Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Nat Commun ; 15(1): 433, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38199997

RESUMO

There is a need to define regions of gene activation or repression that control human kidney cells in states of health, injury, and repair to understand the molecular pathogenesis of kidney disease and design therapeutic strategies. Comprehensive integration of gene expression with epigenetic features that define regulatory elements remains a significant challenge. We measure dual single nucleus RNA expression and chromatin accessibility, DNA methylation, and H3K27ac, H3K4me1, H3K4me3, and H3K27me3 histone modifications to decipher the chromatin landscape and gene regulation of the kidney in reference and adaptive injury states. We establish a spatially-anchored epigenomic atlas to define the kidney's active, silent, and regulatory accessible chromatin regions across the genome. Using this atlas, we note distinct control of adaptive injury in different epithelial cell types. A proximal tubule cell transcription factor network of ELF3, KLF6, and KLF10 regulates the transition between health and injury, while in thick ascending limb cells this transition is regulated by NR2F1. Further, combined perturbation of ELF3, KLF6, and KLF10 distinguishes two adaptive proximal tubular cell subtypes, one of which manifested a repair trajectory after knockout. This atlas will serve as a foundation to facilitate targeted cell-specific therapeutics by reprogramming gene regulatory networks.


Assuntos
Cromatina , Rim , Humanos , Cromatina/genética , Túbulos Renais Proximais , Nível de Saúde , Contagem de Células
2.
bioRxiv ; 2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37333123

RESUMO

There is a need to define regions of gene activation or repression that control human kidney cells in states of health, injury, and repair to understand the molecular pathogenesis of kidney disease and design therapeutic strategies. However, comprehensive integration of gene expression with epigenetic features that define regulatory elements remains a significant challenge. We measured dual single nucleus RNA expression and chromatin accessibility, DNA methylation, and H3K27ac, H3K4me1, H3K4me3, and H3K27me3 histone modifications to decipher the chromatin landscape and gene regulation of the kidney in reference and adaptive injury states. We established a comprehensive and spatially-anchored epigenomic atlas to define the kidney's active, silent, and regulatory accessible chromatin regions across the genome. Using this atlas, we noted distinct control of adaptive injury in different epithelial cell types. A proximal tubule cell transcription factor network of ELF3 , KLF6 , and KLF10 regulated the transition between health and injury, while in thick ascending limb cells this transition was regulated by NR2F1 . Further, combined perturbation of ELF3 , KLF6 , and KLF10 distinguished two adaptive proximal tubular cell subtypes, one of which manifested a repair trajectory after knockout. This atlas will serve as a foundation to facilitate targeted cell-specific therapeutics by reprogramming gene regulatory networks.

4.
Dev Biol ; 464(2): 176-187, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32504627

RESUMO

Chromatin-remodeling complexes play critical roles in establishing gene expression patterns in response to developmental signals. How these epigenetic regulators determine the fate of progenitor cells during development of specific organs is not well understood. We found that genetic deletion of Brg1 (Smarca4), the core enzymatic protein in SWI/SNF, in nephron progenitor cells leads to severe renal hypoplasia. Nephron progenitor cells were depleted in Six2-Cre, Brg1flx/flx mice due to reduced cell proliferation. This defect in self-renewal, together with impaired differentiation resulted in a profound nephron deficit in Brg1 mutant kidneys. Sall1, a transcription factor that is required for expansion and maintenance of nephron progenitors, associates with SWI/SNF. Brg1 and Sall1 bind promoters of many progenitor cell genes and regulate expression of key targets that promote their proliferation.


Assuntos
Diferenciação Celular , Proliferação de Células , DNA Helicases/metabolismo , Néfrons/embriologia , Proteínas Nucleares/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células COS , Chlorocebus aethiops , DNA Helicases/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Transgênicos , Néfrons/citologia , Proteínas Nucleares/genética , Células-Tronco/citologia , Fatores de Transcrição/genética
5.
Dev Dyn ; 249(10): 1217-1242, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32492250

RESUMO

BACKGROUND: Understanding the mechanisms that regulate hair cell (HC) differentiation in the organ of Corti (OC) is essential to designing genetic therapies for hearing loss due to HC loss or damage. We have previously identified Fibroblast Growth Factor 20 (FGF20) as having a key role in HC and supporting cell differentiation in the mouse OC. To investigate the genetic landscape regulated by FGF20 signaling in OC progenitors, we employ Translating Ribosome Affinity Purification combined with Next Generation RNA Sequencing (TRAPseq) in the Fgf20 lineage. RESULTS: We show that TRAPseq targeting OC progenitors effectively enriched for RNA from this rare cell population. TRAPseq identified differentially expressed genes (DEGs) downstream of FGF20, including Etv4, Etv5, Etv1, Dusp6, Hey1, Hey2, Heyl, Tectb, Fat3, Cpxm2, Sall1, Sall3, and cell cycle regulators such as Cdc20. Analysis of Cdc20 conditional-null mice identified decreased cochlea length, while analysis of Sall1-null and Sall1-ΔZn2-10 mice, which harbor a mutation that causes Townes-Brocks syndrome, identified a decrease in outer hair cell number. CONCLUSIONS: We present two datasets: genes with enriched expression in OC progenitors, and DEGs downstream of FGF20 in the embryonic day 14.5 cochlea. We validate select DEGs via in situ hybridization and in vivo functional studies in mice.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Órgão Espiral/metabolismo , Ribossomos/metabolismo , Animais , Diferenciação Celular , Fatores de Crescimento de Fibroblastos/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Terapia Genética , Células Ciliadas Auditivas Externas/metabolismo , Audição , Camundongos , Camundongos Transgênicos , Mutação , Neurogênese , Órgão Espiral/embriologia , Fenótipo , Biossíntese de Proteínas , Análise de Sequência de RNA , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Physiol Rep ; 8(7): e14329, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32281744

RESUMO

Fibrosis is a final common pathway for many causes of progressive chronic kidney disease (CKD). Arginine-glycine-aspartic acid (RGD)-binding integrins are important mediators of the pro-fibrotic response by activating latent TGF-ß at sites of injury and by providing myofibroblasts information about the composition and stiffness of the extracellular matrix. Therefore, blockade of RGD-binding integrins may have therapeutic potential for CKD. To test this idea, we used small-molecule peptidomimetics that potently inhibit a subset of RGD-binding integrins in a murine model of kidney fibrosis. Acute kidney injury leading to fibrosis was induced by administration of aristolochic acid. Continuous subcutaneous administration of CWHM-12, an RGD integrin antagonist, for 28 days improved kidney function as measured by serum creatinine. CWHM-12 significantly reduced Collagen 1 (Col1a1) mRNA expression and scar collagen deposition in the kidney. Protein and gene expression markers of activated myofibroblasts, a major source of extracellular matrix deposition in kidney fibrosis, were diminished by treatment. RNA sequencing revealed that inhibition of RGD integrins influenced multiple pathways that determine the outcome of the response to injury and of repair processes. A second RGD integrin antagonist, CWHM-680, administered once daily by oral gavage was also effective in ameliorating fibrosis. We conclude that targeting RGD integrins with such small-molecule antagonists is a promising therapeutic approach in fibrotic kidney disease.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Antineoplásicos/farmacologia , Integrinas/antagonistas & inibidores , Oligopeptídeos/antagonistas & inibidores , Peptidomiméticos/farmacologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Colágeno/metabolismo , Fibrose/metabolismo , Fibrose/patologia , Fibrose/prevenção & controle , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia
7.
Transl Res ; 209: 90-104, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31085163

RESUMO

Chronic kidney disease (CKD) affects more than 20 million people in the United States and the global burden of this disorder is increasing. Many affected individuals will progress to end stage kidney disease necessitating dialysis or transplantation. CKD is also a major independent contributor to the risk of cardiovascular morbidity and mortality. Tubulointerstitial fibrosis is a final common pathway for most causes of progressive CKD. Currently, there are no clinically available therapies targeting fibrosis that can slow the decline in kidney function. Although it has long been known that TGF-ß signaling is a critical mediator of kidney fibrosis, translating this knowledge to the clinic has been challenging. In this review, we highlight some recent insights into the mechanisms of TGF-ß signaling that target activation of this cytokine at the site of injury or selectively inhibit pro-fibrotic gene expression. Molecules directed at these targets hold the promise of attaining therapeutic efficacy while limiting toxicity seen with global inhibition of TGF-ß. Kidney injury has profound epigenetic effects leading to altered expression of more than a thousand genes. We discuss how drugs targeting epigenetic modifications, some of which are in use for cancer therapy, have the potential to reprogram gene regulatory networks to favor adaptive repair and prevent fibrosis. The lack of reliable biomarkers of kidney fibrosis is a major limitation in designing clinical trials for testing CKD treatments. We conclude by reviewing recent advances in fibrosis biomarker development.


Assuntos
Rim/metabolismo , Rim/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Metilação de DNA/genética , Fibrose , Humanos , Modelos Biológicos
8.
Dev Biol ; 440(1): 13-21, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29705331

RESUMO

Formation of a functional kidney depends on the balance between renewal and differentiation of nephron progenitors. Failure to sustain this balance can lead to kidney failure or stem cell tumors. For nearly 60 years, we have known that signals from an epithelial structure known as the ureteric bud were essential for maintaining this balance. More recently it was discovered that one molecule, Wnt9b, was necessary for both renewal and differentiation of the nephron progenitor cells. How one ligand signaling through one transcription factor promoted two seemingly contradictory cellular processes was unclear. In this study, we show that Wnt9b/beta-catenin signaling alone is sufficient to promote both renewal and differentiation. Moreover, we show that discrete levels of beta-catenin can promote these two disparate fates, with low levels fostering progenitor renewal and high levels driving differentiation. These results provide insight into how Wnt9b regulates distinct target genes that balance nephron progenitor renewal and differentiation.


Assuntos
Néfrons/fisiologia , beta Catenina/metabolismo , beta Catenina/fisiologia , Animais , Diferenciação Celular/genética , Regulação da Expressão Gênica/genética , Rim/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Néfrons/embriologia , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia
9.
Mol Cancer ; 17(1): 78, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29625565

RESUMO

BACKGROUND: SALL1 is a multi-zinc finger transcription factor that regulates organogenesis and stem cell development, but the role of SALL1 in tumor biology and tumorigenesis remains largely unknown. METHODS: We analyzed SALL1 expression levels in human and murine breast cancer cells as well as cancer tissues from different types of breast cancer patients. Using both in vitro co-culture system and in vivo breast tumor models, we investigated how SALL1 expression in breast cancer cells affects tumor cell growth and proliferation, metastasis, and cell fate. Using the gain-of function and loss-of-function strategies, we dissected the molecular mechanism responsible for SALL1 tumor suppressor functions. RESULTS: We demonstrated that SALL1 functions as a tumor suppressor in breast cancer, which is significantly down-regulated in the basal like breast cancer and in estrogen receptor (ER), progesterone receptor (PR) and epidermal growth factor receptor 2 (HER2) triple negative breast cancer patients. SALL1 expression in human and murine breast cancer cells inhibited cancer cell growth and proliferation, metastasis, and promoted cell cycle arrest. Knockdown of SALL1 in breast cancer cells promoted cancer cell growth, proliferation, and colony formation. Our studies revealed that tumor suppression was mediated by recruitment of the Nucleosome Remodeling and Deacetylase (NuRD) complex by SALL1, which promoted cancer cell senescence. We further demonstrated that the mechanism of inhibition of breast cancer cell growth and invasion by SALL1-NuRD depends on the p38 MAPK, ERK1/2, and mTOR signaling pathways. CONCLUSION: Our studies indicate that the developmental control gene SALL1 plays a critical role in tumor suppression by recruiting the NuRD complex and thereby inducing cell senescence in breast cancer cells.


Assuntos
Regulação para Baixo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Técnicas de Cocultura , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo
10.
Oncotarget ; 9(7): 7442-7452, 2018 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-29484122

RESUMO

Similar signaling pathways could operate in both normal hematopoietic stem and progenitor cells (HSPCs) and leukemia stem cells (LSCs). Thus, targeting LSCs signaling without substantial toxicities to normal HSPCs remains challenging. SALL1, is a member of the transcriptional network that regulates stem cell pluripotency, and lacks significant expression in most adult tissues, including normal bone marrow (NBM). We examined the expression and functional characterization of SALL1 in NBM and in acute myeloid leukemia (AML) using in vitro and in vivo assays. We showed that SALL1 is expressed preferentially in LSCs- enriched CD34+CD38- cell subpopulation but not in NBM. SALL1 inhibition resulted in decreased cellular proliferation and in inferior AML engraftment in NSG mice and it was also associated with upregulation of PTEN and downregulation of m-TOR, ß-catenin, and NF-қB expression. These findings suggest that SALL1 inhibition interrupts leukemogenesis. Further studies to validate SALL1 as a potential biomarker for minimal residual disease (MRD) and to determine SALL1's role in prognostication are ongoing. Additionally, pre-clinical evaluation of SALL1 as a therapeutic target in AML is warranted.

11.
Am J Hum Genet ; 102(2): 249-265, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29395072

RESUMO

Townes-Brocks syndrome (TBS) is characterized by a spectrum of malformations in the digits, ears, and kidneys. These anomalies overlap those seen in a growing number of ciliopathies, which are genetic syndromes linked to defects in the formation or function of the primary cilia. TBS is caused by mutations in the gene encoding the transcriptional repressor SALL1 and is associated with the presence of a truncated protein that localizes to the cytoplasm. Here, we provide evidence that SALL1 mutations might cause TBS by means beyond its transcriptional capacity. By using proximity proteomics, we show that truncated SALL1 interacts with factors related to cilia function, including the negative regulators of ciliogenesis CCP110 and CEP97. This most likely contributes to more frequent cilia formation in TBS-derived fibroblasts, as well as in a CRISPR/Cas9-generated model cell line and in TBS-modeled mouse embryonic fibroblasts, than in wild-type controls. Furthermore, TBS-like cells show changes in cilia length and disassembly rates in combination with aberrant SHH signaling transduction. These findings support the hypothesis that aberrations in primary cilia and SHH signaling are contributing factors in TBS phenotypes, representing a paradigm shift in understanding TBS etiology. These results open possibilities for the treatment of TBS.


Assuntos
Anormalidades Múltiplas/genética , Anus Imperfurado/genética , Cílios/metabolismo , Perda Auditiva Neurossensorial/genética , Mutação/genética , Polegar/anormalidades , Fatores de Transcrição/genética , Animais , Citoplasma/metabolismo , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Recém-Nascido , Camundongos , Fenótipo , Ligação Proteica , Proteômica , Transdução de Sinais
12.
Int Urol Nephrol ; 50(3): 501-508, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29404927

RESUMO

BACKGROUND: Fast glomerular filtration rate (GFR) decline is associated with adverse outcomes, but the associated risk factors among patients without chronic kidney disease (CKD) are not well defined. METHODS: From a primary care registry of 37,796, we identified 2219 (6%) adults with at least three estimated (e)GFR values and a baseline eGFR between 60 and 119 ml/min/1.73 m2 during an observation period of 8 years. We defined fast GFR decline as > 5 ml/min/1.73 m2 per year. The outcome measure was incident CKD (eGFR < 60 ml/min/1.73 m2). Clinical and demographic characteristics were compared using Chi-square and independent-samples t tests. RESULTS: Older age, African-American race, unmarried status, hypertension and type 2 diabetes were more common in both fast decliners and those who developed incident CKD (p < 0.0001 to < 0.05). Lower neighborhood socioeconomic status, current smoking and baseline eGFR 90-119 ml/min/1.73 m2 were associated with fast decline (p < 0.01), while baseline eGFR 60-74 ml/min/1.73 m2 with incident CKD (p < 0.05). In multivariate regression models, among fast decliners with mildly reduced baseline eGFR (60-89 ml/min/1.73 m2), older age was significantly associated with incident CKD [odds ratio (OR) 1.04; 95% CI 1.01-1.08], and among those with normal baseline eGFR (≥ 90-119 ml/min/1.73 m2), type 2 diabetes was significantly associated with incident CKD (OR 3.83; 95% CI 1.35-10.89). CONCLUSIONS: Among primary care patients without CKD, GFR is checked infrequently. We have identified patients at high risk of progressive CKD, in whom we suggest a closer monitoring of renal function.


Assuntos
Diabetes Mellitus Tipo 2/epidemiologia , Taxa de Filtração Glomerular , Atenção Primária à Saúde , Insuficiência Renal Crônica/epidemiologia , Insuficiência Renal Crônica/fisiopatologia , Adulto , Fatores Etários , Idoso , Progressão da Doença , Feminino , Humanos , Hipertensão/epidemiologia , Incidência , Masculino , Estado Civil , Pessoa de Meia-Idade , Sistema de Registros , Características de Residência , Fatores de Risco , Fumar/epidemiologia , Fatores de Tempo , Estados Unidos/epidemiologia
13.
BMC Med Genet ; 18(1): 125, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29110636

RESUMO

BACKGROUND: Townes-Brocks syndrome (TBS) is a rare autosomal dominant condition characterized by renal, anal, limb, and auditory abnormalities. TBS diagnosis can be challenging in settings where genetic analysis is not readily available. TBS traits overlap with those of Goldenhar and VACTERL syndromes. CASE PRESENTATION: Here, we present the case of a 5-year-old Brazilian boy born with an anorectal abnormality, limb and external ears malformations, genitourinary anomalies, and a congenital heart defect. Genetic analysis revealed a SALL1 nonsense mutation. The case is discussed in the context of the current literature. CONCLUSIONS: Because of the variability in TBS clinical presentation, genetic analysis is key to the differential diagnosis of TBS relative to phenotypically similar syndromes.


Assuntos
Anormalidades Múltiplas/genética , Anus Imperfurado/genética , Códon sem Sentido , Predisposição Genética para Doença/genética , Perda Auditiva Neurossensorial/genética , Polegar/anormalidades , Fatores de Transcrição/genética , Anormalidades Múltiplas/diagnóstico , Canal Anal/anormalidades , Anus Imperfurado/diagnóstico , Brasil , Pré-Escolar , Diagnóstico Diferencial , Esôfago/anormalidades , Genótipo , Perda Auditiva Neurossensorial/diagnóstico , Cardiopatias Congênitas/diagnóstico , Humanos , Rim/anormalidades , Deformidades Congênitas dos Membros/diagnóstico , Masculino , Fenótipo , Coluna Vertebral/anormalidades , Traqueia/anormalidades
14.
Development ; 144(17): 3080-3094, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28760814

RESUMO

The formation of the proper number of nephrons requires a tightly regulated balance between renal progenitor cell self-renewal and differentiation. The molecular pathways that regulate the transition from renal progenitor to renal vesicle are not well understood. Here, we show that Sall1interacts with the nucleosome remodeling and deacetylase complex (NuRD) to inhibit premature differentiation of nephron progenitor cells. Disruption of Sall1-NuRD in vivo in knock-in mice (ΔSRM) resulted in accelerated differentiation of nephron progenitors and bilateral renal hypoplasia. Transcriptional profiling of mutant kidneys revealed a striking pattern in which genes of the glomerular and proximal tubule lineages were either unchanged or upregulated, and those in the loop of Henle and distal tubule lineages were downregulated. These global changes in gene expression were accompanied by a significant decrease in THP-, NKCC2- and AQP1-positive loop of Henle nephron segments in mutant ΔSRM kidneys. These findings highlight an important function of Sall1-NuRD interaction in the regulation of Six2-positive multipotent renal progenitor cells and formation of the loop of Henle.


Assuntos
Alça do Néfron/embriologia , Alça do Néfron/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Células-Tronco Multipotentes/citologia , Organogênese , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Biomarcadores/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Proliferação de Células/genética , Regulação da Expressão Gênica no Desenvolvimento , Ontologia Genética , Homozigoto , Túbulos Renais/metabolismo , Alça do Néfron/anormalidades , Camundongos , Células-Tronco Multipotentes/metabolismo , Mutação/genética , Organogênese/genética , Ligação Proteica/genética , Fatores de Transcrição/química , Ureter/embriologia , Ureter/metabolismo
15.
Eur Heart J ; 38(9): 675-686, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26491108

RESUMO

AIMS: Congenital anomalies of arterial valves are common birth defects, leading to valvar stenosis. With no pharmaceutical treatment that can prevent the disease progression, prosthetic replacement is the only choice of treatment, incurring considerable morbidity and mortality. Animal models presenting localized anomalies and stenosis of congenital arterial valves similar to that of humans are critically needed research tools to uncover developmental molecular mechanisms underlying this devastating human condition. METHODS AND RESULTS: We generated and characterized mouse models with conditionally altered Notch signalling in endothelial or interstitial cells of developing valves. Mice with inactivation of Notch1 signalling in valvar endothelial cells (VEC) developed congenital anomalies of arterial valves including bicuspid aortic valves and valvar stenosis. Notch1 signalling in VEC was required for repressing proliferation and activating apoptosis of valvar interstitial cells (VIC) after endocardial-to-mesenchymal transformation (EMT). We showed that Notch signalling regulated Tnfα expression in vivo, and Tnf signalling was necessary for apoptosis of VIC and post-EMT development of arterial valves. Furthermore, activation or inhibition of Notch signalling in cultured pig aortic VEC-promoted or suppressed apoptosis of VIC, respectively. CONCLUSION: We have now met the need of critical animal models and shown that Notch-Tnf signalling balances proliferation and apoptosis for post-EMT development of arterial valves. Our results suggest that mutations in its components may lead to congenital anomaly of aortic valves and valvar stenosis in humans.


Assuntos
Estenose da Valva Aórtica/etiologia , Receptor Notch1/metabolismo , Animais , Valva Aórtica/anormalidades , Estenose da Valva Aórtica/embriologia , Estenose da Valva Aórtica/fisiopatologia , Apoptose/fisiologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Homeostase/fisiologia , Células-Tronco Mesenquimais/fisiologia , Camundongos Knockout , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
16.
Am J Physiol Renal Physiol ; 309(10): F852-63, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26311113

RESUMO

It has been postulated that developmental pathways are reutilized during repair and regeneration after injury, but functional analysis of many genes required for kidney formation has not been performed in the adult organ. Mutations in SALL1 cause Townes-Brocks syndrome (TBS) and nonsyndromic congenital anomalies of the kidney and urinary tract, both of which lead to childhood kidney failure. Sall1 is a transcriptional regulator that is expressed in renal progenitor cells and developing nephrons in the embryo. However, its role in the adult kidney has not been investigated. Using a mouse model of TBS (Sall1TBS), we investigated the role of Sall1 in response to acute kidney injury. Our studies revealed that Sall1 is expressed in terminally differentiated renal epithelia, including the S3 segment of the proximal tubule, in the mature kidney. Sall1TBS mice exhibited significant protection from ischemia-reperfusion injury and aristolochic acid-induced nephrotoxicity. This protection from acute injury is seen despite the presence of slowly progressive chronic kidney disease in Sall1TBS mice. Mice containing null alleles of Sall1 are not protected from acute kidney injury, indicating that expression of a truncated mutant protein from the Sall1TBS allele, while causative of congenital anomalies, protects the adult kidney from injury. Our studies further revealed that basal levels of the preconditioning factor heme oxygenase-1 are elevated in Sall1TBS kidneys, suggesting a mechanism for the relative resistance to injury in this model. Together, these studies establish a functional role for Sall1 in the response of the adult kidney to acute injury.


Assuntos
Anormalidades Múltiplas/metabolismo , Injúria Renal Aguda/metabolismo , Anus Imperfurado/metabolismo , Perda Auditiva Neurossensorial/metabolismo , Proteínas Mutantes/metabolismo , Polegar/anormalidades , Fatores de Transcrição/metabolismo , Anormalidades Múltiplas/genética , Injúria Renal Aguda/genética , Animais , Anus Imperfurado/genética , Modelos Animais de Doenças , Perda Auditiva Neurossensorial/genética , Heme Oxigenase-1/genética , Camundongos Transgênicos , Mutação/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Fatores de Transcrição/genética
17.
J Nephrol ; 28(2): 193-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25348221

RESUMO

BACKGROUND AND OBJECTIVES: Acute kidney injury (AKI) after cardiac bypass surgery (CABG) is common and carries a significant association with morbidity and mortality. Since minocycline therapy attenuates kidney injury in animal models of AKI, we tested its effects in patients undergoing CABG. DESIGN, SETTING, PARTICIPANTS AND MEASUREMENTS: This is a randomized, double-blinded, placebo-controlled, multi-center study. We screened high risk patients who were scheduled to undergo CABG in two medical centers between Jan 2008 and June 2011. 40 patients were randomized and 19 patients in each group completed the study. Minocycline prophylaxis was given twice daily, at least for four doses prior to CABG. Primary outcome was defined as AKI [0.3 mg/dl increase in creatinine (Cr)] within 5 days after surgery. Daily serum Cr for 5 days, various clinical and hemodynamic measures and length of stay were recorded. RESULTS: The two groups had similar baseline and intra-operative characteristics. The primary outcome occurred in 52.6% of patients in the minocycline group as compared to 36.8% of patients in the placebo group (p = 0.51). Peak Cr was 1.6 ± 0.7 vs. 1.5 ± 0.7 mg/dl (p = 0.45) in minocycline and placebo groups, respectively. Death at 30 days occurred in 0 vs. 10.5% in the minocycline and placebo groups, respectively (p = 0.48). There were no differences in post-operative length of stay, and cardiovascular events between the two groups. There was a trend towards lower diastolic pulmonary artery pressure [16.8 ± 4.7 vs. 20.7 ± 6.6 mmHg (p = 0.059)] and central venous pressure [11.8 ± 4.3 vs. 14.6 ± 5.6 mmHg (p = 0.13)] in the minocycline group compared to placebo on the first day after surgery. CONCLUSIONS: Minocycline did not protect against AKI post-CABG.


Assuntos
Injúria Renal Aguda/prevenção & controle , Antibacterianos/uso terapêutico , Ponte de Artéria Coronária/efeitos adversos , Minociclina/uso terapêutico , Injúria Renal Aguda/sangue , Injúria Renal Aguda/etiologia , Idoso , Pressão Arterial/efeitos dos fármacos , Pressão Venosa Central/efeitos dos fármacos , Ponte de Artéria Coronária/mortalidade , Creatinina/sangue , Método Duplo-Cego , Feminino , Humanos , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Projetos Piloto
18.
Transl Res ; 165(1): 36-47, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24880148

RESUMO

The nucleosome remodeling and deacetylase (NuRD) complex is one of the major chromatin remodeling complexes found in cells. It plays an important role in regulating gene transcription, genome integrity, and cell cycle progression. Through its impact on these basic cellular processes, increasing evidence indicates that alterations in the activity of this macromolecular complex can lead to developmental defects, oncogenesis, and accelerated aging. Recent genetic and biochemical studies have elucidated the mechanisms of NuRD action in modifying the chromatin landscape. These advances have the potential to lead to new therapeutic approaches to birth defects and cancer.


Assuntos
Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Montagem e Desmontagem da Cromatina , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Feminino , Humanos , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/química , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Neoplasias/genética , Neoplasias/metabolismo , Gravidez , Pesquisa Translacional Biomédica
19.
Development ; 141(5): 1047-58, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24550112

RESUMO

The formation of the proper number of functional nephrons requires a delicate balance between renal progenitor cell self-renewal and differentiation. The molecular factors that regulate the dramatic expansion of the progenitor cell pool and differentiation of these cells into nephron precursor structures (renal vesicles) are not well understood. Here we show that Sall1, a nuclear transcription factor, is required to maintain the stemness of nephron progenitor cells. Transcriptional profiling of Sall1 mutant cells revealed a striking pattern, marked by the reduction of progenitor genes and amplified expression of renal vesicle differentiation genes. These global changes in gene expression were accompanied by ectopic differentiation at E12.5 and depletion of Six2+Cited1+ cap mesenchyme progenitor cells. These findings highlight a novel role for Sall1 in maintaining the stemness of the progenitor cell pool by restraining their differentiation into renal vesicles.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Diferenciação Celular/genética , Feminino , Imuno-Histoquímica , Hibridização In Situ , Rim/citologia , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição/genética
20.
PLoS One ; 7(8): e43098, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912798

RESUMO

During kidney development, canonical Wnt signaling activates differentiation, while the transcription factor Six2 maintains the progenitor pool. These opposing signals help to regulate nephron formation and ensure the full complement of nephrons are formed. Since these two factors control differing fates in kidney mesenchyme, we hypothesized that overexpression of Wnt9b in Six2-expressing cells would disrupt kidney formation and may alter cell differentiation decisions in other tissues. We created a transgenic mouse that conditionally expressed the canonical Wnt ligand in the developing kidney, Wnt9b. The transgene is activated by cre recombinase and expresses GFP. We first tested its biological activity using Hoxb7-cre and found that transgenic Wnt9b was capable of inducing differentiation genes and of rescuing kidney development in Wnt9b(-/-) homozygous deficient mice. In contrast, expression of Wnt9b in cells using Six2-cre caused gastrointestinal distress and severe renal failure in adult mice. Transgenic kidneys had numerous cystic tubules and elevated creatinine values (0.652 ± 0.044) compared to wild-type mice (0.119 ± 0.002). These animals also exhibited a malformed pyloric sphincter, duodenogastric reflux, and a transformation of the distal stomach into proximal fate. The gene expression changes observed for the Wnt9b:EGFP transgene were compared to a stabilized ß-catenin allele to determine that Wnt9b is activating the canonical Wnt pathway in the tissues analyzed. These results demonstrate that expression of Wnt9b in Six2-positive cells disrupts cell fate decisions in the kidney and the gastrointestinal tract.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Rim/embriologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Proteínas Wnt/metabolismo , Animais , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/metabolismo , Hibridização In Situ , Rim/citologia , Camundongos , Camundongos Transgênicos , Piloro/anormalidades , Estômago/embriologia , Transgenes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...