Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(10): 4502-4507, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30765526

RESUMO

With-no-lysine (WNK) kinases regulate renal sodium-chloride cotransporter (NCC) to maintain body sodium and potassium homeostasis. Gain-of-function mutations of WNK1 and WNK4 in humans lead to a Mendelian hypertensive and hyperkalemic disease pseudohypoaldosteronism type II (PHAII). X-ray crystal structure and in vitro studies reveal chloride ion (Cl-) binds to a hydrophobic pocket within the kinase domain of WNKs to inhibit its activity. The mechanism is thought to be important for physiological regulation of NCC by extracellular potassium. To test the hypothesis that WNK4 senses the intracellular concentration of Cl- physiologically, we generated knockin mice carrying Cl--insensitive mutant WNK4. These mice displayed hypertension, hyperkalemia, hyperactive NCC, and other features fully recapitulating human and mouse models of PHAII caused by gain-of-function WNK4. Lowering plasma potassium levels by dietary potassium restriction increased NCC activity in wild-type, but not in knockin, mice. NCC activity in knockin mice can be further enhanced by the administration of norepinephrine, a known activator of NCC. Raising plasma potassium by oral gavage of potassium inactivated NCC within 1 hour in wild-type mice, but had no effect in knockin mice. The results provide compelling support for the notion that WNK4 is a bona fide physiological intracellular Cl- sensor and that Cl- regulation of WNK4 underlies the mechanism of regulation of NCC by extracellular potassium.


Assuntos
Cloretos/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Camundongos , Camundongos Transgênicos , Potássio/administração & dosagem , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Pseudo-Hipoaldosteronismo/genética
2.
J Physiol ; 595(16): 5573-5586, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28555925

RESUMO

KEY POINTS: The highly variable phenotypes observed in patients with classic Bartter's syndrome (BS) remain unsatisfactorily explained. The wide spectrum of functional severity of CLCNKB mutations may contribute to the phenotypic variability, and the genotype-phenotype association has not been established. Low-level expression of the human ClC-Kb channel in mammalian cells impedes the functional study of CLCNKB mutations, and the underlying cause is still unclear. The human ClC-Kb channel is highly degraded by proteasome in human embryonic kidney cells. The C-terminal in-frame green fluorescent protein fusion may slow down the proteasome-mediated proteolysis. Barttin co-expression necessarily improves the stability, membrane trafficking and gating of ClC-Kb. CLCNKB mutations in barttin-binding sites, dimer interface or selectivity filter often have severe functional consequences. The remaining chloride conductance of the ClC-Kb mutant channel significantly correlates with the phenotypes, such as age at diagnosis, plasma chloride concentration, and the degree of calciuria in patients with classic BS. ABSTRACT: Mutations in the CLCNKB gene encoding the human voltage-gated chloride ClC-Kb (hClC-Kb) channel cause classic Bartter's syndrome (BS). In contrast to antenatal BS, classic BS manifests with highly variable phenotypes. The functional severity of the mutant channel has been proposed to explain this phenomenon. Due to difficulties in the expression of hClC-Kb in heterologous expression systems, the functional consequences of mutant channels have not been thoroughly examined, and the genotype-phenotype association has not been established. In this study, we found that hClC-Kb, when expressed in human embryonic kidney (HEK) cells, was unstable due to degradation by proteasome. In-frame fusion of green fluorescent protein (GFP) to the C-terminus of the channel may ameliorate proteasome degradation. Co-expression of barttin increased protein abundance and membrane trafficking of hClC-Kb and markedly increased functional chloride current. We then functionally characterized 18 missense mutations identified in our classic BS cohort and others using HEK cells expressing hClC-Kb-GFP. Most CLCNKB mutations resulted in marked reduction in protein abundance and chloride current, especially those residing at barttin binding sites, dimer interface and selectivity filter. We enrolled classic BS patients carrying homozygous missense mutations with well-described functional consequences and clinical presentations for genotype-phenotype analysis. We found significant correlations of mutant chloride current with the age at diagnosis, plasma chloride concentration and urine calcium excretion rate. In conclusion, hClC-Kb expression in HEK cells is susceptible to proteasome degradation, and fusion of GFP to the C-terminus of hClC-Kb improves protein expression. The functional severity of the CLCNKB mutation is an important determinant of the phenotype in classic BS.


Assuntos
Síndrome de Bartter/genética , Canais de Cloreto/genética , Adolescente , Adulto , Criança , Canais de Cloreto/fisiologia , Células HEK293 , Humanos , Lactente , Pessoa de Meia-Idade , Mutação , Fenótipo , Adulto Jovem
3.
J Biol Chem ; 286(31): 27425-35, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21665951

RESUMO

Hypokalemic periodic paralysis (hypoKPP) is characterized by episodic flaccid paralysis of muscle and acute hypokalemia during attacks. Familial forms of hypoKPP are predominantly caused by mutations of either voltage-gated Ca(2+) or Na(+) channels. The pathogenic gene mutation in non-familial hypoKPP, consisting mainly of thyrotoxic periodic paralysis (TPP) and sporadic periodic paralysis (SPP), is largely unknown. Recently, mutations in KCNJ18, which encodes a skeletal muscle-specific inwardly rectifying K(+) channel Kir2.6, were reported in some TPP patients. Whether mutations of Kir2.6 occur in other patients with non-familial hypoKPP and how mutations of the channel predispose patients to paralysis are unknown. Here, we report one conserved heterozygous mutation in KCNJ18 in two TPP patients and two separate heterozygous mutations in two SPP patients. These mutations result in V168M, R43C, and A200P amino acid substitution of Kir2.6, respectively. Compared with the wild type channel, whole-cell currents of R43C and V168M mutants were reduced by ∼78 and 43%, respectively. No current was detected for the A200P mutant. Single channel conductance and open probability were reduced for R43C and V168M, respectively. Biotinylation assays showed reduced cell surface abundance for R43C and A200P. All three mutants exerted dominant negative inhibition on wild type Kir2.6 as well as wild type Kir2.1, another Kir channel expressed in the skeletal muscle. Thus, mutations of Kir2.6 are associated with SPP as well as TPP. We suggest that decreased outward K(+) current from hypofunction of Kir2.6 predisposes the sarcolemma to hypokalemia-induced paradoxical depolarization during attacks, which in turn leads to Na(+) channel inactivation and inexcitability of muscles.


Assuntos
Paralisia Periódica Hipopotassêmica/genética , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Adulto , Sequência de Aminoácidos , Sequência de Bases , Primers do DNA , Feminino , Humanos , Masculino , Dados de Sequência Molecular
4.
Kidney Int ; 79(7): 730-41, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21228764

RESUMO

We have identified a novel homozygous nonsense mutation (W516X) in the kidney-type electrogenic sodium bicarbonate cotransporter 1 (NBC1) in a patient with isolated proximal renal tubular acidosis (pRTA). To specifically address the pathogenesis of this mutation, we created NBC1 W516X knock-in mice to match the patient's abnormalities. The expression of NBC1 mRNA and protein in the kidneys of NBC1(W516X/W516X) mice were virtually absent, indicating that nonsense-mediated mRNA decay (NMD) is involved in the defective transcription and translation of this mutation. These mice not only recapitulated the phenotypes of this patient with growth retardation, pRTA, and ocular abnormalities, but also showed anemia, volume depletion, prerenal azotemia, and several organ abnormalities, culminating in dehydration and renal failure with early lethality before weaning. In isolated renal proximal tubules, both NBC1 activity and the rate of bicarbonate absorption were markedly reduced. Unexpectedly, there was no compensatory increase in mRNA of distal acid/base transporters. Sodium bicarbonate but not saline administration to these mutant mice markedly prolonged their survival, decreased their protein catabolism and attenuated organ abnormalities. The prolonged survival time uncovered the development of corneal opacities due to corneal edema. Thus, NBC1(W516X/W516X) mice with pRTA represent an animal model for metabolic acidosis and may be useful for testing therapeutic inhibition of NMD in vivo.


Assuntos
Acidose Tubular Renal/metabolismo , Acidose/metabolismo , Túbulos Renais Proximais/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Acidose/tratamento farmacológico , Acidose/genética , Acidose/patologia , Acidose Tubular Renal/tratamento farmacológico , Acidose Tubular Renal/genética , Acidose Tubular Renal/patologia , Fatores Etários , Envelhecimento , Análise de Variância , Anemia/genética , Anemia/metabolismo , Animais , Aquaporina 2/metabolismo , Bicarbonatos/metabolismo , Códon sem Sentido , Opacidade da Córnea/genética , Opacidade da Córnea/metabolismo , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Genótipo , Transtornos do Crescimento/genética , Transtornos do Crescimento/metabolismo , Homozigoto , Humanos , Concentração de Íons de Hidrogênio , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Fenótipo , RNA Mensageiro/metabolismo , Índice de Gravidade de Doença , Bicarbonato de Sódio/administração & dosagem , Bicarbonato de Sódio/metabolismo , Simportadores de Sódio-Bicarbonato/genética , Transcrição Gênica
5.
J Am Soc Nephrol ; 21(11): 1868-77, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20813865

RESUMO

Polymorphisms in the gene encoding sterile 20/SPS1-related proline/alanine-rich kinase (SPAK) associate with hypertension susceptibility in humans. SPAK interacts with WNK kinases to regulate the Na(+)-K(+)-2Cl(-) and Na(+)-Cl(-) co-transporters [collectively, N(K)CC]. Mutations in WNK1/4 and N(K)CC can cause changes in BP and dyskalemia in humans, but the physiologic role of SPAK in vivo is unknown. We generated and analyzed SPAK-null mice by targeting disruption of exons 9 and 10 of SPAK. Compared with SPAK(+/+) littermates, SPAK(+/-) mice exhibited hypotension without significant electrolyte abnormalities, and SPAK(-/-) mice not only exhibited hypotension but also recapitulated Gitelman syndrome with hypokalemia, hypomagnesemia, and hypocalciuria. In the kidney tissues of SPAK(-/-) mice, the expression of total and phosphorylated (p-)NCC was markedly decreased, but that of p-OSR1, total NKCC2, and p-NKCC2 was significantly increased. We observed a blunted response to thiazide but normal response to furosemide in SPAK(-/-) mice. In aortic tissues, total NKCC1 expression was increased but p-NKCC1 was decreased in SPAK-deficient mice. Both SPAK(+/-) and SPAK(-/-) mice had impaired responses to the selective α(1)-adrenergic agonist phenylephrine and the NKCC1 inhibitor bumetanide, suggesting that impaired aortic contractility may contribute to the hypotension of SPAK-null mice. In summary, SPAK-null mice have defects of NCC in the kidneys and NKCC1 in the blood vessels, leading to hypotension through renal salt wasting and vasodilation. SPAK may be a promising target for antihypertensive therapy.


Assuntos
Aorta/fisiopatologia , Síndrome de Gitelman/metabolismo , Síndrome de Gitelman/fisiopatologia , Proteínas Serina-Treonina Quinases/deficiência , Vasoconstrição/fisiologia , Animais , Aorta/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Modelos Animais de Doenças , Diuréticos/farmacologia , Feminino , Furosemida/farmacologia , Hidroclorotiazida/farmacologia , Hipotensão/metabolismo , Hipotensão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/genética , Simportadores de Cloreto de Sódio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Vasoconstrição/efeitos dos fármacos
6.
JCI Insight ; 6(20)2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34499620

RESUMO

The prevailing view is that the ClC-Ka chloride channel (mouse Clc-k1) functions in the thin ascending limb to control urine concentration, whereas the ClC-Kb channel (mouse Clc-k2) functions in the thick ascending limb (TAL) to control salt reabsorption. Mutations of ClC-Kb cause classic Bartter syndrome, characterized by renal salt wasting, with perinatal to adolescent onset. We studied the roles of Clc-k channels in perinatal mouse kidneys using constitutive or inducible kidney-specific gene ablation and 2D and advanced 3D imaging of optically cleared kidneys. We show that Clc-k1 and Clc-k2 were broadly expressed and colocalized in perinatal kidneys. Deletion of Clc-k1 and Clc-k2 revealed that both participated in NKCC2- and NCC-mediated NaCl reabsorption in neonatal kidneys. Embryonic deletion of Clc-k2 caused tubular injury and impaired renal medulla and TAL development. Inducible deletion of Clc-k2 beginning after medulla maturation produced mild salt wasting resulting from reduced NCC activity. Thus, both Clc-k1 and Clc-k2 contributed to salt reabsorption in TAL and distal convoluted tubule (DCT) in neonates, potentially explaining the less-severe phenotypes in classic Bartter syndrome. As opposed to the current understanding that salt wasting in adult patients with Bartter syndrome is due to Clc-k2 deficiency in adult TAL, our results suggest that it originates mainly from defects occurring in the medulla and TAL during development.


Assuntos
Proteínas de Transporte de Ânions/deficiência , Síndrome de Bartter/genética , Canais de Cloreto/deficiência , Medula Renal/crescimento & desenvolvimento , Animais , Feminino , Humanos , Camundongos , Gravidez
7.
Hum Mutat ; 31(12): 1304-15, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20848653

RESUMO

Gitelman syndrome (GS) is characterized by salt-losing hypotension, hypomagnesemia, hypokalemic metabolic alkalosis, and hypocalciuria. To better model human GS caused by a specific mutation in the thiazide-sensitive Na(+) -Cl(-) cotransporter (NCC) gene SLC12A3, we generated a nonsense Ncc Ser707X knockin mouse corresponding to human p.Ser710X (c.2135C>A), a recurrent mutation with severe phenotypes in Chinese GS patients. Compared with wild-type or heterozygous littermates, homozygous (Hom) knockin mice fully recapitulated the phenotype of human GS. The markedly reduced Ncc mRNA and virtually absent Ncc protein expression in kidneys of Hom mice was primarily due to nonsense-mediated mRNA decay (NMD) surveillance mechanisms. Expression of epithelial Na(+) channel (Enac), Ca(2+) channels (Trpv5 and Trpv6), and K(+) channels (Romk1 and maxi-K) were significantly increased. Late distal convoluted tubules (DCT) volume was increased and DCT cell ultrastructure appeared intact. High K(+) intake could not correct hypokalemia but caused a further increase in maxi-K but not Romk1 expression. Renal tissue from a patient with GS also showed the enhanced TRPV5 and ROMK1 expression in distal tubules. We suggest that the upregulation of TRPV5/6 and of ROMK1 and Maxi-K may contribute to hypocalciuria and hypokalemia in Ncc Ser707X knockin mice and human GS, respectively.


Assuntos
Modelos Animais de Doenças , Técnicas de Introdução de Genes/métodos , Síndrome de Gitelman/genética , Receptores de Droga/genética , Serina/genética , Simportadores/genética , Tiazidas/farmacologia , Animais , Sequência de Bases , Canais de Cálcio/metabolismo , Tamanho Celular/efeitos dos fármacos , Códon sem Sentido/genética , Dieta , Feminino , Imunofluorescência , Síndrome de Gitelman/patologia , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Rim/ultraestrutura , Masculino , Camundongos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fenótipo , Cloreto de Potássio/administração & dosagem , Cloreto de Potássio/farmacologia , Estabilidade de RNA/efeitos dos fármacos , Estabilidade de RNA/genética , Receptores de Droga/metabolismo , Sódio/metabolismo , Membro 3 da Família 12 de Carreador de Soluto , Simportadores/metabolismo , Adulto Jovem
8.
Eur J Pediatr ; 168(9): 1129-33, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19050915

RESUMO

BACKGROUND: Progressive renal failure in patients with classic Bartter's syndrome (cBS) due to inactivating mutations in CLCNKB gene is extraordinarily rare. DISCUSSION: We describe a 17-year-old Chinese boy who presented with progressive muscle weakness and renal failure. He was diagnosed as BS of unknown type at the age of 9 months and treated with indomethacin (2 mg/kg/day) and potassium chloride (KCl) supplementation (1.5 mEq/kg/day) for hypokalemia (2.5 mmol/l). At the age of 12 years, serum K+ was 3.0 mmol/l and creatinine reached 2.0 mg/dl. On admission, his blood pressure was normal but volume status was depleted. Urinalysis was essentially normal. Biochemical studies showed hypokalemia (K+ 2.4 mmol/l) with a high transtubular K+ gradient (TTKG) 9.6, metabolic alkalosis (HCO3- 28.4 mmol/l), normomagnesemia (2.0 mg/dl), severe renal failure (BUN 94 mg/dl, Cr 6.3 mg/dl), and hypocalciuria (urine calcium/creatinine ratio 0.02 mg/mg). Abdominal sonography revealed bilateral small size kidneys without nephrocalcinosis or renal stones. After the withdrawal of indomethacin with regular KCl and adequate fluid supplementation for 1 year, serum creatinine and K+ levels have been maintained at 4.0 mg/dl and 3.3 mmol/l, respectively. Direct sequencing of NKCC2, ROMK, ClC-Kb, and NCCT in this patient disclosed a novel homozygous missense mutation (GGG to GAG, G470E) in CLCNKB. This G470E mutation was not identified in 100 healthy Chinese subjects. Long-term therapy of non-steroidal anti-inflammatory drugs (NSAIDs), prolonged hypokalemia, chronic volume depletion, and underlying genetic variety may contribute to the deterioration of his renal function. The cautious use of NSAIDs, aggressive correction of hypokalemia, and avoidance of severe volume depletion may prevent the irreversible renal damage in patients with BS due to a Cl- channel defect.


Assuntos
Canais de Cloreto/genética , Falência Renal Crônica/genética , Mutação Puntual/genética , Simportadores de Cloreto de Sódio-Potássio/genética , Adolescente , Progressão da Doença , Humanos , Hipopotassemia/genética , Masculino , Mutação de Sentido Incorreto/genética , Membro 1 da Família 12 de Carreador de Soluto , Síndrome
9.
Am J Med Sci ; 343(4): 281-5, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21841462

RESUMO

INTRODUCTION: Sporadic periodic paralysis (SPP), the second leading cause of hypokalemic periodic paralysis (HPP) in Asia, has a presentation similar to that of familial periodic paralysis (FPP) and is caused by gene mutations in the calcium (Ca(2+)) (CACNA1S) and sodium (Na(+)) (SCN4A) channels of skeletal muscle. The authors determined whether SPP shares similar genotype and phenotype with FPP. METHODS: Sixty SPP patients who did not have a family history of paralysis, abnormal thyroid function tests and other identifiable causes of HPP, and 8 FPP patients were enrolled. Genomic DNA was isolated from blood leukocytes of all SPP and FPP patients. Genetic analysis of whole S4 segment in CACNA1S and SCN4A was performed. Phenotypic analysis included clinical presentations, laboratory data and precipitating events. RESULTS: All FPP patients had mutations in either CACNA1S or SCN4A, but only 4 SPP patients had de novo mutations in CACNA1S (R1239H) and SCN4A (R669×2, R1135H). SPP patients with de novo mutations manifested a phenotype indistinguishable from that of FPP patients except a later age of onset. SPP patients without mutations also had a later age of onset, significantly fewer attacks of paralysis than FPP patients, and unidentifiable precipitating factors. CONCLUSION: A minority of SPP patients had de novo CACNA1S or SCN4A mutations and may have a variant of FPP. The majority of SPP patients, those without mutations in CACNA1S and SCN4A, represent a unique subgroup of HPP patients, and this form of SPP usually manifests at a later age, is associated with fewer attacks and lacks apparent triggering factors.


Assuntos
Canais de Cálcio/genética , Genótipo , Paralisia Periódica Hipopotassêmica/genética , Fenótipo , Canais de Sódio/genética , Adolescente , Adulto , Canais de Cálcio Tipo L , Criança , Pré-Escolar , Feminino , Humanos , Paralisia Periódica Hipopotassêmica/diagnóstico , Masculino , Canal de Sódio Disparado por Voltagem NAV1.4 , Paralisias Periódicas Familiares/diagnóstico , Paralisias Periódicas Familiares/genética , Mutação Puntual/genética , Adulto Jovem
10.
Clin J Am Soc Nephrol ; 6(3): 630-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21051746

RESUMO

BACKGROUND AND OBJECTIVES: Gitelman's syndrome (GS) is an autosomal recessive renal tubular disorder caused by mutations in the SLC12A3 gene encoding the thiazide-sensitive Na(+)-Cl(-) cotransporter (NCC). Despite meticulous sequencing of genomic DNA, approximately one-third of GS patients are negative or heterozygotes for the known mutations. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Because blood leukocytes express NCC mRNA, we evaluate whether deep intronic mutations contribute to GS patients with uniallelic or undetectable SLC12A3 mutations. Twenty-nine patients with GS (men/women = 16/13), including eight negative and 21 uniallelic SLC12A3 mutations from 19 unrelated families, and normal controls were enrolled in an academic medical center. Analysis of cDNA from blood leukocytes, sequencing of the corresponding introns of genomic DNA for abnormal transcript, and analysis of NCC protein expression from renal biopsy were performed. RESULTS: We identified nine Taiwan aboriginal patients carrying c.1670-191C→T mutations in intron 13 and 10 nonaboriginal patients carrying c.2548+253C→T mutations in intron 21 from 14 families (14/19). These two mutations undetected in 100 healthy subjects created pseudoexons containing new premature termination codons. Haplotype analysis with markers flanking SLC12A3 revealed that both mutations did not have founder effects. Apical NCC expression in the DCT of renal tissue was markedly diminished in two patients carrying deep intronic mutations. CONCLUSIONS: Deep intronic mutations in SLC12A3 causing defective NCC expression can be identified with the RNA-based approach in patients with GS. c.1670-191C→T and c.2548+253C→T are hot spot mutations that can be screened in GS patients with uniallelic or negative SLC12A3 mutations.


Assuntos
Síndrome de Gitelman/genética , Íntrons , Mutação , Receptores de Droga/genética , Simportadores/genética , Centros Médicos Acadêmicos , Adulto , Sequência de Bases , Biópsia , Estudos de Casos e Controles , Análise Mutacional de DNA , Feminino , Predisposição Genética para Doença , Síndrome de Gitelman/metabolismo , Haplótipos , Humanos , Rim/química , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem , Fenótipo , Receptores de Droga/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Membro 3 da Família 12 de Carreador de Soluto , Simportadores/análise , Taiwan
11.
Transl Res ; 150(4): 216-22, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17900509

RESUMO

Focal segmental glomerulosclerosis (FSGS) is characterized by typical sclerosis but also shows other non-sclerotic lesions that provide prognostic informations. The glomerular epithelial hyperplasia lesion (EPHL) that develops earlier than the sclerotic lesions is a key determinant of progression of FSGS. However, the relationship among EPHL, glomeular sclerosis, and macrophage infiltration in FSGS is unclear, and the EPHL-associated markers helpful for prognosis of FSGS have still not been completely identified. Here, we performed clinicopathologic, immunochemical, and molecular analyses to examine whether osteopontin (OPN), a macrophage chemokine, is an injury marker of EPHLs correlating with glomerular sclerosis and macrophage mobilization. First, the FSGS model was induced in Balb/c mice by a single injection of adriamycin, and consecutive sclerosis changes were evaluated. In parallel, we used reverse transcription-polymerase chain reaction and Western blot analyses to determine levels of OPN in isolated glomeruli and urine, respectively. Immunohistochemistry was applied to assess the OPN expression in EPHLs and macrophage infiltration around the glomeruli. Our results showed that, within glomeruli, OPN expressed restrictedly within EPHL; the OPN mRNA and protein of glomeruli increased on day 11, correlating well with the early EPHL, and following sclerosis and macrophage infiltration. In addition, immunohistochemistry (IHC) staining of OPN greatly highlighted early glomerular EPHLs, helping microscopic identification of EPHLs. We propose that the OPN expression in EPHLs could contribute to the progression of FSGS by recruiting macrophage toward the compromised glomeruli. Detection of OPN in glomeruli and urine could be helpful in prognosis of FSGS.


Assuntos
Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/patologia , Glomérulos Renais/patologia , Rim/patologia , Osteopontina/análise , Animais , Biomarcadores/análise , Biomarcadores/urina , Western Blotting , Modelos Animais de Doenças , Doxorrubicina , Epitélio/metabolismo , Epitélio/patologia , Feminino , Regulação da Expressão Gênica , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/urina , Hiperplasia/induzido quimicamente , Hiperplasia/metabolismo , Hiperplasia/patologia , Imuno-Histoquímica , Rim/química , Rim/efeitos dos fármacos , Glomérulos Renais/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Osteopontina/urina , Prognóstico , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa