Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Gastroenterology ; 159(3): 944-955.e8, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32442562

RESUMO

BACKGROUND & AIMS: Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects gastrointestinal tissues, little is known about the roles of gut commensal microbes in susceptibility to and severity of infection. We investigated changes in fecal microbiomes of patients with SARS-CoV-2 infection during hospitalization and associations with severity and fecal shedding of virus. METHODS: We performed shotgun metagenomic sequencing analyses of fecal samples from 15 patients with Coronavirus Disease 2019 (COVID-19) in Hong Kong, from February 5 through March 17, 2020. Fecal samples were collected 2 or 3 times per week from time of hospitalization until discharge; disease was categorized as mild (no radiographic evidence of pneumonia), moderate (pneumonia was present), severe (respiratory rate ≥30/min, or oxygen saturation ≤93% when breathing ambient air), or critical (respiratory failure requiring mechanical ventilation, shock, or organ failure requiring intensive care). We compared microbiome data with those from 6 subjects with community-acquired pneumonia and 15 healthy individuals (controls). We assessed gut microbiome profiles in association with disease severity and changes in fecal shedding of SARS-CoV-2. RESULTS: Patients with COVID-19 had significant alterations in fecal microbiomes compared with controls, characterized by enrichment of opportunistic pathogens and depletion of beneficial commensals, at time of hospitalization and at all timepoints during hospitalization. Depleted symbionts and gut dysbiosis persisted even after clearance of SARS-CoV-2 (determined from throat swabs) and resolution of respiratory symptoms. The baseline abundance of Coprobacillus, Clostridium ramosum, and Clostridium hathewayi correlated with COVID-19 severity; there was an inverse correlation between abundance of Faecalibacterium prausnitzii (an anti-inflammatory bacterium) and disease severity. Over the course of hospitalization, Bacteroides dorei, Bacteroides thetaiotaomicron, Bacteroides massiliensis, and Bacteroides ovatus, which downregulate expression of angiotensin-converting enzyme 2 (ACE2) in murine gut, correlated inversely with SARS-CoV-2 load in fecal samples from patients. CONCLUSIONS: In a pilot study of 15 patients with COVID-19, we found persistent alterations in the fecal microbiome during the time of hospitalization, compared with controls. Fecal microbiota alterations were associated with fecal levels of SARS-CoV-2 and COVID-19 severity. Strategies to alter the intestinal microbiota might reduce disease severity.


Assuntos
Betacoronavirus , Infecções por Coronavirus/microbiologia , Disbiose/virologia , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Pneumonia Viral/microbiologia , Adulto , Idoso , COVID-19 , Feminino , Trato Gastrointestinal/microbiologia , Hong Kong/epidemiologia , Hospitalização/estatística & dados numéricos , Humanos , Masculino , Pessoa de Meia-Idade , Pandemias , Projetos Piloto , SARS-CoV-2
2.
Eye Contact Lens ; 44 Suppl 2: S370-S375, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29944499

RESUMO

OBJECTIVES: To compare the various Pentacam-measured K-readings with the clinical history method (CHM) in eyes that have undergone myopic laser in situ keratomileusis (LASIK). METHODS: In this prospective study, Pentacam examination was performed in 71 eyes 1 month after myopic LASIK. The true net power (TNP) 4 mm, total corneal refractive power (TCRP) 4 mm, equivalent K-reading (EKR) 4.0 mm, and EKR 4.5 mm obtained from the same scan were compared with the K derived from CHM. RESULTS: The average baseline spherical equivalence was -5.44±2.38 D. After LASIK, the mean KCHM was 37.67±2.13 D, TCRP4mm was 37.14±1.79 D, TNP4mm was 36.88±1.76 D, EKR4.0mm was 37.58±1.94 D, and EKR4.5mm was 37.51±1.94 D. TCRP4mm, TNP4mm, and EKR4.5mm showed a statistically significant deviation from the KCHM, with the mean error being 0.53 D, 0.79 D, and 0.16 D, respectively (P<0.05). Only the EKR4.0mm showed no statistically significant difference from the KCHM (mean error 0.09 D, P=0.23). The EKR4.0mm also had the narrowest 95% limits of agreement (LoA) (-1.10 to +1.28 D), whereas both TCRP4mm and TNP4mm had a wider LoA (-0.88 to +1.95 D and -0.62 to +2.20 D, respectively). All four Pentacam K-readings had a strong and statistically significant correlation with the KCHM. CONCLUSIONS: Using the CHM as reference, the EKR4.0mm demonstrated the closest agreement when compared with the EKR4.5mm, TNP4mm, and TCRP4mm obtained from the same scan.


Assuntos
Córnea/fisiopatologia , Ceratomileuse Assistida por Excimer Laser In Situ/métodos , Miopia/cirurgia , Refração Ocular/fisiologia , Adulto , Córnea/cirurgia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Miopia/fisiopatologia , Estudos Prospectivos
3.
Curr Diab Rep ; 17(12): 132, 2017 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-29098478

RESUMO

PURPOSE OF REVIEW: The rising prevalence of obesity and diabetes cannot be fully explained by known risk factors, such as unhealthy diet, a sedentary lifestyle, and family history. This review summarizes the available studies linking persistent organic pollutants (POPs) to obesity and diabetes and discusses plausible underlying mechanisms. RECENT FINDINGS: Increasing evidence suggest that POPs may act as obesogens and diabetogens to promote the development of obesity and diabetes and induce metabolic dysfunction. POPs are synthesized chemicals and are used widely in our daily life. These chemicals are resistant to degradation in chemical or biological processes, which enable them to exist in the environment persistently and to be bio-accumulated in animal and human tissue through the food chain. Increasingly, epidemiologic studies suggest a positive association between POPs and risk of developing diabetes. Understanding the relationship of POPs with obesity and diabetes may shed light on preventive strategies for obesity and diabetes.


Assuntos
Diabetes Mellitus/induzido quimicamente , Diabetes Mellitus/epidemiologia , Poluentes Ambientais/efeitos adversos , Obesidade/induzido quimicamente , Obesidade/epidemiologia , Compostos Orgânicos/efeitos adversos , Animais , Humanos , Fatores de Risco
4.
Clin Exp Ophthalmol ; 44(1): 8-14, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26140309

RESUMO

BACKGROUND: This study aims to compare the effect of conventional corneal collagen cross-linking (CXL) with accelerated corneal collagen cross-linking in treatment of keratoconus. DESIGN: A comparative interventional study was employed. PARTICIPANTS: Participants were consecutive cases of progressive keratoconus receiving either conventional (3 mW/cm(2) irradiance for 30 min) or accelerated CXL (9 mW/cm(2) irradiance for 10 min). METHODS: Clinical and topographic parameters were compared between the two groups. Postoperative corneal stromal demarcation line was measured using anterior segment optical coherence tomography. MAIN OUTCOME MEASURES: Clinical and topographic parameters such as corrected distant visual acuity (CDVA), maximum keratometry (Kmax), mean keratometry (Kmean), demarcation line depth were gathered from medical records. RESULTS: There were a total of 26 eyes with an average follow up of 13.9 ± 6.3 months. Fourteen eyes received conventional CXL, and 12 eyes had accelerated CXL. In the conventional CXL group, CDVA improved significantly (P = 0.021). There was also a significant reduction in Kmax (P = 0.003) and Kmean (P = 0.002). In the accelerated CXL group, no significant changes were found in CDVA (P = 0.395), Kmax (P = 0.388) and Kmean (P = 0.952) postoperatively. A significantly greater reduction in Kmax and Kmean were seen in conventional CXL compared to its accelerated counterpart (P = 0.001 and 0.015, respectively). The demarcation line was deeper in eyes with conventional CXL (P = 0.013), and the depth correlated significantly with the change in Kmean (r = -0.432, P = 0.045). CONCLUSION: Conventional and accelerated CXL are effective in stabilizing keratoconus progression after a mean of 12 months. Patients undergoing conventional CXL showed clinical improvement with greater corneal flattening, which correlated with a deeper corneal stromal demarcation line. This current study is the first to report such correlation.


Assuntos
Colágeno/metabolismo , Substância Própria/metabolismo , Reagentes de Ligações Cruzadas , Ceratocone/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Riboflavina/uso terapêutico , Adulto , Feminino , Seguimentos , Humanos , Ceratocone/metabolismo , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Raios Ultravioleta , Adulto Jovem
5.
Am J Pathol ; 184(2): 409-17, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24262754

RESUMO

We have previously shown that transforming growth factor-ß/Smad3-dependent miRNAs play a critical role in renal inflammation and fibrosis. However, off-target effects of miRNAs limit their therapeutic application. Recently, emerging roles of long noncoding RNAs (lncRNAs) in diseases have been recognized. In this study, we used high-throughput RNA sequencing to identify the Smad3-dependent lncRNAs related to renal inflammation and fibrosis in Smad3 knockout mouse models of unilateral ureteral obstructive nephropathy and immunologically induced anti-glomerular basement membrane glomerulonephritis. Compared with wild-type mice, 151 lncRNAs in the unilateral ureteral obstructive nephropathy kidney and 413 lncRNAs in kidneys with anti-glomerular basement membrane glomerulonephritis were significantly altered in Smad3 knockout mice. Among them, 21 common lncRNAs were up-regulated in wild-type, but down-regulated in Smad3 knockout, kidneys in both disease models in which progressive renal inflammation and fibrosis were abolished when the Smad3 gene was deleted or suppressed. Real-time PCR confirmed these findings and revealed the functional link between Smad3-dependent lncRNAs np_5318/np_17856 and progressive kidney injury. Results demonstrate that the identification and characterization of functional lncRNAs associated with kidney disease may represent a promising research direction into renal disorder and may lead to the development of new lncRNA therapies for kidney diseases.


Assuntos
Inflamação/patologia , Rim/metabolismo , Rim/patologia , RNA Longo não Codificante/metabolismo , Análise de Sequência de RNA , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Feminino , Fibrose , Regulação da Expressão Gênica , Ontologia Genética , Glomerulonefrite/genética , Glomerulonefrite/patologia , Inflamação/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Ligação Proteica , RNA Longo não Codificante/genética , Transdução de Sinais , Obstrução Ureteral/genética , Obstrução Ureteral/patologia
6.
Adv Exp Med Biol ; 888: 253-69, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26663187

RESUMO

Diabetes and diabetic kidney diseases have continually exerted a great burden on our society. Although the recent advances in medical research have led to a much better understanding of diabetic kidney diseases, there is still no successful strategy for effective treatments for diabetic kidney diseases. Recently, treatment of diabetic kidney diseases relies either on drugs that reduce the progression of renal injury or on renal replacement therapies, such as dialysis and kidney transplantation. On the other hand, searching for biomarkers for early diagnosis and effective therapy is also urgent. Discovery of microRNAs has opened to a novel field for posttranscriptional regulation of gene expression. Results from cell culture experiments, experimental animal models, and patients under diabetic conditions reveal the critical role of microRNAs during the progression of diabetic kidney diseases. Functional studies demonstrate not only the capability of microRNAs to regulate expression of target genes, but also their therapeutic potential to diabetic kidney diseases. The existence of microRNAs in plasma, serum, and urine suggests their possibility to be biomarkers in diabetic kidney diseases. Thus, identification of the functional role of microRNAs provides an essentially clinical impact in terms of prevention and treatment of progression in diabetic kidney diseases as it enables us to develop novel, specific therapies and diagnostic tools for diabetic kidney diseases.


Assuntos
Biomarcadores/metabolismo , Nefropatias Diabéticas/genética , Regulação da Expressão Gênica , MicroRNAs/genética , Animais , Nefropatias Diabéticas/diagnóstico , Nefropatias Diabéticas/terapia , Modelos Animais de Doenças , Diagnóstico Precoce , Humanos , Rim/metabolismo , Rim/patologia
7.
Mol Ther ; 21(2): 388-98, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23207693

RESUMO

Blockade of transforming growth factor-ß (TGF-ß) signaling by Smad7 gene therapy is known to prevent experimental renal fibrosis. This study investigated whether Smad7 suppresses renal fibrosis via altering the renal expression of fibrosis-related microRNAs. Application of gene therapy into diseased kidneys of obstructive nephropathy and kidney cells by overexpressing Smad7 restored miR-29b but inhibited the expression of miR-192 and miR-21, resulting in blockade of renal fibrosis. Furthermore, Smad7 overexpression also suppressed advanced glycated end products- and angiotensin II-regulated expression of these microRNAs. In contrast, disruption of Smad7 gene in mice demonstrated opposite results by enhancing the loss of miR-29b and upregulation of miR-192 and miR-21, resulting in promotion of renal fibrosis in ligated kidneys of a model of obstructive nephropathy. More importantly, treatment with anti-miR-29b, miR-21 and miR-192 mimics in Smad7 overexpressing tubular epithelial cells abrogated the suppressive function of Smad7 on renal fibrosis, suggesting that these microRNAs act downstream of Smad7 to override the Smad7 function. In conclusion, Smad7 protects kidneys from fibrosis by regulating TGF-ß/Smad3-mediated renal expression of miR-21, miR-192, and miR-29b. Restored renal miR-29b but suppressed miR-192 and miR-21 may be a mechanism by which gene therapy with Smad7 inhibits renal fibrosis.


Assuntos
Nefropatias/prevenção & controle , MicroRNAs/genética , Proteína Smad7/genética , Fator de Crescimento Transformador beta1/genética , Animais , Western Blotting , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fibrose , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Imuno-Histoquímica , Hibridização In Situ , Nefropatias/genética , Nefropatias/patologia , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
8.
Kidney Int ; 84(6): 1129-44, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23868013

RESUMO

The TGF-ß/Smad3 pathway plays a major role in tissue fibrosis, but the precise mechanisms are not fully understood. Here we identified microRNA miR-433 as an important component of TGF-ß/Smad3-driven renal fibrosis. The miR-433 was upregulated following unilateral ureteral obstruction, a model of aggressive renal fibrosis. In vitro, overexpression of miR-433 enhanced TGF-ß1-induced fibrosis, whereas knockdown of miR-433 suppressed this response. Furthermore, Smad3, but not Smad2, bound to the miR-433 promoter to induce its expression. Delivery of an miR-433 knockdown plasmid to the kidney by ultrasound microbubble-mediated gene transfer suppressed the induction and progression of fibrosis in the obstruction model. The antizyme inhibitor Azin1, an important regulator of polyamine synthesis, was identified as a target of miR-433. Overexpression of miR-433 suppressed Azin1 expression, while, in turn, Azin1 overexpression suppressed TGF-ß signaling and the fibrotic response. Thus, miR-433 is an important component of TGF-ß/Smad3-induced renal fibrosis through the induction of a positive feedback loop to amplify TGF-ß/Smad3 signaling, and may be a potential therapeutic target in tissue fibrosis.


Assuntos
Proteínas de Transporte/metabolismo , Nefropatias/metabolismo , Rim/metabolismo , MicroRNAs/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Doença Antimembrana Basal Glomerular/genética , Doença Antimembrana Basal Glomerular/metabolismo , Doença Antimembrana Basal Glomerular/patologia , Sítios de Ligação , Proteínas de Transporte/genética , Linhagem Celular , Modelos Animais de Doenças , Doxorrubicina , Fibrose , Rim/patologia , Nefropatias/etiologia , Nefropatias/genética , Nefropatias/patologia , Nefropatias/prevenção & controle , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Regiões Promotoras Genéticas , Interferência de RNA , Ratos , Transdução de Sinais , Proteína Smad2/deficiência , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/deficiência , Proteína Smad3/genética , Proteína Smad7/genética , Proteína Smad7/metabolismo , Fatores de Tempo , Transfecção , Fator de Crescimento Transformador beta1/genética , Regulação para Cima , Obstrução Ureteral/complicações
9.
Clin Sci (Lond) ; 124(4): 243-54, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23126427

RESUMO

TGF-ß (transforming growth factor-ß) and BMP-7 (bone morphogenetic protein-7), two key members in the TGF-ß superfamily, play important but diverse roles in CKDs (chronic kidney diseases). Both TGF-ß and BMP-7 share similar downstream Smad signalling pathways, but counter-regulate each other to maintain the balance of their biological activities. During renal injury in CKDs, this balance is significantly altered because TGF-ß signalling is up-regulated by inducing TGF-ß1 and activating Smad3, whereas BMP-7 and its downstream Smad1/5/8 are down-regulated. In the context of renal fibrosis, Smad3 is pathogenic, whereas Smad2 and Smad7 are renoprotective. However, this counter-balancing mechanism is also altered because TGF-ß1 induces Smurf2, a ubiquitin E3-ligase, to target Smad7 as well as Smad2 for degradation. Thus overexpression of renal Smad7 restores the balance of TGF-ß/Smad signalling and has therapeutic effect on CKDs. Recent studies also found that Smad3 mediated renal fibrosis by up-regulating miR-21 (where miR represents microRNA) and miR-192, but down-regulating miR-29 and miR-200 families. Therefore restoring miR-29/miR-200 or suppressing miR-21/miR-192 is able to treat progressive renal fibrosis. Furthermore, activation of TGF-ß/Smad signalling inhibits renal BMP-7 expression and BMP/Smad signalling. On the other hand, overexpression of renal BMP-7 is capable of inhibiting TGF-ß/Smad3 signalling and protects the kidney from TGF-ß-mediated renal injury. This counter-regulation not only expands our understanding of the causes of renal injury, but also suggests the therapeutic potential by targeting TGF-ß/Smad signalling or restoring BMP-7 in CKDs. Taken together, the current understanding of the distinct roles and mechanisms of TGF-ß and BMP-7 in CKDs implies that targeting the TGF-ß/Smad pathway or restoring BMP-7 signalling may represent novel and effective therapies for CKDs.


Assuntos
Proteína Morfogenética Óssea 7/metabolismo , Insuficiência Renal Crônica/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Apoptose/fisiologia , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 7/antagonistas & inibidores , Proliferação de Células , Fibrose/metabolismo , Humanos , Fármacos Renais/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/fisiopatologia , Proteínas Smad/antagonistas & inibidores , Fator de Crescimento Transformador beta/antagonistas & inibidores
10.
J Refract Surg ; 29(1): 49-53, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23311741

RESUMO

PURPOSE: To compare the corneal collagen cross-linking (CXL) demarcation line depth between the central and peripheral cornea after cross-linking using anterior segment optical coherence tomography. METHODS: Retrospective interventional case series of 38 eyes with keratoconus or postoperative LASIK ectasia treated with riboflavin ultraviolet A CXL (UV-X, IROC). CXL demarcation line depth, corneal thickness, and the ratio of the CXL demarcation line depth to the corneal thickness were measured using anterior segment optical coherence tomography at the central cornea and at 2 and 4 mm from the corneal center in four regions: temporal, nasal, superior, and inferior. The CXL demarcation line depths at the center and periphery were compared using the Friedman test. RESULTS: The CXL demarcation line was deepest in the central cornea (302 µm; range: 180 to 397 µm) and was reduced progressively toward the peripheral cornea, at nasal 2 mm (289.5 µm; range: 125 to 370 µm), at nasal 4 mm (206.5 µm; range: 100 to 307 µm), at temporal 2 mm (278.5 µm; range: 128 to 375 µm), and at temporal 4 mm (194 µm; range: 80 to 325 µm) (P<.001). The penetration proportion was greatest at the central cornea (64.8%; range: 44% to 80%) and was decreased toward the periphery, at nasal 2 mm (53.8%; range: 30% to 74%), at nasal 4 mm (33.8%; range: 19% to 53%), at temporal 2 mm (54.1%; range: 29% to 77%), and at temporal 4 mm (34.1%; range: 15% to 54%) (P⩽.001). CONCLUSIONS: Both the CXL demarcation line depth and its proportion over the corneal thickness were greater at the central cornea than the peripheral cornea.


Assuntos
Colágeno/metabolismo , Reagentes de Ligações Cruzadas/uso terapêutico , Ceratocone/tratamento farmacológico , Adolescente , Adulto , Criança , Substância Própria/metabolismo , Substância Própria/patologia , Dilatação Patológica/tratamento farmacológico , Feminino , Humanos , Ceratocone/diagnóstico , Ceratocone/metabolismo , Masculino , Pessoa de Meia-Idade , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Estudos Retrospectivos , Riboflavina/uso terapêutico , Tomografia de Coerência Óptica , Raios Ultravioleta , Adulto Jovem
11.
J Pathol ; 227(2): 175-88, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22190171

RESUMO

TGF-ß1 binds receptor II (TßRII) to exert its biological activities but its functional importance in kidney diseases remains largely unclear. In the present study, we hypothesized that TßRII may function to initiate the downstream TGF-ß signalling and determine the diverse role of TGF-ß1 in kidney injury. The hypothesis was examined in a model of unilateral ureteral obstructive (UUO) nephropathy and in kidney fibroblasts and tubular epithelial cells in which the TßRII was deleted conditionally. We found that disruption of TßRII inhibited severe tubulointerstitial fibrosis in the UUO kidney, which was associated with the impairment of TGF-ß/Smad3 signalling, but not with the ERK/p38 MAP kinase pathway. In contrast, deletion of TßRII enhanced NF-κB signalling and renal inflammation including up-regulation of Il-1ß and Tnfα in the UUO kidney. Similarly, in vitro disruption of TßRII from kidney fibroblasts or tubular epithelial cells inhibited TGF-ß1-induced Smad signalling and fibrosis but impaired the anti-inflammatory effect of TGF-ß1 on IL-1ß-stimulated NF-κB activation and pro-inflammatory cytokine expression. In conclusion, TßRII plays an important but diverse role in regulating renal fibrosis and inflammation. Impaired TGF-ß/Smad3, but not the non-canonical TGF-ß signalling pathway, may be a key mechanism by which disruption of TßRII protects against renal fibrosis. In addition, deletion of TßRII also enhances NF-κB signalling along with up-regulation of renal pro-inflammatory cytokines, which may be associated with the impairment of anti-inflammatory properties of TGF-ß1.


Assuntos
Rim/metabolismo , Nefrite/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Rim/imunologia , Rim/patologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Nefrite/etiologia , Nefrite/genética , Nefrite/patologia , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais , Proteína Smad3/metabolismo , Fatores de Tempo , Transfecção , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/complicações , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Kidney Int ; 81(3): 266-79, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22048127

RESUMO

The mechanism by which TGF-ß regulates renal inflammation and fibrosis is largely unclear; however, it is well accepted that its biological effects are mediated through Smad2 and Smad3 phosphorylation. Following activation, these Smads form heteromeric complex with Smad4 and translocate into the nucleus to bind and regulate the expression of target genes. Here we studied the roles of Smad4 to regulate TGF-ß signaling in a mouse model of unilateral ureteral obstruction using conditional Smad4 knockout mice and in isolated Smad4 mutant macrophages and fibroblasts. Disruption of Smad4 significantly enhanced renal inflammation as evidenced by a greater CD45(+) leukocyte and F4/80(+) macrophage infiltration and upregulation of IL-1ß, TNF-α, MCP-1, and ICAM-1 in the obstructed kidney and in IL-1ß-stimulated macrophages. In contrast, deletion of Smad4 inhibited renal fibrosis and TGF-ß1-induced collagen I expression by fibroblasts. Further studies showed that the loss of Smad4 repressed Smad7 transcription, leading to a loss of functional protein. This, in turn, inhibited IκBα expression but enhanced NF-κB activation, thereby promoting renal inflammation. Interestingly, deletion of Smad4 influenced Smad3-mediated promoter activities and the binding of Smad3 to the COL1A2 promoter, but not Smad3 phosphorylation and nuclear translocation, thereby inhibiting the fibrotic response. Thus, Smad4 may be a key regulator for the diverse roles of TGF-ß1 in inflammation and fibrogenesis by interacting with Smad7 and Smad3 to influence their transcriptional activities in renal inflammation and fibrosis.


Assuntos
Rim/patologia , Nefrite/metabolismo , Proteína Smad3/fisiologia , Proteína Smad4/fisiologia , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Fibrose , Regulação da Expressão Gênica , Interleucina-1beta/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Transcrição Gênica
13.
Stem Cells ; 29(7): 1041-51, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21608077

RESUMO

The pluripotency gene Oct4 encodes a key transcription factor that maintains self-renewal of embryonic stem cell (ESC) and is downregulated upon differentiation of ESCs and silenced in somatic cells. A combination of cis elements, transcription factors, and epigenetic modifications, such as DNA methylation, mediates Oct4 gene expression. Here, we show that the orphan nuclear receptor germ cell nuclear factor (GCNF) initiates Oct4 repression and DNA methylation by the differential recruitment of methyl-CpG binding domain (MBD) and DNA methyltransferases (Dnmts) to the Oct4 promoter. When compared with wild-type ESCs and gastrulating embryos, Oct4 repression is lost and its proximal promoter is significantly hypomethylated in retinoic acid (RA)-differentiated GCNF(-/-) ESCs and GCNF(-/-) embryos. Efforts to characterize mediators of GCNF's repressive function and DNA methylation of the Oct4 promoter identified MBD3, MBD2, and de novo Dnmts as GCNF interacting factors. Upon differentiation, endogenous GCNF binds to the Oct4 proximal promoter and differentially recruits MBD3 and MBD2 as well as Dnmt3A. In differentiated GCNF(-/-) ESCs, recruitment of MBD3 and MBD2 as well as Dnmt3A to Oct4 promoter is lost and subsequently Oct4 repression and DNA methylation failed to occur. Hypomethylation of the Oct4 promoter is also observed in RA-differentiated MBD3(-/-) and Dnmt3A(-/-) ESCs, but not in MBD2(-/-) and Dnmt3B(-/-) ESCs. Thus, recruitment of MBD3, MBD2, and Dnmt3A by GCNF links two events: gene-specific repression and DNA methylation, which occur differentially at the Oct4 promoter. GCNF initiates the repression and epigenetic modification of Oct4 gene during ESC differentiation.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/fisiologia , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Dados de Sequência Molecular , Membro 1 do Grupo A da Subfamília 6 de Receptores Nucleares/genética , Fator 3 de Transcrição de Octâmero/biossíntese , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição/genética
14.
J Refract Surg ; 28(7): 475-81, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22767165

RESUMO

PURPOSE: To report the demarcation line depth after corneal collagen cross-linking (CXL) for keratoconus and postoperative LASIK ectasia with Visante (Carl Zeiss Meditec) optical coherence tomography (OCT) and investigate correlations between this depth and preoperative parameters and postoperative visual and keratometry outcomes. METHODS: Retrospective interventional case series of 40 eyes with keratoconus or postoperative LASIK ectasia treated with riboflavin ultraviolet-A CXL. Ophthalmic evaluation included uncorrected distance visual acuity, corrected distance visual acuity (CDVA), corneal topography (Pentacam, Oculus Optikgeräte GmbH), and postoperative anterior segment OCT. Correlation analyses were performed. Statistical significance was indicated by P<.05. RESULTS: At 6 months postoperative, mean demarcation line depth was 281.4±53.3 µm. Thinner minimal corneal thickness (r=0.413, P=.008), older age (r=-0.490, P=.001), higher grading of ectasia (for both keratoconus and postoperative LASIK ectasia) (r=-0.332, P=.044), female sex (r=0.343, P=.030), postoperative LASIK ectasia (r=-0.420, P=.007), and longer duration of disease (r=-0.377, P=.023) were correlated with shallower demarcation line depth. Forward stepwise multiple linear regression analysis showed that among all preoperative factors, minimal corneal thickness (standardized ß=0.473, P=.003) and age (standardized ß=-0.317, P=.036) were significantly associated with demarcation line depth. The CXL demarcation line depth and change in CDVA (r=-0.16, P=.325) and change of the steepest keratometry at 6 months (r=0.084, P=.637) were not correlated. CONCLUSIONS: The Visante OCT can be used for assessing CXL demarcation line depth. The demarcation line depth may decrease with the severity of ectasia and age.


Assuntos
Colágeno/metabolismo , Substância Própria/metabolismo , Reagentes de Ligações Cruzadas/uso terapêutico , Ceratocone/tratamento farmacológico , Tomografia de Coerência Óptica , Adolescente , Adulto , Criança , Topografia da Córnea , Dilatação Patológica/tratamento farmacológico , Dilatação Patológica/metabolismo , Feminino , Humanos , Ceratocone/metabolismo , Masculino , Pessoa de Meia-Idade , Fármacos Fotossensibilizantes/uso terapêutico , Complicações Pós-Operatórias , Estudos Retrospectivos , Riboflavina/uso terapêutico , Raios Ultravioleta , Acuidade Visual/fisiologia
15.
J Am Soc Nephrol ; 22(5): 802-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21474561

RESUMO

The main function of chemokines is to guide inflammatory cells in their migration to sites of inflammation. During the last 2 decades, an expanding number of chemokines and their receptors have driven broad inquiry into how inflammatory cells are recruited in a variety of diseases. Although this review focuses on chemokines and their receptors in renal injury, proinflammatory IL-17, TGFß, and TWEAK signaling pathways also play a critical role in their expression. Recent studies in transgenic mice as well as blockade of chemokine signaling by neutralizing ligands or receptor antagonists now allow direct interrogation of chemokine action. The emerging role of regulatory T cells and Th17 cells during renal injury also forges tight relationships between chemokines and T cell infiltration in the development of kidney disease. As chemokine receptor blockade inches toward clinical use, the field remains an attractive area with potential for unexpected opportunity in the future.


Assuntos
Quimiocinas/fisiologia , Nefropatias/etiologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/imunologia , Animais , Quimiocinas/genética , Doença Crônica , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Receptores de Quimiocinas/fisiologia , Linfócitos T/imunologia , Linfócitos T Reguladores/fisiologia , Células Th17/fisiologia
16.
J Am Soc Nephrol ; 22(9): 1668-81, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21852586

RESUMO

TGF-ß/Smad signaling plays a role in fibrogenesis, but therapies targeting TGF-ß are ineffective in treating renal fibrosis. Here, we explored the therapeutic potential of targeting TGF-ß-induced microRNA in the progression of renal fibrosis. Microarray analysis and real-time PCR revealed upregulation of miR-21 in tubular epithelial cells (TECs) in response to TGF-ß. Lack of Smad3, but not lack of Smad2, prevented cells from upregulating miR-21 in response to TGF-ß. In addition, Smad3-deficient mice were protected from upregulation of miR-21 and fibrosis in the unilateral ureteral obstruction model. In contrast, conditional knockout of Smad2 enhanced miR-21 expression and renal fibrosis. Furthermore, ultrasound-microbubble-mediated gene transfer of a miR-21-knockdown plasmid halted the progression of renal fibrosis in established obstructive nephropathy. In conclusion, these data demonstrate that Smad3, but not Smad2, signaling increases expression of miR-21, which promotes renal fibrosis. Inhibition of miR-21 may be a therapeutic approach to suppress renal fibrosis.


Assuntos
MicroRNAs/metabolismo , Nefroesclerose/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem Celular , Progressão da Doença , Técnicas de Silenciamento de Genes , Técnicas de Transferência de Genes , Camundongos , MicroRNAs/genética , Ratos , Proteína Smad2/metabolismo , Regulação para Cima
17.
J Am Soc Nephrol ; 22(8): 1462-74, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21784902

RESUMO

TGF-ß/Smad3 signaling promotes fibrosis, but the development of therapeutic interventions involving this pathway will require the identification and ultimate targeting of downstream fibrosis-specific genes. In this study, using a microRNA microarray and real-time PCR, wild-type mice had reduced expression of miR-29 along with the development of progressive renal fibrosis in obstructive nephropathy. In contrast, Smad3 knockout mice had increased expression of miR-29 along with the absence of renal fibrosis in the same model of obstruction. In cultured fibroblasts and tubular epithelial cells, Smad3 mediated TGF-ß(1)-induced downregulation of miR-29 by binding to the promoter of miR-29. Furthermore, miR-29 acted as a downstream inhibitor and therapeutic microRNA for TGF-ß/Smad3-mediated fibrosis. In vitro, overexpression of miR-29b inhibited, but knockdown of miR-29 enhanced, TGF-ß(1)-induced expression of collagens I and III by renal tubular cells. Ultrasound-mediated gene delivery of miR-29b either before or after established obstructive nephropathy blocked progressive renal fibrosis. In conclusion, miR-29 is a downstream inhibitor of TGF-ß/Smad3-mediated fibrosis and may have therapeutic potential for diseases involving fibrosis.


Assuntos
Fibrose/patologia , Nefropatias/patologia , MicroRNAs/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Fibroblastos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/antagonistas & inibidores , Regiões Promotoras Genéticas , Resultado do Tratamento
18.
Mol Metab ; 60: 101493, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35398277

RESUMO

OBJECTIVE: ß cell dedifferentiation may underlie the reversible reduction in pancreatic ß cell mass and function in type 2 diabetes (T2D). We previously reported that ß cell-specific Sirt3 knockout (Sirt3f/f;Cre/+) mice developed impaired glucose tolerance and glucose-stimulated insulin secretion after feeding with high fat diet (HFD). RNA sequencing showed that Sirt3-deficient islets had enhanced expression of Enpp2 (Autotaxin, or ATX), a secreted lysophospholipase which produces lysophosphatidic acid (LPA). Here, we hypothesized that activation of the ATX/LPA pathway contributed to pancreatic ß cell dedifferentiation in Sirt3-deficient ß cells. METHODS: We applied LPA, or lysophosphatidylcoline (LPC), the substrate of ATX for producing LPA, to MIN6 cell line and mouse islets with altered Sirt3 expression to investigate the effect of LPA on ß cell dedifferentiation and its underlying mechanisms. To examine the pathological effects of ATX/LPA pathway, we injected the ß cell selective adeno-associated virus (AAV-Atx-shRNA) or negative control AAV-scramble in Sirt3f/f and Sirt3f/f;Cre/+ mice followed by 6-week of HFD feeding. RESULTS: In Sirt3f/f;Cre/+ mouse islets and Sirt3 knockdown MIN6 cells, ATX upregulation led to increased LPC with increased production of LPA. The latter not only induced reversible dedifferentiation in MIN6 cells and mouse islets, but also reduced glucose-stimulated insulin secretion from islets. In MIN6 cells, LPA induced phosphorylation of JNK/p38 MAPK which was accompanied by ß cell dedifferentiation. The latter was suppressed by inhibitors of LPA receptor, JNK, and p38 MAPK. Importantly, inhibiting ATX in vivo improved insulin secretion and reduced ß cell dedifferentiation in HFD-fed Sirt3f/f;Cre/+ mice. CONCLUSIONS: Sirt3 prevents ß cell dedifferentiation by inhibiting ATX expression and upregulation of LPA. These findings support a long-range signaling effect of Sirt3 which modulates the ATX-LPA pathway to reverse ß cell dysfunction associated with glucolipotoxicity.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Sirtuína 3/metabolismo , Animais , Desdiferenciação Celular , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Sirtuína 3/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Front Med (Lausanne) ; 9: 882240, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35979210

RESUMO

Vernal keratoconjunctivitis (VKC) is an underdiagnosed and underrecognized ocular surface disease with limited epidemiological data in Asia. It is more prevalent in warm, dry, and windy climates, and often has a substantial impact on a patient's quality of life. In rare cases, VKC can be associated with vision loss, either through corticosteroid overuse or inadequate treatment of persistent inflammation. As a potentially severe and complex disease, there is variability with how VKC is managed across Asia and among the various allergic eye diseases. Diagnosis and treatment of patients with VKC is a challenge for many ophthalmologists, since no precise diagnostic criteria have been established, the pathogenesis of the disease is unclear, and anti-allergic treatments are often ineffective in patients with moderate or severe disease. In addition, the choice of treatment and management strategies used for patients varies greatly from country to country and physician to physician. This may be because of a lack of well-defined, standardized guidelines. In response, the Management of Vernal Keratoconjunctivitis in Asia (MOVIA) Expert Working Group (13 experts) completed a consensus program to evaluate, review, and develop best-practice recommendations for the assessment, diagnosis, and management of VKC in Asia. The expert-led recommendations are summarized in this article and based on the currently available evidence alongside the clinical expertise of ophthalmologists from across Asia with specialism and interest in the ocular surface, VKC, and pediatric ophthalmology.

20.
Lab Invest ; 91(6): 837-51, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21383672

RESUMO

Elevated blood level of C-reactive protein (CRP) is associated with increased risk of chronic kidney disease. However, whether this association reflects functional importance of CRP in the pathogenesis of kidney disease remains unclear. In this study, we examined the biological role of CRP in a well-characterized model of progressive kidney disease, unilateral ureteral obstruction (UUO), in mice that express the human CRP gene (CRPtg). Compared with wild-type (Wt) mice at 3 days after UUO, CRPtg mice developed more severe renal inflammation with a significant increase in tubulointerstitial T cells and macrophages, upregulation of proinflammatory cytokines (IL-1ß and TNF-α), chemokines (MCP-1), and adhesion molecules (ICAM-1). Renal fibrosis was also significantly enhanced in CRPtg mice as demonstrated by increased expression of tubulointerstitial α-smooth muscle actin and collagen types I and III compared with Wt mice. Interestingly, on days 7 and 14 after UUO, an equal severity of renal inflammation and fibrosis were observed in CRPtg and Wt mice. These findings suggested that CRP may have a role in the initiation of renal inflammation and fibrosis. Further study revealed that enhanced early renal inflammation and fibrosis on day 3 in CRPtg mice was associated with a significant upregulation of endogenous mouse CRP and FcγRI mRNA and increased activation of both NF-κB/p65 and TGF-ß/Smad2/3 signaling, while equal severity of progressive renal injury at day 7 and day 14 between CRPtg and Wt mice were attributed to equivalent levels of CRP, FcγRI, phospho-NF-κB/p65, and TGF-ß/Smad2/3 signaling. Based on these findings, we conclude that CRP may not only be a biomarker, but also a mediator in the early development of renal inflammation and fibrosis in a mouse model of UUO. Enhanced activation of both NF-κB and TGF-ß/Smad signaling pathways may be mechanisms by which CRP promotes early renal inflammation and fibrosis.


Assuntos
Proteína C-Reativa/metabolismo , Nefropatias/patologia , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia , Obstrução Ureteral/metabolismo , Actinas/metabolismo , Análise de Variância , Animais , Moléculas de Adesão Celular/metabolismo , Quimiocinas/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo II/metabolismo , Citocinas/metabolismo , Fibrose/patologia , Humanos , Imuno-Histoquímica , Nefropatias/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Nefrite/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA