Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38513055

RESUMO

Virtual reality (VR) is a potent educational tool with untapped potential in medical training. However, its integration into medical schools faces challenges such as cybersickness and hesitancy among medical students and professionals. Notably, there has been no systematic assessment of the acceptance of medical educational VR applications by both students and educators. In our single-center study, we enrolled 133 medical students and 14 medical educators. Following a practical demonstration of the established VR anatomy application, Sharecare YOU VR, participants completed a self-reporting survey based on the Technology Acceptance Model (TAM), exploring user acceptance of information technologies and focusing on perceived usefulness (PU), perceived ease of use (PEU), and attitude toward using (ATU). We also sought insights into potential future applications of VR in medical education. Our findings indicate a high level of acceptance among medical students and educators upon structured exposure to VR with significantly positive responses for all three TAM variables (PU, PEU, and ATU). Intriguingly, hands-on experience influenced acceptance. Students envisioned VR's benefits in anatomy, surgery, emergency medicine, and communication skill training with patients. Both students and educators believed that VR could enhance traditional approaches and complement the existing curriculum, anticipating improved preparedness for medical students through VR training applications. In conclusion, our results demonstrate the receptivity of both students and educators to immersive technologies, including VR, in medical education. Importantly, the data suggest that the adoption of VR in this field would be welcomed rather than resisted, potentially enhancing students' self-efficacy and enriching the medical school curriculum.

2.
Cells ; 11(24)2022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36552716

RESUMO

Metabolic effects of physical activity may be reno-protective in the context of hypertension, although exercise stresses kidneys. Aldosterone participates in renal disease in hypertension, but exercise affects the plasma concentration of aldosterone. This study was designed to evaluate whether physical activity and pharmacological treatment by aldosterone have additive effects on renal protection in hypertensive rats. Female spontaneously hypertensive rats (SHR) or normotensive Wistar rats performed voluntary running wheel activity alone or in combination with aldosterone blockade (spironolactone). The following groups were studied: young and pre-hypertensive SHR (n = 5 sedentary; n = 10 running wheels, mean body weight 129 g), 10-month-old Wistar rats (n = 6 sedentary; n = 6 running wheels, mean body weight 263 g), 10-month-old SHRs (n = 18 sedentary, mean body weight 224 g; n = 6 running wheels, mean body weight 272 g; n = 6 aldosterone, mean body weight 219 g; n = 6 aldosterone and running wheels, mean body weight 265 g). Another group of SHRs had free access to running wheels for 6 months and kept sedentary for the last 3 months (n = 6, mean body weight 240 g). Aldosterone was given for the last 4 months. SHRs from the running groups had free access to running wheels beginning at the age of 6 weeks. Renal function was analyzed by microalbuminuria (Alb/Cre), urinary secretion of kidney injury molecule-1 (uKim-1), and plasma blood urea nitrogen (BUN) concentration. Molecular adaptation of the kidney to hypertension and its modification by spironolactone and/or exercise were analyzed by real-time PCR, immunoblots, and histology. After six months of hypertension, rats had increased Alb/Cre and BUN but normal uKim-1. Voluntary free running activity normalized BUN but not Alb/Cre, whereas spironolactone reduced Alb/Cre but not BUN. Exercise constitutively increased renal expression of proprotein convertase subtilisin/kexin type 9 (PCSK9; mRNA and protein) and arginase-2 (mRNA). Spironolactone reduced these effects. uKim-1 increased in rats performing voluntary running wheel activity exercise irrespectively of blood pressure and aldosterone blockade. We observed independent but no additive effects of aldosterone blockade and physical activity on renal function and on molecules potentially affecting renal lipid metabolism.


Assuntos
Hipertensão , Pró-Proteína Convertase 9 , Animais , Feminino , Ratos , Aldosterona , Peso Corporal , Hipertensão/tratamento farmacológico , Rim/metabolismo , Ratos Endogâmicos SHR , Ratos Wistar , RNA Mensageiro/metabolismo , Espironolactona/farmacologia , Atividade Motora/fisiologia
3.
Sci Rep ; 10(1): 7519, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32372032

RESUMO

Tissue macrophages play an important role in organ homeostasis, immunity and the pathogenesis of various inflammation-driven diseases. One major challenge has been to selectively study resident macrophages in highly heterogeneous organs such as kidney. To address this problem, we adopted a Translational Ribosome Affinity Purification (TRAP)- approach and designed a transgene that expresses an eGFP-tagged ribosomal protein (L10a) under the control of the macrophage-specific c-fms promoter to generate c-fms-eGFP-L10a transgenic mice (MacTRAP). Rigorous characterization found no gross abnormalities in MacTRAP mice and confirmed transgene expression across various organs. Immunohistological analyses of MacTRAP kidneys identified eGFP-L10a expressing cells in the tubulointerstitial compartment which stained positive for macrophage marker F4/80. Inflammatory challenge led to robust eGFP-L10a upregulation in kidney, confirming MacTRAP responsiveness in vivo. We successfully extracted macrophage-specific polysomal RNA from MacTRAP kidneys and conducted RNA sequencing followed by bioinformatical analyses, hereby establishing a comprehensive and unique in vivo gene expression and pathway signature of resident renal macrophages. In summary, we created, validated and applied a new, responsive macrophage-specific TRAP mouse line, defining the translational profile of renal macrophages and dendritic cells. This new tool may be of great value for the study of macrophage biology in different organs and various models of injury and disease.


Assuntos
Técnicas Genéticas , Rim/citologia , Macrófagos/metabolismo , Biossíntese de Proteínas , Animais , Células Dendríticas/metabolismo , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Inflamação , Camundongos , Camundongos Transgênicos , RNA/metabolismo , RNA-Seq , Ribossomos/metabolismo , Transgenes
4.
Clin Lab Med ; 39(1): 157-169, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30709504

RESUMO

It is increasingly recognized that calcineurin inhibitors (CNI) such as cyclosporine and tacrolimus are not ideal immunosuppressive agents. Side effects, including increased rates of infection, hypertension, and malignancy, can be severe. Thus, in the past decade, there has been much focus on the development of novel therapeutic agents and strategies designed to replace or minimize CNI exposure in transplant patients. This article reviews potential novel targets in T cells, alloantibody-producing B cells, plasma cells, and complement in transplantation.


Assuntos
Terapia de Imunossupressão/métodos , Transplante , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Fatores Imunológicos/uso terapêutico , Modelos Imunológicos , Linfócitos T/fisiologia , Condicionamento Pré-Transplante/métodos
5.
Physiol Rep ; 6(17): e13842, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30198211

RESUMO

An active lifestyle is generally recommended for hypertensive patients to prevent subsequent end-organ damage. However, experimental data on long-term effects of exercise on hypertension are insufficient and underlying mechanisms are not well understood. This study was aimed to investigate the effect of exercise on renal expression of parathyroid hormone-related protein (PTHrP) and parathyroid hormone receptor type 1 (PTHR1) in spontaneously hypertensive rats (SHR). Twenty-four rats started free running wheel exercise at the age of 1.5 months (pre-hypertensive state) and proceeded for 1.5, 3.0, 6.0, and 10.0 months. Thirty rats kept under standard housing conditions were used as sedentary controls. Kidney function was assessed by measuring plasma creatinine levels and urine albumin-to-creatinine ratios. Renal expression of PTHrP and PTHR1 was analyzed by qRT-PCR and western blot. Renal expression of PTHR1 was markedly increased between the 6th and 10th months in sedentary rats and this increase was significantly lower in SHRs with high physical activity on mRNA (-30%) and protein level (-27%). At the same time, urine albumin-to-creatinine ratio increased (from 65 to 231 mg/g) but somehow lower in exercise performing SHRs (48-196 mg/g). Our data suggest that enhanced exercise, stimulated by allocation of a free running wheel, is associated with lower PTHR1 expression in SHRs and this may contribute to preserved kidney function.


Assuntos
Hipertensão Essencial/metabolismo , Rim/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/genética , Condicionamento Físico Animal/métodos , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Corrida , Animais , Hipertensão Essencial/fisiopatologia , Hipertensão Essencial/terapia , Feminino , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo
6.
Curr Opin Organ Transplant ; 23(1): 51-62, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29176362

RESUMO

PURPOSE OF REVIEW: Finding the right immunosuppressive approach for the individual transplant patient is of utmost importance. In truth, a 'one size fits all' does not exist, and as patients differ, so should therapeutic suppression of the immune system be individualized. For over three decades, biologics have been a central component of transplant immunosuppression. Our growing knowledge of immunological processes combined with biotechnological advances is leading to promising new therapeutic concepts and possibilities including novel biologics. Use of biologics may be appropriate at various phases of kidney transplantation, from desensitization and induction to maintenance therapy and management of acute rejection. Their mechanisms of action include depleting or modulating immune cells, eliminating preformed antibodies, and inhibiting the complement system. Herein, we summarize the current approaches to applying 'established' biologics to prevent and treat allograft rejection in kidney transplantation. We also provide insights into new developments and possible future directions. RECENT FINDINGS: A number of candidate biologics were found to be efficacious in more recent preclinical and early phase clinical trials. Their properties are outlined and their potential for future utilization discussed. SUMMARY: The extraordinary capabilities of biologics are undisputed and our technological progress offers unprecedented opportunities to devise new agents and refine old ones. However, the rationale for their use in kidney allograft recipients must be rigorously examined in every case and, given the significant risk of early and late-onset adverse effects, the risk-to-benefit ratio carefully balanced. We also need to expand and use our knowledge of the underlying physiology of allograft rejection to adjust the characteristics of therapeutic biologics and thus harness their full potential for the benefit of our patients.


Assuntos
Produtos Biológicos/uso terapêutico , Rejeição de Enxerto/tratamento farmacológico , Transplante de Rim/efeitos adversos , Animais , Rejeição de Enxerto/etiologia , Humanos
7.
Cell Tissue Res ; 369(1): 159-170, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28601904

RESUMO

The mouse still represents arguably the most important mammal organism in research for modeling human genetic kidney diseases in vivo. Compared with many other mammal species, the breeding and maintenance of mice in the laboratory is relatively simple and cheap and reproduction cycles are short. In addition to classic gene knockout mouse lines, new molecular biological technologies have led to the development of a plethora of other, more sophisticated, mouse models, allowing the targeting of genes or gene function in a cell-specific, tissue-specific and time-dependent fashion. With the refinement of more recently developed genome-editing technologies, including the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas system and other engineered nucleases such as zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs), our "tool set" of mouse models is expected to rapidly expand. These technological advances hold great promise and should enable us to study and hence understand the biology of inherited kidney diseases in greater detail. By analogy, we may be able to answer questions regarding the impact of individual proteins on the development of human kidney disorders, the underlying mechanisms governing the evolution of the disease and the predicted responsiveness to therapeutic interventions. Moreover, knockout and transgenic mouse models can be highly informative with respect to the effects of genetic variations on renal phenotypes. This review focuses on mouse models that have been devised primarily to study monogenic human kidney diseases, which are typically caused by a single abnormal gene and passed on in a Mendelian pattern. Despite the large number of human hereditary kidney disorders and the multitude of mouse models described in the literature, we attempt to give a balanced overview of several well-known renal pathologies, a few of which are addressed in some detail.


Assuntos
Sistemas CRISPR-Cas , Modelos Animais de Doenças , Doenças Genéticas Inatas , Animais , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Doenças Genéticas Inatas/patologia , Humanos , Camundongos
8.
Eur J Cell Biol ; 94(7-9): 391-400, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26116074

RESUMO

Ca(2+)-activated K(+) channels (KCa) play a pivotal role in the endothelium-dependent hyperpolarization and regulation of vascular tone and blood pressure. For activation, KCa depend on an increase of intracellular calcium which is substantially mediated by Ca(2+)-permeable cation channels including the transient receptor potential V4 (TRPV4). It has been proposed that KCa and Ca(2+)-permeable cation channels may be clustered in localized positions within the cell membrane to form functional units and that caveolae may constitute the scaffolding for such microcompartmental organization. Here, we sought to elucidate the composition and functional relevance of these microcompartments in vitro and in vivo. We show that TRPV4 and small-conductance KCa2.3 are enriched in caveolae of human microvascular endothelial cells. Using immunoprecipitation, immunocytology and superresolution microscopy, we found a caveolae-dependent association between caveolin-1, TRPV4 and small conductance KCa2.3, but not intermediate conductance KCa3.1, in endothelial cells under static condition. Mechanical stimulation of cells via exposure to shear stress led to a partial de-novo colocalization of KCa3.1 with Cav-1 and TRPV4. In a mouse model of genetic Cav-1 deficiency, we found significantly reduced KCa-mediated currents as determined by patch-clamping in carotid artery endothelial cells (CAEC) from Cav-1(-/-) mice compared to wildtype. Functionally, Cav-1 deficiency was associated with impaired endothelium-derived hyperpolarizing factor (EDHF)-mediated vasodilation in response to shear stress and acetylcholine. In summary, our findings provide evidence for a dynamic microcompartmentation of TRPV4/KCa in caveolae of endothelial cells and highlight the importance of Cav-1 for endothelial KCa functions and flow-induced vasodilation.


Assuntos
Cavéolas/fisiologia , Caveolina 1/metabolismo , Células Endoteliais/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Caveolina 1/genética , Compartimento Celular , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Vasodilatação/fisiologia
9.
Kidney Int ; 86(6): 1116-29, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24940801

RESUMO

Identifying new biomarkers and therapeutic targets for podocytopathies such as focal segmental glomerulosclerosis (FSGS) requires a detailed analysis of transcriptional changes in podocytes over the course of disease. Here we used translating ribosome affinity purification (TRAP) to isolate and profile podocyte-specific mRNA in two different models of FSGS. We expressed enhanced green fluorescent protein-tagged to ribosomal protein L10a in podocytes under the control of the collagen-1α1 promoter, enabling one-step podocyte-specific mRNA isolation over the course of disease. This TRAP protocol robustly enriched known podocyte-specific mRNAs. We crossed Col1α1-eGFP-L10a mice with the Actn4(-/-) and Actn4(+/K256E) models of FSGS and analyzed podocyte transcriptional profiles at 2, 6, and 44 weeks of age. Two upregulated podocyte genes in murine FSGS (CXCL1 and DMPK) were found to be upregulated at the protein level in biopsies from patients with FSGS, validating this approach. There was no dilution of podocyte-specific transcripts during disease. These are the first podocyte-specific RNA expression data sets during aging and in two models of FSGS. This approach identified new podocyte proteins that are upregulated in FSGS and defines novel biomarkers and therapeutic targets for human glomerular disease.


Assuntos
Actinina/genética , Envelhecimento/genética , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/metabolismo , Podócitos/metabolismo , RNA Mensageiro/análise , Envelhecimento/metabolismo , Animais , Biomarcadores/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Knockout , Miotonina Proteína Quinase/genética , Miotonina Proteína Quinase/metabolismo , Proteínas de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Biossíntese de Proteínas , Proteína Ribossômica L10 , Proteínas Ribossômicas/genética , Transcriptoma
10.
J Am Soc Nephrol ; 25(9): 1979-90, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24652793

RESUMO

Myofibroblasts secrete matrix during chronic injury, and their ablation ameliorates fibrosis. Development of new biomarkers and therapies for CKD will be aided by a detailed analysis of myofibroblast gene expression during the early stages of fibrosis. However, dissociating myofibroblasts from fibrotic kidney is challenging. We therefore adapted translational ribosome affinity purification (TRAP) to isolate and profile mRNA from myofibroblasts and their precursors during kidney fibrosis. We generated and characterized a transgenic mouse expressing an enhanced green fluorescent protein (eGFP)-tagged L10a ribosomal subunit protein under control of the collagen1α1 promoter. We developed a one-step procedure for isolation of polysomal RNA from collagen1α1-eGFPL10a mice subject to unilateral ureteral obstruction and analyzed and validated the resulting transcriptional profiles. Pathway analysis revealed strong gene signatures for cell proliferation, migration, and shape change. Numerous novel genes and candidate biomarkers were upregulated during fibrosis, specifically in myofibroblasts, and we validated these results by quantitative PCR, in situ, and Western blot analysis. This study provides a comprehensive analysis of early myofibroblast gene expression during kidney fibrosis and introduces a new technique for cell-specific polysomal mRNA isolation in kidney injury models that is suited for RNA-sequencing technologies.


Assuntos
Rim/metabolismo , Rim/patologia , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Animais , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Fibrose , Perfilação da Expressão Gênica/métodos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Rim/lesões , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína Ribossômica L10 , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Regulação para Cima , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
11.
J Clin Invest ; 124(3): 1242-54, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24569379

RESUMO

Acute kidney injury (AKI) promotes an abrupt loss of kidney function that results in substantial morbidity and mortality. Considerable effort has gone toward identification of diagnostic biomarkers and analysis of AKI-associated molecular events; however, most studies have adopted organ-wide approaches and have not elucidated the interplay among different cell types involved in AKI pathophysiology. To better characterize AKI-associated molecular and cellular events, we developed a mouse line that enables the identification of translational profiles in specific cell types. This strategy relies on CRE recombinase-dependent activation of an EGFP-tagged L10a ribosomal protein subunit, which allows translating ribosome affinity purification (TRAP) of mRNA populations in CRE-expressing cells. Combining this mouse line with cell type-specific CRE-driver lines, we identified distinct cellular responses in an ischemia reperfusion injury (IRI) model of AKI. Twenty-four hours following IRI, distinct translational signatures were identified in the nephron, kidney interstitial cell populations, vascular endothelium, and macrophages/monocytes. Furthermore, TRAP captured known IRI-associated markers, validating this approach. Biological function annotation, canonical pathway analysis, and in situ analysis of identified response genes provided insight into cell-specific injury signatures. Our study provides a deep, cell-based view of early injury-associated molecular events in AKI and documents a versatile, genetic tool to monitor cell-specific and temporal-specific biological processes in disease modeling.


Assuntos
Injúria Renal Aguda/metabolismo , Traumatismo por Reperfusão/metabolismo , Transcriptoma , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Animais , Ontologia Genética , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Rim/irrigação sanguínea , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos , Biossíntese de Proteínas , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Traumatismo por Reperfusão/genética , Proteínas Ribossômicas/biossíntese , Proteínas Ribossômicas/genética
12.
J Clin Invest ; 123(9): 4023-35, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23979159

RESUMO

Acute kidney injury predisposes patients to the development of both chronic kidney disease and end-stage renal failure, but the molecular details underlying this important clinical association remain obscure. We report that kidney injury molecule-1 (KIM-1), an epithelial phosphatidylserine receptor expressed transiently after acute injury and chronically in fibrotic renal disease, promotes kidney fibrosis. Conditional expression of KIM-1 in renal epithelial cells (Kim1(RECtg)) in the absence of an injury stimulus resulted in focal epithelial vacuolization at birth, but otherwise normal tubule histology and kidney function. By 4 weeks of age, Kim1(RECtg) mice developed spontaneous and progressive interstitial kidney inflammation with fibrosis, leading to renal failure with anemia, proteinuria, hyperphosphatemia, hypertension, cardiac hypertrophy, and death, analogous to progressive kidney disease in humans. Kim1(RECtg) kidneys had elevated expression of proinflammatory monocyte chemotactic protein-1 (MCP-1) at early time points. Heterologous expression of KIM-1 in an immortalized proximal tubule cell line triggered MCP-1 secretion and increased MCP-1-dependent macrophage chemotaxis. In mice expressing a mutant, truncated KIM-1 polypeptide, experimental kidney fibrosis was ameliorated with reduced levels of MCP-1, consistent with a profibrotic role for native KIM-1. Thus, sustained KIM-1 expression promotes kidney fibrosis and provides a link between acute and recurrent injury with progressive chronic kidney disease.


Assuntos
Falência Renal Crônica/metabolismo , Rim/metabolismo , Proteínas de Membrana/metabolismo , Animais , Citocinas/metabolismo , Fibrose , Receptor Celular 1 do Vírus da Hepatite A , Mediadores da Inflamação/metabolismo , Rim/patologia , Falência Renal Crônica/imunologia , Falência Renal Crônica/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Nefrite/metabolismo , Fenótipo , Podócitos/metabolismo , Insuficiência Renal Crônica/imunologia , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia
13.
Kidney Int ; 82(2): 172-83, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22437410

RESUMO

Chronic kidney disease (CKD) remains one of the leading causes of death in the developed world, and acute kidney injury (AKI) is now recognized as a major risk factor in its development. Understanding the factors leading to CKD after acute injury are limited by current animal models of AKI, which concurrently target various kidney cell types including epithelial, endothelial, and inflammatory cells. Here, we developed a mouse model of kidney injury using the Six2-Cre-LoxP technology to selectively activate expression of the simian diphtheria toxin (DT) receptor in renal epithelia derived from the metanephric mesenchyme. By adjusting the timing and dose of DT, a highly selective model of tubular injury was created to define the acute and chronic consequences of isolated epithelial injury. The DT-induced sublethal tubular epithelial injury was confined to the S1 and S2 segments of the proximal tubule rather than being widespread in the metanephric mesenchyme-derived epithelial lineage. Acute injury was promptly followed by inflammatory cell infiltration and robust tubular cell proliferation, leading to complete recovery after a single toxin insult. In striking contrast, three insults to renal epithelial cells at 1-week intervals resulted in maladaptive repair with interstitial capillary loss, fibrosis, and glomerulosclerosis, which was highly correlated with the degree of interstitial fibrosis. Thus, selective epithelial injury can drive the formation of interstitial fibrosis, capillary rarefaction, and potentially glomerulosclerosis, substantiating a direct role for damaged tubule epithelium in the pathogenesis of CKD.


Assuntos
Injúria Renal Aguda/complicações , Células Epiteliais/patologia , Glomerulonefrite/etiologia , Túbulos Renais Proximais/patologia , Injúria Renal Aguda/genética , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Apoptose , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Fibrose , Glomerulonefrite/genética , Glomerulonefrite/imunologia , Glomerulonefrite/metabolismo , Glomerulonefrite/patologia , Glomerulonefrite/fisiopatologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Túbulos Renais Proximais/imunologia , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/fisiopatologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regeneração , Fatores de Tempo
14.
J Am Soc Nephrol ; 23(5): 785-91, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22362911

RESUMO

Visualizing podocyte foot processes requires electron microscopy, a technique that depends on special equipment, requires immunogold for colabeling, and does not take advantage of the growing number of in vivo fluorophores available. To address these limitations, we developed a genetic strategy to allow detailed visualization of single podocytes and their foot processes by conventional fluorescence microscopy. We generated a transgenic mouse line expressing a GFP-Cre-ERT2 fusion protein under the control of the collagen α1(I) promoter with strong podocyte expression. Administration of submaximal tamoxifen allowed genetic labeling of single podocytes when crossed with a Cre-reporter line. Of three different reporter systems that we evaluated for the ability to reveal fine structural details of podocytes, bigenic Coll1α1GCE;Gt(ROSA)26Sor(tm9(CAG-tdTomato)) mice allowed podocyte labeling with a strong and homogeneous reporter signal that was easily observed by epifluorescence. We could easily detect anatomic features of podocytes down to tertiary foot processes, and we were able to visualize and quantitate ultrastructural changes to foot processes after podocyte injury. In summary, using this method of genetic labeling and conventional fluorescence microscopy to visualize podocyte foot processes will complement electron microscopy and facilitate the analysis of podocytes and their precursors in vivo.


Assuntos
Microscopia de Fluorescência/métodos , Podócitos/fisiologia , Animais , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
15.
Pediatr Nephrol ; 27(2): 183-93, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21311912

RESUMO

Chronic kidney diseases (CKD), independent of their primary cause, lead to progressive, irreversible loss of functional renal parenchyma. Renal pathology in CKD is characterized by tubulointerstitial fibrosis with excessive matrix deposition produced by myofibroblasts. Because blocking the formation of these scar-forming cells represents a logical therapeutic target for patients with progressive fibrotic kidney disease, the origin of renal myofibroblasts is a subject of intense investigation. Although the traditional view holds that resident fibroblasts are the myofibroblast precursor, for the last 10 years, injured epithelial cells have been thought to directly contribute to the myofibroblast pool by the process of epithelial-to-mesenchymal transition (EMT). The recent application of genetic fate mapping techniques in mouse fibrosis models has provided new insights into the cell hierarchies in fibrotic kidney disease and results cast doubt on the concept that EMT is a source of myofibroblast recruitment in vivo, but rather point to the resident pericyte/perivascular fibroblast as the myofibroblast progenitor pool. This review will highlight recent findings arguing against EMT as a direct contributor to the kidney myofibroblast population and review the use of genetic fate mapping to elucidate the cellular mechanisms of kidney homeostasis and disease.


Assuntos
Nefropatias/patologia , Rim/patologia , Miofibroblastos/fisiologia , Doença Crônica , Transição Epitelial-Mesenquimal , Fibroblastos/fisiologia , Humanos , Células-Tronco/fisiologia , Fator de Crescimento Transformador beta/fisiologia
16.
Proc Natl Acad Sci U S A ; 106(34): 14518-23, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19706538

RESUMO

Proliferation of interstitial fibroblasts is a hallmark of progressive renal fibrosis commonly resulting in chronic kidney failure. The intermediate-conductance Ca(2+)-activated K(+) channel (K(Ca)3.1) has been proposed to promote mitogenesis in several cell types and contribute to disease states characterized by excessive proliferation. Here, we hypothesized that K(Ca)3.1 activity is pivotal for renal fibroblast proliferation and that deficiency or pharmacological blockade of K(Ca)3.1 suppresses development of renal fibrosis. We found that mitogenic stimulation up-regulated K(Ca)3.1 in murine renal fibroblasts via a MEK-dependent mechanism and that selective blockade of K(Ca)3.1 functions potently inhibited fibroblast proliferation by G(0)/G(1) arrest. Renal fibrosis induced by unilateral ureteral obstruction (UUO) in mice was paralleled by a robust up-regulation of K(Ca)3.1 in affected kidneys. Mice lacking K(Ca)3.1 (K(Ca)3.1(-/-)) showed a significant reduction in fibrotic marker expression, chronic tubulointerstitial damage, collagen deposition and alphaSMA(+) cells in kidneys after UUO, whereas functional renal parenchyma was better preserved. Pharmacological treatment with the selective K(Ca)3.1 blocker TRAM-34 similarly attenuated progression of UUO-induced renal fibrosis in wild-type mice and rats. In conclusion, our data demonstrate that K(Ca)3.1 is involved in renal fibroblast proliferation and fibrogenesis and suggest that K(Ca)3.1 may represent a therapeutic target for the treatment of fibrotic kidney disease.


Assuntos
Fibroblastos/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Rim/efeitos dos fármacos , Pirazóis/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Fibrose/etiologia , Fibrose/prevenção & controle , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/fisiologia , Rim/metabolismo , Rim/patologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Obstrução Ureteral/complicações
17.
Circulation ; 119(17): 2323-32, 2009 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-19380617

RESUMO

BACKGROUND: It has been proposed that activation of endothelial SK3 (K(Ca)2.3) and IK1 (K(Ca)3.1) K+ channels plays a role in the arteriolar dilation attributed to an endothelium-derived hyperpolarizing factor (EDHF). However, our understanding of the precise function of SK3 and IK1 in the EDHF dilator response and in blood pressure control remains incomplete. To clarify the roles of SK3 and IK1 channels in the EDHF dilator response and their contribution to blood pressure control in vivo, we generated mice deficient for both channels. METHODS AND RESULTS: Expression and function of endothelial SK3 and IK1 in IK1(-/-)/SK3(T/T) mice was characterized by patch-clamp, membrane potential measurements, pressure myography, and intravital microscopy. Blood pressure was measured in conscious mice by telemetry. Combined IK1/SK3 deficiency in IK1(-/-)/SK3(T/T) (+doxycycline) mice abolished endothelial K(Ca) currents and impaired acetylcholine-induced smooth muscle hyperpolarization and EDHF-mediated dilation in conduit arteries and in resistance arterioles in vivo. IK1 deficiency had a severe impact on acetylcholine-induced EDHF-mediated vasodilation, whereas SK3 deficiency impaired NO-mediated dilation to acetylcholine and to shear stress stimulation. As a consequence, SK3/IK1-deficient mice exhibited an elevated arterial blood pressure, which was most prominent during physical activity. Overexpression of SK3 in IK1(-/-)/SK3(T/T) mice partially restored EDHF- and nitric oxide-mediated vasodilation and lowered elevated blood pressure. The IK1-opener SKA-31 enhanced EDHF-mediated vasodilation and lowered blood pressure in SK3-deficient IK1(+/+)/SK3(T/T) (+doxycycline) mice to normotensive levels. CONCLUSIONS: Our study demonstrates that endothelial SK3 and IK1 channels have distinct stimulus-dependent functions, are major players in the EDHF pathway, and significantly contribute to arterial blood pressure regulation. Endothelial K(Ca) channels may represent novel therapeutic targets for the treatment of hypertension.


Assuntos
Fatores Biológicos/fisiologia , Hipertensão/etiologia , Vasodilatação , Animais , Fatores Biológicos/metabolismo , Pressão Sanguínea/fisiologia , Cálcio/metabolismo , Potenciais da Membrana , Camundongos , Camundongos Knockout , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/fisiologia , Canais de Potássio Shaw/deficiência , Canais de Potássio Ativados por Cálcio de Condutância Baixa/deficiência
18.
Br J Pharmacol ; 157(4): 509-26, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19302590

RESUMO

The arterial endothelium critically contributes to blood pressure control by releasing vasodilating autacoids such as nitric oxide, prostacyclin and a third factor or pathway termed 'endothelium-derived hyperpolarizing factor' (EDHF). The nature of EDHF and EDHF-signalling pathways is not fully understood yet. However, endothelial hyperpolarization mediated by the Ca(2+)-activated K(+) channels (K(Ca)) has been suggested to play a critical role in initializing EDHF-dilator responses in conduit and resistance-sized arteries of many species including humans. Endothelial K(Ca) currents are mediated by the two K(Ca) subtypes, intermediate-conductance K(Ca) (KCa3.1) (also known as, a.k.a. IK(Ca)) and small-conductance K(Ca) type 3 (KCa2.3) (a.k.a. SK(Ca)). In this review, we summarize current knowledge about endothelial KCa3.1 and KCa2.3 channels, their molecular and pharmacological properties and their specific roles in endothelial function and, particularly, in the EDHF-dilator response. In addition we focus on recent experimental evidences derived from KCa3.1- and/or KCa2.3-deficient mice that exhibit severe defects in EDHF signalling and elevated blood pressures, thus highlighting the importance of the KCa3.1/KCa2.3-EDHF-dilator system for blood pressure control. Moreover, we outline differential and overlapping roles of KCa3.1 and KCa2.3 for EDHF signalling as well as for nitric oxide synthesis and discuss recent evidence for a heterogeneous (sub) cellular distribution of KCa3.1 (at endothelial projections towards the smooth muscle) and KCa2.3 (at inter-endothelial borders and caveolae), which may explain their distinct roles for endothelial function. Finally, we summarize the interrelations of altered KCa3.1/KCa2.3 and EDHF system impairments with cardiovascular disease states such as hypertension, diabetes, dyslipidemia and atherosclerosis and discuss the therapeutic potential of KCa3.1/KCa2.3 openers as novel types of blood pressure-lowering drugs.


Assuntos
Fatores Biológicos/fisiologia , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/fisiopatologia , Descoberta de Drogas , Endotélio Vascular/fisiopatologia , Canais de Potássio Cálcio-Ativados/biossíntese , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia , Vasodilatadores/uso terapêutico , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Modelos Biológicos , Canais de Potássio Cálcio-Ativados/agonistas , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Canais de Potássio Cálcio-Ativados/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Vasodilatadores/farmacologia
19.
Pflugers Arch ; 458(2): 291-302, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19037656

RESUMO

Gardos channel, the erythrocyte Ca(2+)-activated K(+) channel (K(Ca)3.1), is considered a major regulator of red blood cell (RBC) volume by mediating efflux of potassium and thus cell dehydration and shrinkage. However, the functional importance of K(Ca)3.1 in RBC in vivo is incompletely understood. Here, we used K(Ca)3.1(-/-)-mice to investigate the consequences of K(Ca)3.1 deficiency for RBC indices, functions, and sequestration. RBCs of K(Ca)3.1(-/-)-mice of all ages were mildly macrocytic but their biconcave appearance being preserved. RBC number, total hemoglobin, and hematocrit were unchanged in the adult K(Ca)3.1(-/-)-mice and increased in the premature K(Ca)3.1(-/-)-mice. Filterability, Ca(2+)-dependent volume decrease and osmotic tolerance of RBCs lacking K(Ca)3.1 were noticeably reduced when compared to RBC of wild-type littermates. Deformability to increasing shear stress was unchanged. Strikingly, K(Ca)3.1(-/-)-mice developed progressive splenomegaly which was considerable ( approximately 200% of controls) in the >6-month-old mice and was paralleled by increased iron deposition in the aged mice presumably as a consequence of enhanced RBC sequestration. Daily injections of the K(Ca)3.1-blocker TRAM-34 (120 mg/kg) also produced mild splenomegaly in wild-type mice. We conclude that genetic deficit of erythroid K(Ca)3.1 causes mild RBC macrocytosis, presumably leading to reduced filterability, and impairs volume regulation. These RBC defects result in mild but progressive splenomegaly.


Assuntos
Deformação Eritrocítica/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/fisiologia , Esplenomegalia/etiologia , Animais , Morte Celular/efeitos dos fármacos , Tamanho Celular , Contagem de Eritrócitos , Índices de Eritrócitos/efeitos dos fármacos , Eritrócitos , Citometria de Fluxo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/deficiência , Ferro/metabolismo , Camundongos , Pirazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA