Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Semin Cell Dev Biol ; 91: 119-131, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-29857053

RESUMO

There is a global epidemic of chronic kidney disease (CKD) characterized by a progressive loss of nephrons, ascribed in large part to a rising incidence of hypertension, metabolic syndrome, and type 2 diabetes mellitus. There is a ten-fold variation in nephron number at birth in the general population, and a 50% overall decrease in nephron number in the last decades of life. The vicious cycle of nephron loss stimulating hypertrophy by remaining nephrons and resulting in glomerulosclerosis has been regarded as maladaptive, and only partially responsive to angiotensin inhibition. Advances over the past century in kidney physiology, genetics, and development have elucidated many aspects of nephron formation, structure and function. Parallel advances have been achieved in evolutionary biology, with the emergence of evolutionary medicine, a discipline that promises to provide new insight into the treatment of chronic disease. This review provides a framework for understanding the origins of contemporary developmental nephrology, and recent progress in evolutionary biology. The establishment of evolutionary developmental biology (evo-devo), ecological developmental biology (eco-devo), and developmental origins of health and disease (DOHaD) followed the discovery of the hox gene family, the recognition of the contribution of cumulative environmental stressors to the changing phenotype over the life cycle, and mechanisms of epigenetic regulation. The maturation of evolutionary medicine has contributed to new investigative approaches to cardiovascular disease, cancer, and infectious disease, and promises the same for CKD. By incorporating these principles, developmental nephrology is ideally positioned to answer important questions regarding the fate of nephrons from embryo through senescence.


Assuntos
Biologia do Desenvolvimento/métodos , Evolução Molecular , Néfrons/metabolismo , Insuficiência Renal Crônica/genética , Animais , Biologia do Desenvolvimento/tendências , Epigênese Genética/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Néfrons/citologia , Néfrons/embriologia , Organogênese/genética , Insuficiência Renal Crônica/embriologia , Insuficiência Renal Crônica/patologia
2.
Am J Physiol Renal Physiol ; 308(10): F1155-66, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25694483

RESUMO

Cystinosis is an inherited disorder resulting from a mutation in the CTNS gene, causing progressive proximal tubular cell flattening, the so-called swan-neck lesion (SNL), and eventual renal failure. To determine the role of oxidative stress in cystinosis, histologic sections of kidneys from C57BL/6 Ctns(-/-) and wild-type mice were examined by immunohistochemistry and morphometry from 1 wk to 20 mo of age. Additional mice were treated from 1 to 6 mo with vehicle or mitoquinone (MitoQ), an antioxidant targeted to mitochondria. The leading edge of the SNL lost mitochondria and superoxide production, and became surrounded by a thickened tubular basement membrane. Progression of the SNL as determined by staining with lectin from Lotus tetragonolobus accelerated after 3 mo, but was delayed by treatment with MitoQ (38 ± 4% vs. 28 ± 1%, P < 0.01). Through 9 mo, glomeruli had retained renin staining and intact macula densa, whereas SNL expressed transgelin, an actin-binding protein, but neither kidney injury molecule-1 (KIM-1) nor cell death was observed. After 9 mo, clusters of proximal tubules exhibited localized oxidative stress (4-hydroxynonenal binding), expressed KIM-1, and underwent apoptosis, leading to the formation of atubular glomeruli and accumulation of interstitial collagen. We conclude that nephron integrity is initially maintained in the Ctns(-/-) mouse by adaptive flattening of cells of the SNL through loss of mitochondria, upregulation of transgelin, and thickened basement membrane. This adaptation ultimately fails in adulthood, with proximal tubular disruption, formation of atubular glomeruli, and renal failure. Antioxidant treatment targeted to mitochondria delays initiation of the SNL, and may provide therapeutic benefit in children with cystinosis.


Assuntos
Adaptação Fisiológica/fisiologia , Cistinose/patologia , Cistinose/fisiopatologia , Túbulos Renais Proximais/patologia , Túbulos Renais Proximais/fisiopatologia , Estresse Oxidativo/fisiologia , Sistemas de Transporte de Aminoácidos Neutros/deficiência , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Cistinose/genética , Modelos Animais de Doenças , Feminino , Receptor Celular 1 do Vírus da Hepatite A , Túbulos Renais Proximais/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mutação/genética , Compostos Organofosforados/farmacologia , Superóxidos/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/farmacologia
3.
Am J Pathol ; 184(7): 1957-66, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24815352

RESUMO

In polycystic kidney disease (PKD), renal parenchyma is destroyed by cysts, hypothesized to obstruct nephrons. A signature of unilateral ureteral obstruction, proximal tubular atrophy leads to formation of atubular glomeruli. To determine whether this process occurs in PKD, kidneys from pcy mice (moderately progressive PKD), kidneys from cpk mice (rapidly progressive PKD), and human autosomal dominant PKD were examined in early and late stages. Integrity of the glomerulotubular junction and proximal tubular mass were determined in sections stained with Lotus tetragonolobus lectin. Development of proximal tubular atrophy and atubular glomeruli was determined in serial sections of individual glomeruli. In pcy mice, most glomerulotubular junctions were normal at 20 weeks, but by 30 weeks, 56% were atrophic and 25% of glomeruli were atubular; glomerulotubular junction integrity decreased with increasing cyst area (r = 0.83, P < 0.05). In cpk mice, all glomerulotubular junctions were normal at 10 days, but by 19 days, 26% had become abnormal. In early-stage autosomal dominant PKD kidneys, 50% of glomeruli were atubular or attached to atrophic tubules; in advanced disease, 100% were abnormal. Thus, proximal tubular injury in cystic kidneys closely parallels that observed with ureteral obstruction. These findings support the hypothesis that, in renal cystic disorders, cyst-dependent obstruction of medullary and cortical tubules initiates a process culminating in widespread destruction of proximal convoluted tubules at the glomerulotubular junction.


Assuntos
Glomérulos Renais/patologia , Túbulos Renais Proximais/patologia , Doenças Renais Policísticas/patologia , Obstrução Ureteral/complicações , Adulto , Animais , Cistos , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Pediatr Nephrol ; 29(4): 537-41, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23949631

RESUMO

Most chronic kidney disease in children results from congenital or inherited disorders, which can be studied in mouse models. Following 2 weeks of unilateral ureteral obstruction (UUO) in the adult mouse, nephron loss is due to proximal tubular mitochondrial injury and cell death. In neonatal mice, proximal tubular cell death is delayed beyond 2 weeks of complete UUO, and release of partial UUO allows remodeling of remaining nephrons. Progressive cyst expansion develops in polycystic kidney disease (PKD), a common inherited renal disorder. The polycystic kidney and fibrosis (pcy)-mutant mouse (which develops late-onset PKD) develops thinning of the glomerulotubular junction in parallel with growth of cysts in adulthood. Renal insufficiency in nephropathic cystinosis, a rare inherited renal disorder, results from progressive tubular cystine accumulation. In the Ctns knockout mouse (a model of cystinosis), proximal tubular cells become flattened, with loss of mitochondria and thickening of tubular basement membrane. In each model, persistent obstructive or metabolic stress leads ultimately to the formation of atubular glomeruli. The initial "fight" response (proximal tubular survival) switches to a "flight" response (proximal tubular cell death) with ongoing oxidative injury and mitochondrial damage. Therapies should be directed at reducing proximal tubular mitochondrial oxidative injury to enhance repair and regeneration.


Assuntos
Nefropatias/congênito , Nefropatias/patologia , Túbulos Renais Proximais/lesões , Túbulos Renais Proximais/patologia , Animais , Humanos
6.
Pediatr Nephrol ; 26(9): 1381-5, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21222000

RESUMO

Congenital urinary tract obstruction is a major cause of progressive renal disease in children. We developed a model of partial unilateral ureteral obstruction (UUO) in the neonatal mouse, in which nephrogenesis at birth is similar to that of the midtrimester human fetus. The proximal tubule responds to UUO by undergoing apoptosis and necrosis, likely due to mitochondrial sensitivity to hypoxia and reactive oxygen species in the face of reduced endogenous antiapoptotic factors such as eNOS. Damage to the glomerulotubular junction is followed by scission and formation of atubular glomeruli and aglomerular tubules. This is an orchestrated process, with atubular glomeruli surrounded by a continuous layer of regenerated parietal epithelial cells. Relief of UUO at 7 days of age results in remodeling of the renal parenchyma by adulthood. In contrast to proximal tubular destruction, collecting ducts remain dilated and patent, with remodeling due to apoptosis and proliferation (a process associated with recruitment of intercalated cells as progenitor cells following UUO in the fetal monkey). Formation of atubular glomeruli occurs in other renal disorders (congenital nephrotic syndrome and cystinosis), and may represent a maladaptive response to proximal tubular injury reflecting an evolutionary adaptation by an ancestor we share with aglomerular marine fish.


Assuntos
Glomérulos Renais/patologia , Túbulos Renais Proximais/patologia , Obstrução Ureteral/patologia , Animais , Animais Recém-Nascidos , Apoptose , Doença Crônica , Modelos Animais de Doenças , Progressão da Doença , Humanos , Glomérulos Renais/metabolismo , Túbulos Renais Proximais/metabolismo , Camundongos , Necrose , Obstrução Ureteral/complicações , Obstrução Ureteral/metabolismo
7.
Am J Physiol Renal Physiol ; 298(1): F62-71, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19889956

RESUMO

To investigate the role of endogenous inducible nitric oxide synthase (iNOS) in the response of the developing kidney to unilateral ureteral obstruction (UUO), neonatal iNOS null mutant (-/-) and wild-type (WT) mice were subjected to partial or complete UUO. At 7 and 21 days of age, apoptosis, renin, vascular endothelial growth factor (VEGF), fibroblasts (anti-fibroblast-specific peptide 1), myofibroblasts (alpha-smooth muscle actin), macrophages (F4/80), and collagen were measured in kidney tissue. Compared with WT, renal parenchymal thickness was increased, with preservation of the papilla, in -/- mice with partial UUO, but decreased in -/- mice with complete UUO. Ureteral peristalsis increased with severity of pelvic dilatation in WT, and increased further in -/- mice with partial UUO. Apoptosis, fibroblasts, and macrophages were increased in -/- mice with complete UUO, but there was no effect of iNOS on other histological parameters following complete UUO. Renin was decreased in -/- mice with partial UUO. There was no effect of iNOS genotype on renal collagen accumulation at either 7 or 21 days of age. These results are consistent with an injurious role for endogenous iNOS following partial UUO by inhibiting ureteral peristalsis and increasing renal renin although renal fibrosis is not affected. In contrast, in mice with complete UUO, iNOS attenuates apoptosis and enhances renal parenchymal thickness. Alterations in the severity of ureteral obstruction may therefore influence the effect of iNOS on long-term renal injury.


Assuntos
Animais Recém-Nascidos/metabolismo , Hidronefrose/metabolismo , Hidronefrose/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia , Actinas/metabolismo , Animais , Apoptose/fisiologia , Colágeno/metabolismo , Modelos Animais de Doenças , Feminino , Fibroblastos/patologia , Rim/metabolismo , Rim/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Renina/metabolismo , Índice de Gravidade de Doença
8.
J Am Soc Nephrol ; 19(2): 197-206, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18199796

RESUMO

Functional nephrons can be lost through a process of glomerulotubular disconnection. Progressive development of atubular glomeruli seems to play a major role in a number of renal disorders, including glomerular diseases, ascribed to injury to the glomerulotubular junction as a result of proteinuria; however, formation of atubular glomeruli is even more common in tubulointerstitial disorders, such as obstructive nephropathy. Toxic nephropathy is also associated with the formation of atubular glomeruli, suggesting susceptibility of the glomerulotubular junction to toxic injury. Narrowing or other abnormalities of the glomerulotubular junction are described as precursors of glomerulotubular disconnection. Cystinosis represents a dramatic example of progressive injury to the glomerulotubular junction, with formation of the "swan-neck deformity" following degenerative tubular cell changes attributable to apoptosis. Significant numbers of atubular glomeruli have been reported in chronic pyelonephritis and renal allograft rejection; this suggests interstitial inflammation as a stimulus for the formation of atubular glomeruli. Because of difficulties in morphologic recognition, it is likely that glomerulotubular disconnection is an underappreciated mechanism in the progression of renal disease. A better understanding of the vulnerability of the glomerulotubular junction and its protection from injury should lead to better strategies for preserving renal function in many nephropathies.


Assuntos
Nefropatias/patologia , Glomérulos Renais/patologia , Túbulos Renais/patologia , Néfrons/patologia , Animais , Progressão da Doença , Humanos , Nefropatias/fisiopatologia , Glomérulos Renais/fisiopatologia , Túbulos Renais/fisiopatologia , Néfrons/fisiopatologia
9.
Am J Physiol Renal Physiol ; 293(3): F895-903, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17626154

RESUMO

In rodent models of obstructive nephropathy, exogenous epidermal growth factor (EGF) attenuates tubule cell death in rats and exacerbates cell death in mice. To identify species differences in EGF receptor (EGFR) regulation and signaling, cell lysates were prepared from rat, mouse, and human proximal tubule cells (PTC) and compared by immunoblot analysis for expression and phosphorylation of Src and EGFR. Frozen kidney tissue was also analyzed. Results indicate mouse PTC have constitutive Src- and EGFR-kinase activities not detected in rat or human PTC. Immunoblots of rat, mouse, and human kidney homogenates confirmed this finding in vivo. Src-specific inhibitor PP2 and EGFR kinase inhibitor AG1478 decreased EGF-induced apoptosis in mouse PTC by 74% (P < 0.001) and 70% (P < 0.001), respectively. Expression of a constitutive Src mutant cDNA in rat PTC rendered cells susceptible to EGF-induced death. EGF decreased stretch-induced apoptosis by 66% (P < 0.001) relative to vehicle control in human PTC, similar to rat PTC response. We conclude that elevated Src activity in mouse tubular cells alters downstream EGFR signaling and increases susceptibility to EGF-induced cell death. The unexpected finding that a therapeutic agent (EGF) in rats is detrimental in mice underscores the importance of determining which animal best represents the response of human kidneys to a given agent.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Rim/metabolismo , Quinases da Família src/metabolismo , Animais , Humanos , Rim/efeitos dos fármacos , Rim/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Especificidade da Espécie
10.
Am J Physiol Renal Physiol ; 293(1): F262-8, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17442727

RESUMO

The renin-angiotensin system is activated in the developing kidney and is necessary for normal renal development, but is further activated by unilateral ureteral obstruction (UUO). During nephrogenesis, there is a switch from a preponderance of angiotensin AT(2) to AT(1) receptors in the rat. We examined the renal cellular response to angiotensin II receptor inhibition in the neonatal rat subjected to partial UUO under anesthesia within 48 h of birth. Group I ("early") received saline vehicle, losartan (AT(1) inhibitor), or PD-123319 (AT(2) inhibitor) during the completion of nephrogenesis in the first 10 days of life. Group II ("late") received each of the three treatments throughout the subsequent 10 days of life. Kidneys were harvested at 21 days, and the distribution of renin, apoptosis, macrophages, alpha-smooth muscle actin, and collagen was determined. Losartan and PD-123319 each increased vascular renin distribution in both kidneys. Partial UUO reduced growth and increased apoptosis, macrophages, alpha-smooth muscle actin, and collagen in the obstructed kidney. Early losartan treatment further increased alpha-smooth muscle actin and collagen in the obstructed kidney and induced apoptosis, macrophages, and collagen in the contralateral kidney. Late losartan treatment had no effect on any of the parameters in either kidney, and PD-123319 had no effect on either kidney. We conclude that selective inhibition of AT(1) receptors during nephrogenesis (but not during subsequent renal maturation) exacerbates injury to the obstructed kidney and also injures the contralateral kidney. These results suggest that angiotensin II receptor blockers should be avoided in the developing hydronephrotic kidney.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/toxicidade , Animais Recém-Nascidos/fisiologia , Nefropatias/etiologia , Nefropatias/patologia , Obstrução Ureteral/complicações , Envelhecimento/fisiologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Colágeno/metabolismo , Feminino , Imuno-Histoquímica , Rim/patologia , Losartan/toxicidade , Macrófagos/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Sódio/fisiologia , Regulação para Cima/efeitos dos fármacos
11.
Am J Physiol Renal Physiol ; 292(1): F168-74, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16788139

RESUMO

Chronic unilateral ureteral obstruction (UUO) in the neonatal rat causes delayed renal maturation, tubular apoptosis, and interstitial inflammation. Vascular endothelial growth factor (VEGF) acts as a survival factor for tubular cells and reduces renal injury in several models of renal disease. To determine whether exogenous VEGF attenuates renal injury from UUO, rats were subjected within the first 48 h of life to sham operation, partial UUO, or complete UUO. Saline vehicle or VEGF(121) (50 mg/kg) was injected twice daily for 7 days, after which kidneys were harvested for histological study. The density of peritubular capillaries was measured with platelet-endothelial cell adhesion molecule-1 immunostaining, proliferating nuclei were detected by proliferating-cell nuclear antigen staining, apoptosis by the transferase-mediated dUTP nick end-labeling technique, macrophages by ED-1 immunostaining, and collagen by Sirius red staining. Glomerular number and maturation index were also determined in each group. Following chronic complete UUO in the neonatal rat, peritubular capillary density was significantly decreased. Cortical capillary density was further reduced by exogenous VEGF in the partially obstructed kidney. While UUO also decreased glomerular number and delayed glomerular maturation, exogenous VEGF exerted no additional effects. Cellular proliferation and tubular apoptosis increased in proportion to the severity of obstruction, but exogenous VEGF had no additional effects on proliferation, tubular apoptosis, or macrophage infiltration. However, VEGF reduced interstitial apoptosis in the kidney with partial UUO. We conclude that VEGF does not have salutary effects on the renal lesions caused by chronic UUO in the neonatal rat and may actually worsen obstructive nephropathy by aggravating the interstitial lesions.


Assuntos
Animais Recém-Nascidos/fisiologia , Rim/metabolismo , Obstrução Ureteral/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Apoptose/efeitos dos fármacos , Capilares/patologia , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Rim/crescimento & desenvolvimento , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/patologia , Tamanho do Órgão/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Ratos , Ratos Sprague-Dawley
12.
Am J Physiol Renal Physiol ; 292(1): F158-67, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16788140

RESUMO

Obstructive nephropathy constitutes a major cause of renal impairment in children. Chronic unilateral ureteral obstruction (UUO) impairs maturation of the developing kidney and leads to tubular apoptosis and interstitial inflammation. Vascular endothelial growth factor (VEGF) is involved in recovery from various forms of renal injury. We questioned whether the renal expression of endogenous VEGF and its receptor (VEGFR2/Flk-1) is modified by UUO in early development. Neonatal rats were subjected to partial or complete UUO or sham operation. The distribution of immunoreactive VEGF in each kidney was examined after 7, 14, or 28 days. Adult rats were also subjected to sham operation or complete UUO. Tubular VEGF increased between 14 and 28 days in sham-operated rats and in some partially obstructed neonatal rats but decreased with complete UUO. Parallel changes were found by Western blotting, but not by RT-PCR. Immunoreactive VEGF colocalized with mitochondria in proximal and distal tubules and also appeared in type A intercalated cells, glomerular vascular endothelium, and podocytes. While neonatal microvascular renal VEGFR2 receptor staining was strongly positive regardless of UUO, staining was weak in sham-operated adults but increased following UUO. Parallel changes in VEGFR2 expression were verified by RT-PCR and Western blotting. We conclude that endogenous renal VEGF is developmentally regulated in the neonatal rat and is differentially regulated by partial and complete UUO. Following UUO in the adult, the VEGF receptor is upregulated. Endogenous VEGF may serve an adaptive role in responding to tubular injury caused by UUO and may modulate adaptation by the contralateral kidney.


Assuntos
Animais Recém-Nascidos/fisiologia , Nefropatias/metabolismo , Nefropatias/patologia , Rim/metabolismo , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Western Blotting , Gliceraldeído-3-Fosfato Desidrogenases/biossíntese , Gliceraldeído-3-Fosfato Desidrogenases/genética , Imuno-Histoquímica , Rim/patologia , Córtex Renal/metabolismo , Nefropatias/etiologia , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , ATPases Translocadoras de Prótons/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Obstrução Ureteral/complicações , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
13.
Am J Physiol Renal Physiol ; 292(3): F946-55, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17107943

RESUMO

Congenital urinary tract obstruction is the most important cause of renal insufficiency in infants and children, and angiotensin-converting enzyme (ACE) inhibitors attenuate the progression of renal disease in adults. ACE inhibitors are increasingly utilized in children with progressive renal disease. Because angiotensin is necessary for normal renal development, we examined the effects of ACE inhibition both during and immediately following the period of postnatal nephrogenesis in the neonatal rat subjected to sham operation or partial unilateral ureteral obstruction (UUO) under general anesthesia within the first 48 h of life. Rats in group I received enalapril 30 mg/kg body wt (or vehicle) daily for the first 10 days, while in group II, the 10 days of treatment began 10 days after surgery. Kidneys were harvested at day 21 and analyzed for apoptosis (TUNEL), interstitial macrophages (ED-1 immunohistochemistry), myofibroblasts (alpha-smooth muscle actin), and collagen (Sirius red). Partial UUO delayed glomerular maturation and increased ipsilateral renal macrophage infiltration, alpha-smooth muscle actin and Sirius red staining. In group I, enalapril increased myofibroblast accumulation in sham-operated kidneys, but not in obstructed kidneys. In contrast, in group II, enalapril further increased macrophage, myofibroblast, and collagen accumulation following partial UUO. The relative abundance of components of the kallikrein-kinin system, measured by Western blot, was not altered by partial UUO in the 14- and 28-day-old rat. Thus, in contrast to its salutary effects at later ages, ACE inhibition can worsen injury to the partially obstructed kidney during renal maturation even after the completion of nephrogenesis.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Nefropatias/etiologia , Rim/efeitos dos fármacos , Obstrução Ureteral/complicações , Actinas/metabolismo , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Colágeno/metabolismo , Contraindicações , Enalapril/farmacologia , Rim/metabolismo , Rim/patologia , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Córtex Renal/patologia , Nefropatias/patologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Medula Renal/metabolismo , Medula Renal/patologia , Pelve Renal/efeitos dos fármacos , Pelve Renal/patologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Cininogênios/metabolismo , Macrófagos/patologia , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor B2 da Bradicinina/metabolismo , Calicreínas Teciduais/metabolismo
14.
Nat Clin Pract Nephrol ; 2(3): 157-68, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16932414

RESUMO

Obstructive nephropathy is a major cause of renal failure, particularly in infants and children. Cellular and molecular mechanisms responsible for the progression of the tubular atrophy and interstitial fibrosis-processes that lead to nephron loss-have been elucidated in the past 5 years. Following urinary tract obstruction and tubular dilatation, a cascade of events results in upregulation of the intrarenal renin-angiotensin system, tubular apoptosis and macrophage infiltration of the interstitium. This is followed by accumulation of interstitial fibroblasts through proliferation of resident fibroblasts and epithelial-mesenchymal transformation of renal tubular cells. Under the influence of cytokines, chemokines and other signaling molecules produced by tubular and interstitial cells, fibroblasts undergo transformation to myofibroblasts that induce expansion of the extracellular matrix. The cellular interactions that regulate development of interstitial inflammation, tubular apoptosis and interstitial fibrosis are complex. Changes in renal gene expression and protein production afford many potential biomarkers of disease progression and targets for therapeutic manipulation. These include signaling molecules and receptors involved in macrophage recruitment and proliferation, tubular death signals and survival factors, and modulators of epithelial-mesenchymal transformation. Targeted gene deletion and various forms of gene therapy have been used in experimental obstructive nephropathy, mostly rodent models of unilateral ureteral obstruction or cell culture techniques. Further refinement of these models is needed to develop a matrix of biomarkers with clinical predictive value, as well as molecular therapies that will prevent or reverse the renal structural and functional consequences of obstructive nephropathy.


Assuntos
Obstrução Ureteral/urina , Apoptose , Biomarcadores/urina , Quimiocina CCL7 , Fator de Crescimento Epidérmico/urina , Fibrose , Terapia Genética , Humanos , Inflamação/etiologia , Rim/patologia , Proteínas Quimioatraentes de Monócitos/urina , Fator de Crescimento Transformador beta/urina , Fator de Necrose Tumoral alfa/urina , Obstrução Ureteral/complicações , Obstrução Ureteral/congênito , Obstrução Ureteral/terapia
15.
Kidney Int ; 68(2): 504-14, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16014027

RESUMO

BACKGROUND: Epidermal growth factor (EGF) markedly attenuates tubular apoptosis induced by unilateral ureteral obstruction (UUO) in the neonatal rat, and reduces apoptosis induced by mechanical stretch of cultured rat tubular cells. METHODS: To investigate the role of EGF in modulating apoptosis resulting from UUO, neonatal wild type and mutant mice lacking EGF (knockout), or with diminished EGF receptor activity (waved-2 mutant) were compared to control mice for tubular apoptosis and atrophy. Rat and mouse kidneys were compared for localization of the EGF receptor. Apoptosis was also measured in cultured mouse tubular cells subjected to stretch and exposed to EGF. RESULTS: UUO reduced endogenous renal EGF expression in wild-type mice. Unlike the rat, exogenous EGF did not decrease tubular apoptosis or atrophy in the obstructed kidney, and significantly increased stretch-induced apoptosis of cultured mouse tubular cells. Tubular apoptosis was 50% lower in the obstructed kidney of EGF knockout and waved-2 mice relative to wild type and heterozygous animals. Exogenous EGF increased tubular apoptosis and doubled atrophy in the obstructed kidney of waved-2 mice. Species differences in EGF receptor localization were detected in 3-day-old kidneys. CONCLUSION: EGF acts as a survival factor in the neonatal rat, but potentiates tubular cell death in the neonatal mouse. Species differences are maintained in cultured cells, suggesting that differences in EGF receptor signaling underlie these opposing effects.


Assuntos
Apoptose/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Hidronefrose/patologia , Hidronefrose/fisiopatologia , Animais , Animais Recém-Nascidos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Receptores ErbB/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Especificidade da Espécie
16.
Nephron Exp Nephrol ; 100(3): e113-23, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15824514

RESUMO

BACKGROUND: Renal interstitial inflammation is a consequence of unilateral ureteral obstruction (UUO). Following ischemia/reperfusion, adenosine reduces renal inflammation and injury, effects which are potentiated by type 4 phosphodiesterase inhibitors. We therefore studied the effects of A2A adenosine receptor agonist (ATL146e), and PDE4 inhibitor (rolipram) in mice subjected to UUO. METHODS: Mice were subjected to UUO or sham operation, and received either vehicle or ATL146e + rolipram by osmotic minipump for 1 or 7 days. At 1, 3, 7, or 14 days after operation, renal macrophage infiltration, apoptosis, proliferation, tubular atrophy, and interstitial fibrosis were quantitated, and expressions of IL-6 and TGF-beta mRNA were determined. RESULTS: ATL146e + rolipram reduced macrophage infiltration by 40% after 3 days UUO (p < 0.05). Tubular apoptosis, tubular atrophy, and interstitial fibrosis were increased by 7 or 14 days UUO, but were unaffected by ATL146e + rolipram. However, cellular proliferation was increased by ATL146e + rolipram in the obstructed kidney. ATL146e + rolipram had no effect on the renal expression of IL-6 and TGF-beta mRNA. CONCLUSIONS: A2A receptor activation and PDE4 inhibition transiently reduce renal macrophage infiltration, but do not ameliorate the renal response to UUO. We speculate that the persistent stimulus for inflammation triggered by UUO cannot be reversed by agents that suppress inflammatory cell activation alone.


Assuntos
Ácidos Cicloexanocarboxílicos/farmacologia , Inflamação/fisiopatologia , Nefropatias/fisiopatologia , Inibidores de Fosfodiesterase/farmacologia , Purinas/farmacologia , Receptores A2 de Adenosina/fisiologia , Rolipram/farmacologia , Obstrução Ureteral/complicações , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , 3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Animais , Apoptose , Proliferação de Células , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Fibrose , Camundongos , Camundongos Endogâmicos C57BL , Receptores A2 de Adenosina/efeitos dos fármacos
17.
Semin Perinatol ; 28(2): 124-31, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15200251

RESUMO

Significant advances have been made recently in elucidating the cellular consequences of urinary tract obstruction during renal development. Urinary tract obstruction impairs growth and maturation of the kidney, and can also cause renal maldevelopment. This includes a reduction in the number of nephrons, tubular atrophy, and progressive interstitial fibrosis. Apoptosis (programmed cell death) accounts for much of the loss of tubular epithelial cells. Factors contributing to apoptosis include stretching of cells in dilated tubules, altered renal production of growth factors, and infiltration of the renal interstitium by macrophages. Two major controversies remain regarding the surgical management of congenital obstructive nephropathy: first, which fetuses with bladder outlet obstruction should undergo prenatal intervention, and second, which infants should undergo early pyeloplasty for ureteropelvic junction obstruction? Even after successful surgery for congential obstructive nephropathy, all patients should be followed for hypertension, proteinuria, or renal deterioration.


Assuntos
Obstrução Ureteral/congênito , Obstrução Uretral/congênito , Obstrução do Colo da Bexiga Urinária/congênito , Animais , Apoptose , Modelos Animais de Doenças , Feminino , Humanos , Hipertensão , Recém-Nascido , Gravidez , Proteinúria , Obstrução Ureteral/complicações , Obstrução Ureteral/patologia , Obstrução Ureteral/cirurgia , Obstrução Uretral/complicações , Obstrução Uretral/patologia , Obstrução Uretral/cirurgia , Obstrução do Colo da Bexiga Urinária/complicações , Obstrução do Colo da Bexiga Urinária/patologia , Obstrução do Colo da Bexiga Urinária/cirurgia
18.
Pediatr Nephrol ; 18(4): 335-41, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12700958

RESUMO

Macrophages play important roles during renal inflammation. Infiltrating macrophages produce proinflammatory mediators and induce apoptosis in a variety of target cells. Because proximal tubules are frequently damaged in inflammatory processes, we investigated murine macrophages (J774) in the induction of apoptosis in murine PKSV-PR proximal tubule cells. PKSV-PR cells were co-cultured with activated or non-activated macrophages. Apoptosis was assessed by Annexin-V-FITC/propidium iodide staining and analyzed by fluorescence-activated cell sorting. Macrophages were separated from tubule cells with transwell membranes to distinguish soluble factor-mediated from direct cell-to-cell contact-mediated apoptosis. Cell supernatants from activated and non-activated macrophages were analyzed for induction of apoptosis. Activated (but not non-activated) macrophages induced tubule cell apoptosis in co-culture. Soluble factors were mainly responsible for induction of apoptosis; membrane separation and transfer of cell supernatant from activated macrophages showed similar levels of apoptosis induction. Although tumor necrosis factor (TNF)-alpha and transforming growth factor (TGF)-beta(1), measured by ELISA, increased significantly in supernatants from activated macrophages, blocking TNF-alpha and TGF-beta did not decrease apoptosis in PKSV-PR cells co-cultured with macrophages. Moreover, exogenous addition of TNF-alpha, TGF-beta, anti-Fas antibody, or TRAIL failed to induce apoptosis in tubule cells. We conclude that inflammatory macrophages mediate proximal tubule cell death, directing apoptosis mainly via release of unidentified soluble factors.


Assuntos
Apoptose/fisiologia , Túbulos Renais Proximais/citologia , Macrófagos/fisiologia , Animais , Proteínas Reguladoras de Apoptose , Linhagem Celular , Técnicas de Cocultura , Meios de Cultivo Condicionados , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Ativação de Macrófagos/fisiologia , Glicoproteínas de Membrana/farmacologia , Camundongos , Frações Subcelulares/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/farmacologia
19.
Kidney Int ; 63(2): 564-75, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12631121

RESUMO

BACKGROUND: To elucidate the sequence of renal responses leading to nephron loss in obstructive nephropathy, we examined the evolution of segmental nephron cellular changes consequent to chronic unilateral ureteral obstruction (UUO) in the neonatal mouse. METHODS: Neonatal mice were subjected to UUO or sham-operation, and kidneys were harvested 5, 12 or 19 days after surgery. Proximal tubules (PT), distal tubules (DT) and collecting ducts (CD) were identified with lectins. Histomorphometric quantitation was made for cellular necrosis, apoptosis, proliferation, tubular dilatation, tubular basement membrane (TBM) thickening, interstitial collagen, and glomerular maturation. The distribution of hypoxic tissue was determined using pimonidazole as a marker. Additional studies were performed by mechanically stretching monolayer cultures of mouse proximal tubular and collecting duct cells, and measuring apoptosis. RESULTS: Neonatal UUO induced an arrest of glomerular maturation throughout the period of study. Chronic UUO induced hypoxia, tubular necrosis, proliferation, and TBM thickening in the PT, but stimulated apoptosis in the DT and CD. Tubular dilation in the obstructed kidney was most severe in CD and least severe in PT. Tubular cell apoptosis closely paralleled tubular dilation (P < 0.05), and fibrosis surrounding individual tubules also correlated with tubular dilation (P < 0.001). Mechanical stretching of cultured mouse tubular cells induced apoptosis directly proportional to the magnitude of axial strain: apoptosis was consistently greater in CD than in PT cells (P < 0.05). CONCLUSIONS: Following UUO, the co-localization of hypoxia with cellular proliferation, necrosis, and TBM thickening of the PT is consistent with ischemic injury resulting from vasoconstriction. In contrast, a selective dilation of the distal portion of the nephron (DT and CD), which results from the greater tubular compliance there, leads to stretch-induced epithelial cell apoptosis, along with a progressive peritubular fibrosis. Nephron loss in the obstructed developing kidney likely results from complex, segment-specific cellular responses.


Assuntos
Apoptose , Túbulos Renais/patologia , Túbulos Renais/fisiopatologia , Néfrons/patologia , Néfrons/fisiopatologia , Obstrução Ureteral/patologia , Obstrução Ureteral/fisiopatologia , Animais , Animais Recém-Nascidos , Membrana Basal/patologia , Divisão Celular , Feminino , Fibrose , Hipóxia/etiologia , Hipóxia/patologia , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/fisiopatologia , Masculino , Camundongos , Necrose , Obstrução Ureteral/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA