Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Sci Rep ; 11(1): 12903, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145342

RESUMO

Abdominal aortic aneurysm (AAA) is a complex disease which is incompletely accounted for. Basement membrane (BM) Collagen IV (COL4A1/A2) is abundant in the artery wall, and several lines of evidence indicate a protective role of baseline COL4A1/A2 in AAA development. Using Col4a1/a2 hemizygous knockout mice (Col4a1/a2+/-, 129Svj background) we show that partial Col4a1/a2 deficiency augmented AAA formation. Although unchallenged aortas were morphometrically and biomechanically unaffected by genotype, explorative proteomic analyses of aortas revealed a clear reduction in BM components and contractile vascular smooth muscle cell (VSMC) proteins, suggesting a central effect of the BM in maintaining VSMCs in the contractile phenotype. These findings were translated to human arteries by showing that COL4A1/A2 correlated to BM proteins and VSMC markers in non-lesioned internal mammary arteries obtained from coronary artery bypass procedures. Moreover, in human AAA tissue, MYH11 (VSMC marker) was depleted in areas of reduced COL4 as assessed by immunohistochemistry. Finally, circulating COL4A1 degradation fragments correlated with AAA progression in the largest Danish AAA cohort, suggesting COL4A1/A2 proteolysis to be an important feature of AAA formation. In sum, we identify COL4A1/A2 as a critical regulator of VSMC phenotype and a protective factor in AAA formation.


Assuntos
Aneurisma da Aorta Abdominal/etiologia , Membrana Basal/metabolismo , Colágeno Tipo IV/deficiência , Predisposição Genética para Doença , Alelos , Animais , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Biomarcadores , Biópsia , Colágeno Tipo IV/genética , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Estudos de Associação Genética , Genótipo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteólise , Proteoma , Proteômica/métodos
2.
PLoS One ; 16(4): e0249909, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33848312

RESUMO

Congenital hearing loss affects 1 in every 1000 births, with genetic mutations contributing to more than 50% of all cases. X-linked nonsyndromic hereditary hearing loss is associated with six loci (DFNX1-6) and five genes. Recently, the missense mutation (c.1771G>A, p.Gly591Ser) in COL4A6, encoding the basement membrane (BM) collagen α6(IV) chain, was shown to be associated with X-linked congenital nonsyndromic hearing loss with cochlear malformation. However, the mechanism by which the COL4A6 mutation impacts hereditary hearing loss has not yet been elucidated. Herein, we investigated Col4a6 knockout (KO) effects on hearing function and cochlear formation in mice. Immunohistochemistry showed that the collagen α6(IV) chain was distributed throughout the mouse cochlea within subepithelial BMs underlying the interdental cells, inner sulcus cells, basilar membrane, outer sulcus cells, root cells, Reissner's membrane, and perivascular BMs in the spiral limbus, spiral ligament, and stria vascularis. However, the click-evoked auditory brainstem response analysis did not show significant changes in the hearing threshold of Col4a6 KO mice compared with wild-type (WT) mice with the same genetic background. In addition, the cochlear structures of Col4a6 KO mice did not exhibit morphological alterations, according to the results of high-resolution micro-computed tomography and histology. Hence, loss of Col4a6 gene expression in mice showed normal click ABR thresholds and normal cochlear formation, which differs from humans with the COL4A6 missense mutation c.1771G>A, p.Gly591Ser. Therefore, the deleterious effects in the auditory system caused by the missense mutation in COL4A6 are likely due to the dominant-negative effects of the α6(IV) chain and/or α5α6α5(IV) heterotrimer with an aberrant structure that would not occur in cases with loss of gene expression.


Assuntos
Cóclea/metabolismo , Colágeno Tipo IV/genética , Surdez/patologia , Animais , Limiar Auditivo , Cóclea/química , Cóclea/diagnóstico por imagem , Cóclea/patologia , Colágeno Tipo IV/deficiência , Surdez/congênito , Surdez/genética , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação de Sentido Incorreto , Fenótipo , Multimerização Proteica , Microtomografia por Raio-X
3.
Am J Physiol Heart Circ Physiol ; 320(5): H1862-H1872, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33769915

RESUMO

There are currently no Food and Drug Administration-approved treatments for heart failure with preserved ejection fraction (HFpEF). Here we compared the effects of exercise with and without α/ß-adrenergic blockade with carvedilol in Col4a3-/- Alport mice, a model of the phenogroup 3 subclass of HFpEF with underlying renal dysfunction. Alport mice were assigned to the following groups: no treatment control (n = 29), carvedilol (n = 11), voluntary exercise (n = 9), and combination carvedilol and exercise (n = 8). Cardiac function was assessed by echocardiography after 4-wk treatments. Running activity of Alport mice was similar to wild types at 1 mo of age but markedly reduced at 2 mo (1.3 ± 0.40 vs. 4.5 ± 1.02 km/day, P < 0.05). There was a nonsignificant trend for increased running activity at 2 mo by carvedilol in the combination treatment group. Combination treatments conferred increased body weight of Col4a3-/- mice (22.0 ± 1.18 vs. 17.8 ± 0.29 g in untreated mice, P < 0.01), suggesting improved physiology, and heart rates declined by similar increments in all carvedilol-treatment groups. The combination treatment improved systolic parameters; stroke volume (30.5 ± 1.99 vs. 17.8 ± 0.77 µL, P < 0.0001) as well as ejection fraction and global longitudinal strain compared with controls. Myocardial performance index was normalized by all interventions (P < 0.0001). Elevated osteopontin plasma levels in control Alport mice were significantly lowered only by combination treatment, and renal function of the Alport group assessed by urine albumin creatinine ratio was significantly improved by all treatments. The results support synergistic roles for exercise and carvedilol to augment cardiac systolic function of Alport mice with moderately improved renal functions but no change in diastole.NEW & NOTEWORTHY In an Alport mouse model of heart failure with preserved ejection fraction (HFpEF), exercise and carvedilol synergistically improved systolic function without affecting diastole. Carvedilol alone or in combination with exercise also improved kidney function. Molecular analyses indicate that the observed improvements in cardiorenal functions were mediated at least in part by effects on serum osteopontin and related inflammatory cytokine cascades. The work presents new potential therapeutic targets and approaches for HFpEF.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Carvedilol/farmacologia , Colágeno Tipo IV/deficiência , Terapia por Exercício , Insuficiência Cardíaca/terapia , Nefrite Hereditária/terapia , Osteopontina/sangue , Disfunção Ventricular Esquerda/terapia , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Autoantígenos/genética , Biomarcadores/sangue , Colágeno Tipo IV/genética , Terapia Combinada , Diástole , Modelos Animais de Doenças , Regulação para Baixo , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Camundongos da Linhagem 129 , Camundongos Knockout , Nefrite Hereditária/sangue , Nefrite Hereditária/genética , Nefrite Hereditária/fisiopatologia , Recuperação de Função Fisiológica , Sístole , Disfunção Ventricular Esquerda/sangue , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/fisiopatologia
4.
Int J Mol Sci ; 21(4)2020 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-32098220

RESUMO

Histone deacetylases have been a target of therapy for organ fibrosis. Here, we report the protective effect of CG200745 (CG), a novel histone deacetylase inhibitor, on tubulointerstitial fibrosis in Col4a3-/- mice, a murine model of Alport syndrome. Morphological analyses revealed CG treatment markedly alleviated kidney fibrosis in Col4a3-/- mice at the age of 7 weeks. CG prevented the activation of transforming growth factor ß (TGFß) and its downstream SMAD signaling in the kidney of Col4a3-/- mice. As critical upstream regulators of TGFß signaling, immunoblotting of whole kidney lysate of Col4a3-/- mice reveled that intra-renal renin-angiotensin system (RAS) was activated with concurrent upregulation of inflammation and apoptosis, which were effectively suppressed by CG treatment. CG suppressed both activation of RAS and up-regulation of TGFß signals in angiotensin II-stimulated HK-2 cells, a human kidney proximal tubular epithelial cell line. CG inhibited activation of TGFß-driven signals and fibrosis in NRK-49F cells, a rat kidney fibroblast cell line, under angiotensin II-rich conditions. Collectively, CG was found to be effective both in proximal tubular epithelial cells by inhibiting local RAS and TGFß signaling activation, as well as in fibroblasts by blocking their transition to myofibroblasts, attenuating renal fibrosis in a murine model of Alport syndrome.


Assuntos
Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Túbulos Renais Proximais/metabolismo , Naftalenos/farmacologia , Nefrite Hereditária , Transdução de Sinais , Animais , Autoantígenos/metabolismo , Linhagem Celular , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/metabolismo , Modelos Animais de Doenças , Fibrose , Humanos , Túbulos Renais Proximais/patologia , Masculino , Camundongos , Camundongos Knockout , Nefrite Hereditária/tratamento farmacológico , Nefrite Hereditária/genética , Nefrite Hereditária/metabolismo , Nefrite Hereditária/patologia , Ratos , Fator de Crescimento Transformador beta/metabolismo
5.
Hum Gene Ther ; 30(7): 865-881, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30808234

RESUMO

Alport syndrome is a genetic disease caused by mutations in type IV collagen and is characterized by progressive kidney disease. The Col4α3-/- mouse model recapitulates the main features of human Alport syndrome. Previously, it was reported that kidney microRNA-21 (miR-21) expression is significantly increased in Col4α3-/- mice, and administration of anti-miR-21 oligonucleotides (anti-miR-21) attenuates kidney disease progression in Col4α3-/- mice, indicating that miR-21 is a viable therapeutic target for Alport syndrome. However, the expression pattern of miR-21 in the kidneys of patients with human Alport syndrome has not been evaluated. Paraffin-embedded kidney specimens were obtained from 27 patients with Alport syndrome and from 10 normal controls. They were evaluated for miR-21 expression and for in situ hybridization and mRNA expression by quantitative polymerase chain reaction. In addition, anti-miR-21 was administrated to Col4α3-/- mice at different stages of disease, and changes in proteinuria, kidney function, and survival were monitored. Transcriptomic analysis of mouse kidney was conducted using RNA sequencing. miR-21 expression was significantly elevated in kidney specimens from patients with Alport syndrome compared to normal controls. Elevated renal miR-21 expression positively correlated with 24 h urine protein, serum blood urea nitrogen, serum creatinine, and severity of kidney pathology. On histological evaluation, high levels of miR-21 were localized to damaged tubular epithelial cells and glomeruli. Kidney specimens from both humans and mice with Alport syndrome exhibited abnormal expression of genes involved in kidney injury, fibrosis, inflammation, mitochondrial function, and lipid metabolism. Administration of anti-miR-21 to Alport mice resulted in slowing of kidney function decline, partial reversal of abnormal gene expression associated with disease pathology, and improved survival. Increased levels of miR-21 in human Alport kidney samples showed a correlation with kidney disease severity measured by proteinuria, biomarkers of kidney function, and kidney histopathology scores. These human data, combined with the finding that a reduction of miR-21 in Col4α3-/- mice improves kidney phenotype and survival, support miR-21 as a viable therapeutic target for the treatment of Alport syndrome.


Assuntos
Regulação da Expressão Gênica , Predisposição Genética para Doença , MicroRNAs/genética , Nefrite Hereditária/genética , Adolescente , Animais , Autoantígenos , Biomarcadores , Biópsia , Criança , Colágeno Tipo IV/deficiência , Modelos Animais de Doenças , Feminino , Fibrose , Perfilação da Expressão Gênica , Estudos de Associação Genética , Humanos , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Knockout , Nefrite Hereditária/diagnóstico , Nefrite Hereditária/metabolismo , Índice de Gravidade de Doença
6.
Am J Physiol Renal Physiol ; 316(5): F830-F837, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30724107

RESUMO

The glomerular basement membrane (GBM) is a critical component of the kidney's blood filtration barrier. Alport syndrome, a hereditary disease leading to kidney failure, is caused by the loss or dysfunction of the GBM's major collagen type IV (COL4) isoform α3α4α5. The constituent COL4 α-chains assemble into heterotrimers in the endoplasmic reticulum before secretion into the extracellular space. If any one of the α3-, α4-, or α5-chains is lost due to mutation of one of the genes, then the entire heterotrimer is lost. Patients with Alport syndrome typically have mutations in the X-linked COL4A5 gene or uncommonly have the autosomal recessive form of the disease due to COL4A3 or COL4A4 mutations. Treatment for Alport syndrome is currently limited to angiotensin-converting enzyme inhibition or angiotensin receptor blockers. Experimental approaches in Alport mice have demonstrated that induced expression of COL4A3, either widely or specifically in podocytes of Col4a3-/- mice, can abrogate disease progression even after establishment of the abnormal GBM. While targeting podocytes in vivo for gene therapy is a significant challenge, the more accessible glomerular endothelium could be amenable for mutant gene repair. In the present study, we expressed COL4A3 in Col4a3-/- Alport mice using an endothelial cell-specific inducible transgenic system, but collagen-α3α4α5(IV) was not detected in the GBM or elsewhere, and the Alport phenotype was not rescued. Our results suggest that endothelial cells do not express the Col4a3/a4/a5 genes and should not be viewed as a target for gene therapy.


Assuntos
Autoantígenos/metabolismo , Colágeno Tipo IV/metabolismo , Células Endoteliais/metabolismo , Terapia Genética , Glomérulos Renais/irrigação sanguínea , Nefrite Hereditária/terapia , Animais , Autoantígenos/genética , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/genética , Modelos Animais de Doenças , Células Endoteliais/patologia , Predisposição Genética para Doença , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Nefrite Hereditária/genética , Nefrite Hereditária/metabolismo , Nefrite Hereditária/patologia , Fenótipo , Subunidades Proteicas
7.
Kidney Int ; 93(1): 147-158, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28843411

RESUMO

We examined activin receptor type IIA (ActRIIA) activation in chronic kidney disease (CKD) by signal analysis and inhibition in mice with Alport syndrome using the ActRIIA ligand trap RAP-011 initiated in 75-day-old Alport mice. At 200 days of age, there was severe CKD and associated Mineral and Bone Disorder (CKD-MBD), consisting of osteodystrophy, vascular calcification, cardiac hypertrophy, hyperphosphatemia, hyperparathyroidism, elevated FGF23, and reduced klotho. The CKD-induced bone resorption and osteoblast dysfunction was reversed, and bone formation was increased by RAP-011. ActRIIA inhibition prevented the formation of calcium apatite deposits in the aortic adventitia and tunica media and significantly decreased the mean aortic calcium concentration from 0.59 in untreated to 0.36 mg/g in treated Alport mice. Aortic ActRIIA stimulation in untreated mice increased p-Smad2 levels and the transcription of sm22α and αSMA. ActRIIA inhibition reversed aortic expression of the osteoblast transition markers Runx2 and osterix. Heart weight was significantly increased by 26% in untreated mice but remained normal during RAP-011 treatment. In 150-day-old mice, GFR was significantly reduced by 55%, but only by 30% in the RAP-011-treated group. In 200-day-old mice, the mean BUN was 100 mg/dl in untreated mice compared to 60 mg/dl in the treated group. In the kidneys of 200-day-old mice, ActRIIA and p-Smad2 were induced and MCP-1, fibronectin, and interstitial fibrosis were stimulated; all were attenuated by RAP-011 treatment. Hence, the activation of ActRIIA signaling during early CKD contributes to the CKD-MBD components of osteodystrophy and cardiovascular disease and to renal fibrosis. Thus, the inhibition of ActRIIA signaling is efficacious in improving and delaying CKD-MBD in this model of Alport syndrome.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Reabsorção Óssea/metabolismo , Cardiomegalia/metabolismo , Distúrbio Mineral e Ósseo na Doença Renal Crônica/metabolismo , Nefrite Hereditária/metabolismo , Insuficiência Renal Crônica/metabolismo , Calcificação Vascular/metabolismo , Actinas/metabolismo , Receptores de Activinas Tipo II/antagonistas & inibidores , Receptores de Activinas Tipo II/genética , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiopatologia , Remodelação Óssea , Reabsorção Óssea/genética , Reabsorção Óssea/fisiopatologia , Reabsorção Óssea/prevenção & controle , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Osso e Ossos/fisiopatologia , Cardiomegalia/genética , Cardiomegalia/fisiopatologia , Cardiomegalia/prevenção & controle , Distúrbio Mineral e Ósseo na Doença Renal Crônica/genética , Distúrbio Mineral e Ósseo na Doença Renal Crônica/fisiopatologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/prevenção & controle , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Modelos Animais de Doenças , Fator de Crescimento de Fibroblastos 23 , Fibrose , Taxa de Filtração Glomerular , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Nefrite Hereditária/tratamento farmacológico , Nefrite Hereditária/genética , Nefrite Hereditária/fisiopatologia , Fosforilação , Proteínas Recombinantes de Fusão/farmacologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/fisiopatologia , Insuficiência Renal Crônica/prevenção & controle , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Transcrição Sp7/metabolismo , Calcificação Vascular/genética , Calcificação Vascular/fisiopatologia , Calcificação Vascular/prevenção & controle , Remodelação Vascular
8.
J Pathol ; 244(4): 408-420, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29266233

RESUMO

Mutations in the α1 (COL4A1) or α2 (COL4A2) chains of collagen type IV, a major component of the vascular basement membrane, cause intracerebral haemorrhages with variable expressivity and reduced penetrance by mechanisms that remain poorly understood. Here we sought to investigate the cellular mechanisms of COL4A1-related intracerebral haemorrhage and identify a marker for haemorrhage risk stratification. A combination of histological, immunohistochemical, and electron microscopy analyses were used to analyse the brain parenchyma, cerebrovasculature, and retinal vessels of mice expressing the disease-causing COL4A1 p.G498V mutation. Mutant mice developed cerebral microhaemorrhages and macroscopic haemorrhages (macrohaemorrhages), the latter with reduced penetrance, mimicking the human disease. Microhaemorrhages that occurred in early postnatal life were associated with a transient, generalized increase in blood-brain barrier permeability at the level of capillaries. Macrohaemorrhages, which occurred later in life, originated from deep brain arteries with focal loss of smooth muscle cells. Similar smooth muscle cell loss was detected in retinal arteries, and a time-course analysis of arterial lesions showed that smooth muscle cells are recruited normally in arterial wall during development, but undergo progressive apoptosis-mediated degeneration. By assessing in parallel the extent of these retinal arterial lesions and the presence/absence of macrohaemorrhages, we found that the arterial lesion load in the retina is strongly correlated with the burden of macrohaemorrhages. We conclude that microhaemorrhages and macrohaemorrhages are driven by two distinct mechanisms. Moreover, smooth muscle cell degeneration is a critical factor underlying the partial penetrance of COL4A1-related macrohaemorrhages, and retinal imaging is a promising tool for identifying high-risk patients. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Hemorragia Cerebral/patologia , Colágeno Tipo IV/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Acidente Vascular Cerebral/patologia , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Proliferação de Células , Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/genética , Modelos Animais de Doenças , Progressão da Doença , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Predisposição Genética para Doença , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/ultraestrutura , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/ultraestrutura , Penetrância , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Artéria Retiniana/metabolismo , Artéria Retiniana/patologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Fatores de Tempo
9.
Sci Rep ; 7(1): 11696, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28916834

RESUMO

The glomerulus exercises its filtration barrier function by establishing a complex filtration apparatus consisting of podocyte foot processes, glomerular basement membrane and endothelial cells. Disruption of any component of the glomerular filtration barrier leads to glomerular dysfunction, frequently manifested as proteinuria. Ultrastructural studies of the glomerulus by transmission electron microscopy (TEM) and conventional scanning electron microscopy (SEM) have been routinely used to identify and classify various glomerular diseases. Here we report the application of newly developed helium ion scanning microscopy (HIM) to examine the glomerulopathy in a Col4a3 mutant/Alport syndrome mouse model. Our study revealed unprecedented details of glomerular abnormalities in Col4a3 mutants including distorted podocyte cell bodies and disorganized primary processes. Strikingly, we observed abundant filamentous microprojections arising from podocyte cell bodies and processes, and presence of unique bridging processes that connect the primary processes and foot processes in Alport mice. Furthermore, we detected an altered glomerular endothelium with disrupted sub-endothelial integrity. More importantly, we were able to clearly visualize the complex, three-dimensional podocyte and endothelial interface by HIM. Our study demonstrates that HIM provides nanometer resolution to uncover and rediscover critical ultrastructural characteristics of the glomerulopathy in Col4a3 mutant mice.


Assuntos
Autoantígenos/genética , Colágeno Tipo IV/genética , Glomerulonefrite/patologia , Glomérulos Renais/ultraestrutura , Animais , Colágeno Tipo IV/deficiência , Células Endoteliais/patologia , Glomérulos Renais/patologia , Lasers de Gás , Camundongos , Camundongos Mutantes/genética , Microscopia Confocal , Podócitos/patologia , Podócitos/ultraestrutura
10.
PLoS One ; 12(5): e0177685, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28542346

RESUMO

Alport syndrome (AS) is a clinically and genetically heterogeneous, progressive nephropathy caused by mutations in COL4A3, COL4A4, and COL4A5, which encode type IV collagen. The large sizes of these genes and the absence of mutation hot spots have complicated mutational analysis by routine polymerase chain reaction (PCR)-based approaches. Here, in order to design a rapid and effective method for the genetic diagnosis of AS, we developed a strategy by utilizing targeted capture associated with next-generation sequencing (NGS) to analyze COL4A3, COL4A4, and COL4A5 simultaneously in 20 AS patients. All the coding exons and flanking sequences of COL4A3, COL4A4, and COL4A5 from the probands were captured followed by HiSeq 2500 sequencing. Candidate mutations were validated by classic Sanger sequencing and quantitative (q)PCR. Sixteen patients (16/20, 75%) showed X-linked inheritance, and four patients (4/20, 20%) showed autosomal recessive inheritance. None of the individuals had autosomal-dominant AS. Fifteen novel mutations, 6 known mutations, and 2 novel fragment deletions were detected by targeted capture and NGS. Of these novel mutations, 12, 3, and 2 mutations were detected in COL4A5, COL4A4, and COL4A3, respectively. A comparison of the clinical manifestations caused by different types of mutations in COL4A5 suggested that nonsense mutations and glycine substitution by an acidic amino acid are more severe than the other missense mutations. Pathogenic mutations were detected in 20 patients. These novel mutations can expand the genotypic spectrum of AS. Our results demonstrated that targeted capture and NGS technology are effective in the genetic diagnosis of AS.


Assuntos
Povo Asiático/genética , Autoantígenos/genética , Colágeno Tipo IV/genética , Mutação , Nefrite Hereditária/genética , Adolescente , Adulto , Criança , Pré-Escolar , China , Colágeno Tipo IV/deficiência , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Deleção de Sequência , Adulto Jovem
11.
Kidney Int ; 91(6): 1347-1361, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28249676

RESUMO

Angiotensin-converting enzyme 2 (ACE2) is a monocarboxypeptidase in the renin-angiotensin system that catalyzes the breakdown of angiotensin II to angiotensin 1-7. We have reported that ACE2 expression in the kidney is reduced in experimental Alport syndrome but the impact of this finding on disease progression has not been studied. Accordingly, we evaluated effects of murine recombinant ACE2 treatment in Col4a3 knockout mice, a model of Alport syndrome characterized by proteinuria and progressive renal injury. Murine recombinant ACE2 (0.5 mg/kg/day) was administered from four to seven weeks of age via osmotic mini-pump. Pathological changes were attenuated by murine recombinant ACE2 treatment which ameliorated kidney fibrosis as shown by decreased expression of COL1α1 mRNA, less accumulation of extracellular matrix proteins, and inhibition of transforming growth factor-ß signaling. Further, increases in proinflammatory cytokine expression, macrophage infiltration, inflammatory signaling pathway activation, and heme oxygenase-1 levels in Col4a3 knockout mice were also reduced by murine recombinant ACE2 treatment. Lastly, murine recombinant ACE2 influenced the turnover of renal ACE2, as it suppressed the expression of tumor necrosis factor-α converting enzyme, a negative regulator of ACE2. Thus, treatment with exogenous ACE2 alters angiotensin peptide metabolism in the kidneys of Col4a3 knockout mice and attenuates the progression of Alport syndrome nephropathy.


Assuntos
Rim/efeitos dos fármacos , Nefrite Hereditária/tratamento farmacológico , Peptidil Dipeptidase A/administração & dosagem , Albuminúria/tratamento farmacológico , Albuminúria/etiologia , Albuminúria/metabolismo , Enzima de Conversão de Angiotensina 2 , Angiotensinas/metabolismo , Animais , Autoantígenos/genética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fibrose , Predisposição Genética para Doença , Mediadores da Inflamação/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrite Hereditária/complicações , Nefrite Hereditária/genética , Nefrite Hereditária/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Proteínas Recombinantes/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo
12.
Kidney Int ; 91(6): 1336-1346, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27927599

RESUMO

Blockers of the renin-angiotensin system are effective in the treatment of experimental and clinical diabetic nephropathy. An approach different from blocking the formation or action of angiotensin II (1-8) that could also be effective involves fostering its degradation. Angiotensin-converting enzyme 2 (ACE2) is a monocarboxypeptidase that cleaves angiotensin II (1-8) to form angiotensin (1-7). Therefore, we examined the renal effects of murine recombinant ACE2 in mice with streptozotocin-induced diabetic nephropathy as well as that of amplification of circulating ACE2 using minicircle DNA delivery prior to induction of experimental diabetes. This delivery resulted in a long-term sustained and profound increase in serum ACE2 activity and enhanced ability to metabolize an acute angiotensin II (1-8) load. In mice with streptozotocin-induced diabetes pretreated with minicircle ACE2, ACE2 protein in plasma increased markedly and this was associated with a more than 100-fold increase in serum ACE2 activity. However, minicircle ACE2 did not result in changes in urinary ACE2 activity as compared to untreated diabetic mice. In both diabetic groups, glomerular filtration rate increased significantly and to the same extent as compared to non-diabetic controls. Albuminuria, glomerular mesangial expansion, glomerular cellularity, and glomerular size were all increased to a similar extent in minicircle ACE2-treated and untreated diabetic mice, as compared to non-diabetic controls. Recombinant mouse ACE2 given for 4 weeks by intraperitoneal daily injections in mice with streptozotocin-induced diabetic nephropathy also failed to improve albuminuria or kidney pathology. Thus, a profound augmentation of ACE2 confined to the circulation failed to ameliorate the glomerular lesions and hyperfiltration characteristic of early diabetic nephropathy. These findings emphasize the importance of targeting the kidney rather than the circulatory renin angiotensin system to combat diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/etiologia , Rim/enzimologia , Peptidil Dipeptidase A/sangue , Albuminúria/enzimologia , Albuminúria/etiologia , Albuminúria/genética , Angiotensina I/metabolismo , Angiotensina II/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , Autoantígenos/genética , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/genética , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/enzimologia , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/enzimologia , Nefropatias Diabéticas/fisiopatologia , Feminino , Taxa de Filtração Glomerular , Rim/patologia , Rim/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/metabolismo , Peptidil Dipeptidase A/administração & dosagem , Peptidil Dipeptidase A/genética , Proteínas Recombinantes/administração & dosagem , Fatores de Tempo
13.
PLoS One ; 10(11): e0141231, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26555339

RESUMO

Alport syndrome is a genetic disease of collagen IV (α3, 4, 5) resulting in renal failure. This study was designed to investigate sex-phenotype correlations and evaluate the contribution of macrophage infiltration to disease progression using Col4a3 knock out (Col4a3KO) mice, an established genetic model of autosomal recessive Alport syndrome. No sex differences in the evolution of body mass loss, renal pathology, biomarkers of tubular damage KIM-1 and NGAL, or deterioration of kidney function were observed during the life span of Col4a3KO mice. These findings confirm that, similar to human autosomal recessive Alport syndrome, female and male Col4a3KO mice develop renal failure at the same age and with similar severity. The specific contribution of macrophage infiltration to Alport disease, one of the prominent features of the disease in human and Col4a3KO mice, remains unknown. This study shows that depletion of kidney macrophages in Col4a3KO male mice by administration of clodronate liposomes, prior to clinical onset of disease and throughout the study period, does not protect the mice from renal failure and interstitial fibrosis, nor delay disease progression. These results suggest that therapy targeting macrophage recruitment to kidney is unlikely to be effective as treatment of Alport syndrome.


Assuntos
Ácido Clodrônico/uso terapêutico , Colágeno Tipo IV/deficiência , Macrófagos/efeitos dos fármacos , Nefrite Hereditária/fisiopatologia , Animais , Apoptose , Autoantígenos/genética , Ácido Clodrônico/administração & dosagem , Ácido Clodrônico/farmacologia , Colágeno Tipo IV/genética , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Feminino , Rim/patologia , Falência Renal Crônica/etiologia , Lipossomos , Macrófagos/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Modelos Animais , Nefrite Hereditária/tratamento farmacológico , Nefrite Hereditária/genética , Nefrite Hereditária/imunologia , Caracteres Sexuais
14.
J Proteome Res ; 14(12): 5202-14, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26487288

RESUMO

The efficiency of the inhibition of the angiotensin converting enzyme, the most widely used therapy for the Alport syndrome, depends on the onset of the therapy-the earlier the better. Hence, early progressive biomarkers are urgently required to allow for preclinical diagnosis, an early start of possible therapy as well as the monitoring of this therapy. In the present study, an improved comprehensive and precise proteomic approach has been applied to the serum of juvenile Alport-mice, nontreated and treated, and wild-type controls of various ages to search for biomarkers. With a total of 2542 stringently altered proteins, the serum composition clearly shows a dependency on age, that is, stage, and therapy. Initially, the serum constituents indicate an enhanced extracellular matrix remodeling, cell damage, and the production of particular acute phase proteins. A panel of 15 potential biomarker candidates has been identified. In later stages, renal filtration failure and systemic acute phase reaction determine the composition of the serum; an effect that is well-known for manifested human Alport syndrome. With a small number of mouse urine samples, for example, the proteomic results for gelsolin could be verified using ELISA. Once verified in man, these early biomarkers would allow for a sensitive and specific diagnosis of the Alport syndrome in children as well as facilitate the monitoring of a possible therapy.


Assuntos
Autoantígenos/genética , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/genética , Nefrite Hereditária/sangue , Nefrite Hereditária/genética , Animais , Autoantígenos/sangue , Biomarcadores/sangue , Proteínas Sanguíneas/isolamento & purificação , Criança , Colágeno Tipo IV/sangue , Modelos Animais de Doenças , Progressão da Doença , Diagnóstico Precoce , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Humanos , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Nefrite Hereditária/diagnóstico , Proteômica , Espectrometria de Massas em Tandem
15.
Cell Biochem Funct ; 33(7): 503-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26449633

RESUMO

The aim of this study was to elucidate functional and molecular effects of mycophenolic acid (MPA) on non-lymphatic, kidney epithelial cells treated with transforming growth factor (TGF). MPA effects were studied using HK2 cells incubated with EGF and TGF. The reversibility of these effects was verified using guanosine and 8-aminoguanosine. The following assays were applied: cell proliferation, viability, collagen matrix contraction, scratch wound closure, spindle index, FACS with anti-CD29 and anti-CD326, promoter demethylation of RAS protein activator like 1 (RASAL1), as well as gene expression of RASAL1, integrin 1ß (ITGB1) (CD29) and epithelial cell adhesion molecule (EpCam) (CD326). Cell proliferation was inhibited by increasing concentrations of MPA, whereas neither apoptosis nor cytotoxicity was detected. Stimulation with EGF and/or TGF led to a significant collagen matrix contraction that was successfully inhibited by MPA. In addition, scratch wound closure was inhibited by incubation with TGF alone or with EGF. Under the same conditions, cell morphology (spindle shape) and molecular phenotype (ITGB1(High)EpCam(Low)/ITGB1(Low)EpCam(High)) were both significantly changed, suggesting an epithelial to mesenchymal transformation. Cell morphology and motility, as well as molecular phenotype, were reversible after MPA treatment with TGF transformation in both presence/absence of EGF, thereby suggesting a correlation with the previously described antifibrotic effects of MPA. Dysregulation of TGF signal transduction appears to be related to progression of fibrosis. A TGF-transformed kidney epithelial cell line derived from human proximal tubules was used to study whether the immunosuppressive drug: MPA possesses any functional or molecular antifibrotic effects. Functional and morphological in vitro changes induced by both the TGF and epithelial-growth-factor were reversible by treatment with MPA. An inhibitory effect of MPA on the TGF pathway appears to be responsible for the previously described antifibrotic effects of the MPA in the COL4A3-deficient mouse model of renal fibrosis.


Assuntos
Movimento Celular/efeitos dos fármacos , Colágeno Tipo IV/deficiência , Rim/efeitos dos fármacos , Rim/patologia , Ácido Micofenólico/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Autoantígenos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Fator de Crescimento Epidérmico/metabolismo , Fibrose/tratamento farmacológico , Humanos , Camundongos , Fator de Crescimento Transformador beta/metabolismo
16.
Circulation ; 131(18): 1555-65, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25753534

RESUMO

BACKGROUND: Collagen type IV alpha1 (COL4A1) and alpha2 (COL4A2) form heterotrimers critical for vascular basement membrane stability and function. Patients with COL4A1 or COL4A2 mutations suffer from diverse cerebrovascular diseases, including cerebral microbleeds, porencephaly, and fatal intracerebral hemorrhage (ICH). However, the pathogenic mechanisms remain unknown, and there is a lack of effective treatment. METHODS AND RESULTS: Using Col4a1 and Col4a2 mutant mouse models, we investigated the genetic complexity and cellular mechanisms underlying the disease. We found that Col4a1 mutations cause abnormal vascular development, which triggers small-vessel disease, recurrent hemorrhagic strokes, and age-related macroangiopathy. We showed that allelic heterogeneity, genetic context, and environmental factors such as intense exercise or anticoagulant medication modulated disease severity and contributed to phenotypic heterogeneity. We found that intracellular accumulation of mutant collagen in vascular endothelial cells and pericytes was a key triggering factor of ICH. Finally, we showed that treatment of mutant mice with a US Food and Drug Administration-approved chemical chaperone resulted in a decreased collagen intracellular accumulation and a significant reduction in ICH severity. CONCLUSIONS: Our data are the first to show therapeutic prevention in vivo of ICH resulting from Col4a1 mutation and imply that a mechanism-based therapy promoting protein folding might also prevent ICH in patients with COL4A1 and COL4A2 mutations.


Assuntos
Hemorragia Cerebral/prevenção & controle , Colágeno Tipo IV/genética , Heterogeneidade Genética , Fragmentos de Peptídeos/genética , Animais , Vasos Sanguíneos/anormalidades , Vasos Sanguíneos/embriologia , Barreira Hematoencefálica , Encéfalo/irrigação sanguínea , Encéfalo/embriologia , Hemorragia Cerebral/genética , Colágeno/metabolismo , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/fisiologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Feminino , Interação Gene-Ambiente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Neovascularização Fisiológica/genética , Fragmentos de Peptídeos/deficiência , Fragmentos de Peptídeos/fisiologia , Pericitos/metabolismo , Fenótipo , Condicionamento Físico Animal , Porencefalia/genética , Vasos Retinianos/embriologia
17.
J Clin Invest ; 125(1): 141-56, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25415439

RESUMO

MicroRNA-21 (miR-21) contributes to the pathogenesis of fibrogenic diseases in multiple organs, including the kidneys, potentially by silencing metabolic pathways that are critical for cellular ATP generation, ROS production, and inflammatory signaling. Here, we developed highly specific oligonucleotides that distribute to the kidney and inhibit miR-21 function when administered subcutaneously and evaluated the therapeutic potential of these anti-miR-21 oligonucleotides in chronic kidney disease. In a murine model of Alport nephropathy, miR-21 silencing did not produce any adverse effects and resulted in substantially milder kidney disease, with minimal albuminuria and dysfunction, compared with vehicle-treated mice. miR-21 silencing dramatically improved survival of Alport mice and reduced histological end points, including glomerulosclerosis, interstitial fibrosis, tubular injury, and inflammation. Anti-miR-21 enhanced PPARα/retinoid X receptor (PPARα/RXR) activity and downstream signaling pathways in glomerular, tubular, and interstitial cells. Moreover, miR-21 silencing enhanced mitochondrial function, which reduced mitochondrial ROS production and thus preserved tubular functions. Inhibition of miR-21 was protective against TGF-ß-induced fibrogenesis and inflammation in glomerular and interstitial cells, likely as the result of enhanced PPARα/RXR activity and improved mitochondrial function. Together, these results demonstrate that inhibition of miR-21 represents a potential therapeutic strategy for chronic kidney diseases including Alport nephropathy.


Assuntos
MicroRNAs/genética , Nefrite Hereditária/terapia , Oligorribonucleotídeos Antissenso/genética , Animais , Autoantígenos/genética , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/genética , Progressão da Doença , Fibrose/metabolismo , Rim/metabolismo , Rim/patologia , Redes e Vias Metabólicas/genética , Camundongos da Linhagem 129 , MicroRNAs/metabolismo , Nefrite Hereditária/metabolismo , Nefrite Hereditária/patologia , Espécies Reativas de Oxigênio/metabolismo , Transcriptoma , Regulação para Cima
19.
J Am Soc Nephrol ; 25(2): 260-75, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24262798

RESUMO

Thin-basement-membrane nephropathy (TBMN) and Alport syndrome (AS) are progressive collagen IV nephropathies caused by mutations in COL4A3/A4/A5 genes. These nephropathies invariably present with microscopic hematuria and frequently progress to proteinuria and CKD or ESRD during long-term follow-up. Nonetheless, the exact molecular mechanisms by which these mutations exert their deleterious effects on the glomerulus remain elusive. We hypothesized that defective trafficking of the COL4A3 chain causes a strong intracellular effect on the cell responsible for COL4A3 expression, the podocyte. To this end, we overexpressed normal and mutant COL4A3 chains (G1334E mutation) in human undifferentiated podocytes and tested their effects in various intracellular pathways using a microarray approach. COL4A3 overexpression in the podocyte caused chain retention in the endoplasmic reticulum (ER) that was associated with activation of unfolded protein response (UPR)-related markers of ER stress. Notably, the overexpression of normal or mutant COL4A3 chains differentially activated the UPR pathway. Similar results were observed in a novel knockin mouse carrying the Col4a3-G1332E mutation, which produced a phenotype consistent with AS, and in biopsy specimens from patients with TBMN carrying a heterozygous COL4A3-G1334E mutation. These results suggest that ER stress arising from defective localization of collagen IV chains in human podocytes contributes to the pathogenesis of TBMN and AS through activation of the UPR, a finding that may pave the way for novel therapeutic interventions for a variety of collagenopathies.


Assuntos
Colágeno Tipo IV/deficiência , Estresse do Retículo Endoplasmático/fisiologia , Membrana Basal Glomerular/metabolismo , Nefrite Hereditária/metabolismo , Podócitos/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Animais , Autoantígenos/genética , Autoantígenos/fisiologia , Biópsia , Células Cultivadas , Colágeno Tipo IV/genética , Colágeno Tipo IV/fisiologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Chaperona BiP do Retículo Endoplasmático , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Membrana Basal Glomerular/patologia , Proteínas de Choque Térmico/biossíntese , Proteínas de Choque Térmico/genética , Heterozigoto , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Mutação de Sentido Incorreto , Nefrite Hereditária/genética , Nefrite Hereditária/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Podócitos/patologia , Mutação Puntual , Análise Serial de Proteínas , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição/metabolismo , Transfecção
20.
Hum Mol Genet ; 23(2): 283-92, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24001601

RESUMO

Haemorrhagic stroke accounts for ∼20% of stroke cases and porencephaly is a clinical consequence of perinatal cerebral haemorrhaging. Here, we report the identification of a novel dominant G702D mutation in the collagen domain of COL4A2 (collagen IV alpha chain 2) in a family displaying porencephaly with reduced penetrance. COL4A2 is the obligatory protein partner of COL4A1 but in contrast to most COL4A1 mutations, the COL4A2 mutation does not lead to eye or kidney disease. Analysis of dermal biopsies from a patient and his unaffected father, who also carries the mutation, revealed that both display basement membrane (BM) defects. Intriguingly, defective collagen IV incorporation into the dermal BM was observed in the patient only and was associated with endoplasmic reticulum (ER) retention of COL4A2 in primary dermal fibroblasts. This intracellular accumulation led to ER stress, unfolded protein response activation, reduced cell proliferation and increased apoptosis. Interestingly, the absence of ER retention of COL4A2 and ER stress in cells from the unaffected father indicate that accumulation and/or clearance of mutant COL4A2 from the ER may be a critical modifier for disease development. Our analysis also revealed that mutant collagen IV is degraded via the proteasome. Importantly, treatment of patient cells with a chemical chaperone decreased intracellular COL4A2 levels, ER stress and apoptosis, demonstrating that reducing intracellular collagen accumulation can ameliorate the cellular phenotype of COL4A2 mutations. Importantly, these data highlight that manipulation of chaperone levels, intracellular collagen accumulation and ER stress are potential therapeutic options for collagen IV diseases including haemorrhagic stroke.


Assuntos
Membrana Basal/fisiopatologia , Encefalopatias/tratamento farmacológico , Colágeno Tipo IV/deficiência , Hemiplegia/tratamento farmacológico , Hemorragias Intracranianas/tratamento farmacológico , Fenilbutiratos/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Apoptose/efeitos dos fármacos , Membrana Basal/efeitos dos fármacos , Encefalopatias/genética , Encefalopatias/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Hemiplegia/genética , Hemiplegia/patologia , Humanos , Hemorragias Intracranianas/complicações , Hemorragias Intracranianas/genética , Masculino , Mutação , Fenótipo , Porencefalia , Complexo de Endopeptidases do Proteassoma/metabolismo , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA