptores de Peptídeos/metabolismo , Insuficiência Cardíaca/tratamento farmacológico
4.
iScience ; 26(6): 106830, 2023 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-37250770

RESUMO

Apolipoprotein L1 (APOL1) high-risk genotypes are associated with increased risk of chronic kidney disease (CKD) in people of West African ancestry. Given the importance of endothelial cells (ECs) in CKD, we hypothesized that APOL1 high-risk genotypes may contribute to disease via EC-intrinsic activation and dysfunction. Single cell RNA sequencing (scRNA-seq) analysis of the Kidney Precision Medicine Project dataset revealed APOL1 expression in ECs from various renal vascular compartments. Utilizing two public transcriptomic datasets of kidney tissue from African Americans with CKD and a dataset of APOL1-expressing transgenic mice, we identified an EC activation signature; specifically, increased intercellular adhesion molecule 1 (ICAM-1) expression and enrichment in leukocyte migration pathways. In vitro, APOL1 expression in ECs derived from genetically modified human induced pluripotent stem cells and glomerular ECs triggered changes in ICAM-1 and platelet endothelial cell adhesion molecule 1 (PECAM-1) leading to an increase in monocyte attachment. Overall, our data suggest the involvement of APOL1 as an inducer of EC activation in multiple renal vascular beds with potential effects beyond the glomerular vasculature.

5.
Kidney Int ; 101(4): 752-765, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34968552

RESUMO

IgA nephropathy (IgAN) is the most common primary glomerulonephritis worldwide and defined by the presence of IgA-containing immune complexes in the mesangium that induce an inflammation leading to glomerulonephritis. Since we poorly understand early mechanisms of glomerular injury in IgAN we performed single-cell RNA sequencing (scRNA-seq) analysis of glomerulus-associated cells using SMARTseq2-technology at the early stage of IgAN in grouped ddY-mice. Cell-specific molecular signatures unraveled a key role of endothelial cells in the early pathogenesis of IgAN, especially in the recruitment and infiltration of immune cells. Mesangial and podocyte cells demonstrated less molecular changes. Several intra-glomerular paracrine pathways were detected, such as mesangial cell-derived Slit3 potentially activating Robo-receptors in podocyte/endothelial cells. Surprisingly, proximal tubular cells were strongly affected at the early stage and potential glomerulo-tubular cell-cell crosstalk pathways were identified. Importantly, many of the cellular transcriptomic signatures identified in this well-established mouse model were also detected in published bulk transcriptomic data in human IgAN. Moreover, we validated the functionality of key cell-cell crosstalk pathways using cell culture models, such as the effect of the Slit-Robo signalling axis. Thus, our study provides important novel molecular insights into the pathogenesis of early IgAN-associated glomerulopathy.


Assuntos
Glomerulonefrite por IGA , Glomerulonefrite , Animais , Células Endoteliais/patologia , Feminino , Mesângio Glomerular/patologia , Glomerulonefrite/metabolismo , Humanos , Imunoglobulina A/metabolismo , Glomérulos Renais/patologia , Masculino , Proteínas de Membrana/genética , Camundongos , Análise de Sequência de RNA
6.
Kidney Int ; 100(4): 809-823, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34147551

RESUMO

Inflammatory pathways are activated in most glomerular diseases but molecular mechanisms driving them in kidney tissue are poorly known. We identified retinoic acid receptor responder 1 (Rarres1) as a highly podocyte-enriched protein in healthy kidneys. Studies in podocyte-specific knockout animals indicated that Rarres1 was not needed for the normal development or maintenance of the glomerulus filtration barrier and did not modulate the outcome of kidney disease in a model of glomerulonephritis. Interestingly, we detected an induction of Rarres1 expression in glomerular and peritubular capillary endothelial cells in IgA and diabetic kidney disease, as well as in ANCA-associated vasculitis. Analysis of publicly available RNA data sets showed that the induction of Rarres1 expression was a common molecular mechanism in chronic kidney diseases. A conditional knock-in mouse line, overexpressing Rarres1 specifically in endothelial cells, did not show any obvious kidney phenotype. However, the overexpression promoted the progression of kidney damage in a model of glomerulonephritis. In line with this, conditional knock-out mice, lacking Rarres1 in endothelial cells, were partially protected in the disease model. Mechanistically, Rarres1 promoted inflammation and fibrosis via transcription factor Nuclear Factor-κB signaling pathway by activating receptor tyrosine kinase Axl. Thus, induction of Rarres1 expression in endothelial cells is a prevalent molecular mechanism in human glomerulopathies and this seems to have a pathogenic role in driving inflammation and fibrosis via the Nuclear Factor-κB signaling pathway.


Assuntos
Nefropatias Diabéticas , NF-kappa B , Animais , Nefropatias Diabéticas/genética , Células Endoteliais , Proteínas de Membrana , Camundongos , Receptores do Ácido Retinoico , Transdução de Sinais
7.
Nat Commun ; 12(1): 2141, 2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33837218

RESUMO

Molecular characterization of the individual cell types in human kidney as well as model organisms are critical in defining organ function and understanding translational aspects of biomedical research. Previous studies have uncovered gene expression profiles of several kidney glomerular cell types, however, important cells, including mesangial (MCs) and glomerular parietal epithelial cells (PECs), are missing or incompletely described, and a systematic comparison between mouse and human kidney is lacking. To this end, we use Smart-seq2 to profile 4332 individual glomerulus-associated cells isolated from human living donor renal biopsies and mouse kidney. The analysis reveals genetic programs for all four glomerular cell types (podocytes, glomerular endothelial cells, MCs and PECs) as well as rare glomerulus-associated macula densa cells. Importantly, we detect heterogeneity in glomerulus-associated Pdgfrb-expressing cells, including bona fide intraglomerular MCs with the functionally active phagocytic molecular machinery, as well as a unique mural cell type located in the central stalk region of the glomerulus tuft. Furthermore, we observe remarkable species differences in the individual gene expression profiles of defined glomerular cell types that highlight translational challenges in the field and provide a guide to design translational studies.


Assuntos
Células Endoteliais/metabolismo , Mesângio Glomerular/metabolismo , Podócitos/metabolismo , Biossíntese de Proteínas/genética , Transcriptoma/fisiologia , Animais , Separação Celular , Biologia Computacional , Citometria de Fluxo , Heterogeneidade Genética , Mesângio Glomerular/citologia , Humanos , Masculino , Camundongos , RNA-Seq , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptores da Fosfolipase A2/genética , Análise de Célula Única , Especificidade da Espécie
8.
J Am Soc Nephrol ; 30(9): 1573-1586, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31285284

RESUMO

BACKGROUND: Inflammatory processes play an important role in the pathogenesis of glomerulopathies. Finding novel ways to suppress glomerular inflammation may offer a new way to stop disease progression. However, the molecular mechanisms that initiate and drive inflammation in the glomerulus are still poorly understood. METHODS: We performed large-scale gene expression profiling of glomerulus-associated G protein-coupled receptors (GPCRs) to identify new potential therapeutic targets for glomerulopathies. The expression of Gprc5b in disease was analyzed using quantitative PCR and immunofluorescence, and by analyzing published microarray data sets. In vivo studies were carried out in a podocyte-specific Gprc5b knockout mouse line. Mechanistic studies were performed in cultured human podocytes. RESULTS: We identified an orphan GPCR, Gprc5b, as a novel gene highly enriched in podocytes that was significantly upregulated in common human glomerulopathies, including diabetic nephropathy, IgA nephropathy, and lupus nephritis. Similar upregulation of Gprc5b was detected in LPS-induced nephropathy in mice. Studies in podocyte-specific Gprc5b knockout mice showed that Gprc5b was not essential for normal development of the glomerular filtration barrier. However, knockout mice were partially protected from LPS-induced proteinuria and recruitment of inflammatory cells. Mechanistically, RNA sequencing in Gprc5b knockouts mice and experiments in cultured human podocytes showed that Gpr5cb regulated inflammatory response in podocytes via NF-κB signaling. CONCLUSIONS: GPRC5b is a novel podocyte-specific receptor that regulates inflammatory response in the glomerulus by modulating the NF-κB signaling pathway. Upregulation of Gprc5b in human glomerulopathies suggests that it may play a role in their pathogenesis.


Assuntos
Nefropatias/genética , NF-kappa B/metabolismo , Receptores Acoplados a Proteínas G/genética , Animais , Células Cultivadas , Quimiocina CCL2/genética , Nefropatias Diabéticas/genética , Perfilação da Expressão Gênica , Barreira de Filtração Glomerular/crescimento & desenvolvimento , Glomerulonefrite por IGA/genética , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Nefropatias/patologia , Lipopolissacarídeos , Nefrite Lúpica/genética , Camundongos , Camundongos Knockout , Podócitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/genética
9.
Sci Rep ; 9(1): 8888, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31221975

RESUMO

Podocytes have an important role in the pathogenesis of diabetic nephropathy (DN). Podocyte foot process effacement, mediated largely by the actin-based cytoskeleton of foot processes, is commonly detected in DN and is believed to be a key pathogenic event in the development of proteinuria. In this study, we identified coronin 2b (Coro2b), a member of known actin-regulating proteins, the coronins, as a highly podocyte-enriched molecule located at the cytoplasmic side of the apical plasma membrane. Studies in human renal biopsies show that glomerular Coro2b expression is significantly down-regulated in patients with DN. Studies in knockout mice indicate that Coro2b is not required for the development or maintenance of the glomerular filtration barrier. Moreover, inactivation of Coro2b specifically in podocytes does not affect the outcome of nephropathy in a streptozotocin-induced diabetes model. However, Coro2b seems to modulate the reorganization of foot processes under pathological conditions as Coro2b knockout podocytes are partially protected from protamine sulfate perfusion-induced foot process effacement. Taken together, our study suggests a role for Coro2b in the pathogenesis of glomerulopathies. Further studies regarding the involvement of Coro2b in podocyte health and diseases are warranted.


Assuntos
Pé Diabético/metabolismo , Nefropatias Diabéticas/metabolismo , Regulação para Baixo , Proteínas dos Microfilamentos/metabolismo , Podócitos/metabolismo , Protaminas/metabolismo , Humanos
10.
Artigo em Inglês | MEDLINE | ID: mdl-30083135

RESUMO

Over the past two decades it has become increasing clear that injury and loss of podocytes is an early and common clinical observation presented in many forms of glomerulopathy and chronic kidney disease. Identification of disease-causing monogenic mutations in numerous podocyte-expressed genes as well as studies conducted using preclinical animal models have shown that the podocyte plays a central role in establishing kidney dysfunction. In this review, we summarize current knowledge regarding the potential for podocyte-targeted therapies and give our view on how a deeper understanding of the molecular makeup of the podocyte will enable future therapeutic interventions. Specifically, we recount some of the currently described podocentric strategies for therapy and summarize the status and evolution of various model systems used to facilitate our understanding of the molecular and functional underpinnings of podocyte biology.

11.
J Am Soc Nephrol ; 29(6): 1679-1689, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29636387

RESUMO

Background Renal glomeruli are the primary target of injury in diabetic nephropathy (DN), and the glomerular podocyte has a key role in disease progression.Methods To identify potential novel therapeutic targets for DN, we performed high-throughput molecular profiling of G protein-coupled receptors (GPCRs) using human glomeruli.Results We identified an orphan GPCR, Gprc5a, as a highly podocyte-specific gene, the expression of which was significantly downregulated in glomeruli of patients with DN compared with those without DN. Inactivation of Gprc5a in mice resulted in thickening of the glomerular basement membrane and activation of mesangial cells, which are two hallmark features of DN in humans. Compared with wild-type mice, Gprc5a-deficient animals demonstrated increased albuminuria and more severe histologic changes after induction of diabetes with streptozotocin. Mechanistically, Gprc5a modulated TGF-ß signaling and activation of the EGF receptor in cultured podocytes.Conclusions Gprc5a has an important role in the pathogenesis of DN, and further study of the podocyte-specific signaling activity of this protein is warranted.


Assuntos
Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Podócitos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Actinas/metabolismo , Albuminúria/etiologia , Animais , Linhagem Celular , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/patologia , Regulação para Baixo , Receptores ErbB/metabolismo , Fibronectinas/metabolismo , Membrana Basal Glomerular/patologia , Humanos , Células Mesangiais , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/metabolismo
12.
Physiol Rep ; 5(5)2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28292877

RESUMO

The leptin-deficient BTBRob/ob mouse develops progressive albuminuria and morphological lesions similar to human diabetic nephropathy (DN), although whether glomerular hyperfiltration, a recognized feature of early DN that may contribute to renal injury, also occurs in this model is not known. Leptin replacement has been shown to reverse the signs of renal injury in this model, but in contrast, the expected renoprotection by angiotensin-converting enzyme (ACE) inhibition in BTBRob/ob mice seems to be limited. Therefore, to investigate the potential renal benefits of improved metabolic control in this model, we studied the effect of treatment with the dual peroxisome proliferator-activated receptor (PPAR) α/γ agonist AZD6610 and compared it with the ACE inhibitor enalapril. AZD6610 lowered plasma glucose and triglyceride concentrations and increased liver size, but had no significant effect in reducing albuminuria, whereas enalapril did have an effect. Nephrin and WT1 mRNA expression decreased in the kidneys of BTBRob/ob mice, consistent with podocyte injury and loss, but was unaffected by either drug treatment: at the protein level, both nephrin and WT1-positive cells per glomerulus were decreased. Mesangial matrix expansion was reduced in AZD6610-treated mice. GFR, measured by creatinine clearance, was increased in BTBRob/ob mice, but unaffected by either treatment. Unexpectedly, enalapril-treated mice showed intrarenal arteriolar vascular remodeling with concentric thickening of vessel walls. In summary, we found that the BTBRob/ob mouse model shows some similarities to the early changes seen in human DN, but that ACE inhibition or PPARα/γ agonism afforded limited or no kidney protection.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Glicemia/metabolismo , Diabetes Mellitus/metabolismo , Nefropatias Diabéticas/metabolismo , Rim/efeitos dos fármacos , PPAR alfa/antagonistas & inibidores , PPAR gama/antagonistas & inibidores , Animais , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Enalapril/farmacologia , Rim/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Triglicerídeos/sangue , Proteínas WT1
13.
Drug Discov Today ; 20(10): 1228-34, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26096184

RESUMO

The majority of chronic kidney disease (CKD) cases have their origin in the glomerulus, the microvascular unit of the nephron that serves as a filter tasked with forming primary urine. This selective filtration process is determined to a large extent by the functional capacity of the podocyte, a highly differentiated cell type that enwraps the outer aspect of the glomerular capillary wall. In this short review, we describe the biology of the podocyte, its central role in the etiology of various glomerulopathies and highlight current and future opportunities to exploit the unique properties of this cell type for developing kidney-specific therapeutics.


Assuntos
Glomérulos Renais/patologia , Terapia de Alvo Molecular , Podócitos/metabolismo , Animais , Desenho de Fármacos , Humanos , Insuficiência Renal Crônica/fisiopatologia
15.
PLoS One ; 10(3): e0122067, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25807495

RESUMO

BACKGROUND: Podocyte foot process effacement accompanied by actin cytoskeleton rearrangements is a cardinal feature of many progressive human proteinuric diseases. RESULTS: By microarray profiling of mouse glomerulus, SCHIP1 emerged as one of the most highly enriched transcripts. We detected Schip1 protein in the kidney glomerulus, specifically in podocytes foot processes. Functionally, Schip1 inactivation in zebrafish by morpholino knock-down results in foot process disorganization and podocyte loss leading to proteinuria. In cultured podocytes Schip1 localizes to cortical actin-rich regions of lamellipodia, where it forms a complex with Nherf2 and ezrin, proteins known to participate in actin remodeling stimulated by PDGFß signaling. Mechanistically, overexpression of Schip1 in vitro causes accumulation of cortical F-actin with dissolution of transversal stress fibers and promotes cell migration in response to PDGF-BB stimulation. Upon actin disassembly by latrunculin A treatment, Schip1 remains associated with the residual F-actin-containing structures, suggesting a functional connection with actin cytoskeleton possibly via its interaction partners. A similar assay with cytochalasin D points to stabilization of cortical actin cytoskeleton in Schip1 overexpressing cells by attenuation of actin depolymerisation. CONCLUSIONS: Schip1 is a novel glomerular protein predominantly expressed in podocytes, necessary for the zebrafish pronephros development and function. Schip1 associates with the cortical actin cytoskeleton network and modulates its dynamics in response to PDGF signaling via interaction with the Nherf2/ezrin complex. Its implication in proteinuric diseases remains to be further investigated.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fosfoproteínas/metabolismo , Podócitos/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Actinas/metabolismo , Animais , Becaplermina , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Movimento Celular , Células Cultivadas , Citocalasina D/metabolismo , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Glomérulos Renais/metabolismo , Microscopia de Fluorescência , Oligonucleotídeos Antissenso/metabolismo , Podócitos/citologia , Pronefro/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Pseudópodes/metabolismo , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Tiazolidinas/farmacologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética
16.
J Am Soc Nephrol ; 26(3): 647-62, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25071083

RESUMO

Rhophilin-1 is a Rho GTPase-interacting protein, the biologic function of which is largely unknown. Here, we identify and describe the functional role of Rhophilin-1 as a novel podocyte-specific protein of the kidney glomerulus. Rhophilin-1 knockout mice were phenotypically normal at birth but developed albuminuria at about 2 weeks of age. Kidneys from severely albuminuric mice revealed widespread podocyte foot process effacement, thickening of the glomerular basement membrane, and FSGS-like lesions. The absence of any overt changes in the expression of podocyte proteins at the onset of proteinuria suggested that the primary cause of podocyte abnormalities in Rhpn1-null mice was the result of cell-autonomous, Rhophilin-1-dependent signaling events. In culture, Rhophilin-1 was detected at the plasma membrane leading edge of primary podocytes, where it elicited remodeling of the actin cytoskeleton network. This effect of Rhophilin-1 on actin cytoskeleton organization associated with inhibitory effects on Rho-dependent phosphorylation of the myosin regulatory light chain and stress fiber formation. Conversely, phosphorylation of myosin regulatory light chain increased in podocyte foot processes of Rhpn1(-/-) mice, implicating altered actinomyosin contractility in foot process effacement and compromised filtration capacity. Targeted deletion of RhoA in podocytes of Rhophilin-1 knockout mice exacerbated the renal injury. Taken together, our results indicate that Rhophilin-1 is essential for the integrity of the glomerular filtration barrier and that this protein is a key determinant of podocyte cytoskeleton architecture.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Citoesqueleto/fisiologia , Podócitos/fisiologia , Animais , Células Cultivadas , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout
17.
Nephron Extra ; 4(3): 146-54, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25404935

RESUMO

BACKGROUND/AIMS: The transmembrane proteins Neph1 and nephrin form a complex in the slit diaphragm (SD) of podocytes. As recent studies indicate an involvement of this complex in the polymerization of the actin cytoskeleton and proteinuria, we wanted to study the subcellular localization of Neph1 in the normal human kidney and its expression in focal segmental glomerulosclerosis (FSGS), minimal change nephrotic syndrome (MCNS), and the corresponding experimental models of Adriamycin-induced nephropathy (ADR) and puromycin aminonucleoside nephrosis (PAN). All these disorders are characterized by substantial foot process effacement (FPE) and proteinuria. MATERIALS AND METHODS: Kidney biopsies from patients with primary FSGS (perihilar type) and MCNS were compared to normal renal tissue. Mouse and rat kidney cortices from days 7 and 14 after Adriamycin injection and days 2 and 4 after puromycin aminonucleoside injection, respectively, were compared to control mouse and rat kidney. Polyclonal antibodies against Neph1 and nephrin were used for immunoelectron microscopy, and semiquantification was performed. RESULTS: We localized Neph1 mainly to, and in close proximity to, the SD. Double staining of Neph1 and nephrin showed the proteins to be in close connection in the SD. The total amount of Neph1 in the podocytes was significantly reduced in FSGS, MCNS, ADR, and PAN. The reduction of Neph1 was also seen in areas with and without FPE. Nephrin was reduced in MCNS and PAN but unchanged in FSGS. CONCLUSION: With nephrin (but not Neph1) unchanged in FSGS, there might be a disruption of the complex and an involvement of Neph1 in its pathogenesis.

18.
Pediatr Nephrol ; 29(4): 505-11, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23824180

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic cause of end-stage renal disease, affecting approximately 1 in 1,000 people. The disease is characterized by the development of numerous large fluid-filled renal cysts over the course of decades. These cysts compress the surrounding renal parenchyma and impair its function. Mutations in two genes are responsible for ADPKD. The protein products of both of these genes, polycystin-1 and polycystin-2, localize to the primary cilium and participate in a wide variety of signaling pathways. Polycystin-1 undergoes several proteolytic cleavages that produce fragments which manifest biological activities. Recent results suggest that the production of polycystin-1 cleavage fragments is necessary and sufficient to account for at least some, although certainly not all, of the physiological functions of the parent protein.


Assuntos
Regulação da Expressão Gênica/fisiologia , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Humanos , Mutação , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Transcrição Gênica
19.
Mol Cell Biol ; 33(20): 4030-40, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23938297

RESUMO

Platelet-derived growth factor A-chain (PDGF-A) exists in two evolutionarily conserved isoforms, PDGF-Along and PDGF-Ashort, generated by alternative RNA splicing. They differ by the presence (in PDGF-Along) or absence (in PDGF-Ashort) of a carboxy-terminal heparin/heparan sulfate proteoglycan-binding motif. In mice, similar motifs present in other members of the PDGF and vascular endothelial growth factor (VEGF) families have been functionally analyzed in vivo, but the specific physiological importance of PDGF-Along has not been explored previously. Here, we analyzed the absolute and relative expression of the two PDGF-A splice isoforms during early postnatal organ development in the mouse and report on the generation of a Pdgfa allele (Pdgfa(Δex6)) incapable of producing PDGF-Along due to a deletion of the exon 6 splice acceptor site. In situations of limiting PDGF-A signaling through PDGF receptor alpha (PDGFRα), or in mice lacking PDGF-C, homozygous carriers of Pdgfa(Δex6) showed abnormal development of the lung, intestine, and vertebral column, pinpointing developmental processes where PDGF-Along may play a physiological role.


Assuntos
Sequência de Bases , Mucosa Intestinal/metabolismo , Pulmão/metabolismo , Camundongos Transgênicos/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Deleção de Sequência , Coluna Vertebral/metabolismo , Processamento Alternativo , Motivos de Aminoácidos , Animais , Animais Recém-Nascidos , Sítios de Ligação , Éxons , Efeito Fundador , Regulação da Expressão Gênica no Desenvolvimento , Heparina/metabolismo , Intestinos/anormalidades , Intestinos/crescimento & desenvolvimento , Pulmão/anormalidades , Pulmão/crescimento & desenvolvimento , Camundongos , Camundongos Transgênicos/metabolismo , Dados de Sequência Molecular , Morfogênese/genética , Fator de Crescimento Derivado de Plaquetas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Coluna Vertebral/anormalidades , Coluna Vertebral/crescimento & desenvolvimento
20.
Kidney Int ; 82(10): 1071-83, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22832517

RESUMO

Pleckstrin homology domain-containing, family H (with MyTH4 domain), member 2 (Plekhh2) is a 1491-residue intracellular protein highly enriched in renal glomerular podocytes for which no function has been ascribed. Analysis of renal biopsies from patients with focal segmental glomerulosclerosis revealed a significant reduction in total podocyte Plekhh2 expression compared to controls. Sequence analysis indicated a putative α-helical coiled-coil segment as the only recognizable domain within the N-terminal half of the polypeptide, while the C-terminal half contains two PH, a MyTH4, and a FERM domain. We identified a phosphatidylinositol-3-phosphate consensus-binding site in the PH1 domain required for Plekhh2 localization to peripheral regions of cell lamellipodia. The N-terminal half of Plekkh2 is not necessary for lamellipodial targeting but mediates self-association. Yeast two-hybrid screening showed that Plekhh2 directly interacts through its FERM domain with the focal adhesion protein Hic-5 and actin. Plekhh2 and Hic-5 coprecipitated and colocalized at the soles of podocyte foot processes in situ and Hic-5 partially relocated from focal adhesions to lamellipodia in Plekhh2-expressing podocytes. In addition, Plekhh2 stabilizes the cortical actin cytoskeleton by attenuating actin depolymerization. Our findings suggest a structural and functional role for Plekhh2 in the podocyte foot processes.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Junções Célula-Matriz/metabolismo , Glomerulosclerose Segmentar e Focal/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Podócitos/metabolismo , Citoesqueleto de Actina/patologia , Animais , Sítios de Ligação , Biópsia , Células CHO , Células COS , Estudos de Casos e Controles , Chlorocebus aethiops , Cricetinae , Cricetulus , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/patologia , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Domínio LIM/metabolismo , Camundongos , Fosfatos de Fosfatidilinositol/metabolismo , Podócitos/patologia , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Estrutura Secundária de Proteína , Transporte Proteico , Pseudópodes/metabolismo , Análise de Sequência de Proteína , Transfecção , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...