Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; : 107520, 2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38950862

RESUMO

Acute kidney injury (AKI) is a common condition associated with significant morbidity, mortality, and cost. Injured kidney tissue can regenerate after many forms of AKI. However, there are no treatments in routine clinical practice to encourage recovery. In part, this shortcoming is due to an incomplete understanding of the genetic mechanisms that orchestrate kidney recovery. The advent of high-throughput sequencing technologies and genetic mouse models has opened an unprecedented window into the transcriptional dynamics that accompany both successful and maladaptive repair. AKI recovery shares similar cell-state transformations with kidney development, which can suggest common mechanisms of gene regulation. Several powerful bioinformatic strategies have been developed to infer the activity of gene regulatory networks by combining multiple forms of sequencing data at single-cell resolution. These studies highlight shared stress responses but also key changes in gene regulatory networks controlling metabolism. Furthermore, chromatin immunoprecipitation studies in injured kidneys have revealed dynamic epigenetic modifications at enhancer elements near target genes. This review will highlight how these studies have enhanced our understanding of gene regulation in injury response and regeneration.

2.
Am J Physiol Renal Physiol ; 326(2): F178-F188, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-37994409

RESUMO

Chronic kidney disease is increasing at an alarming rate and correlates with the increase in diabetes, obesity, and hypertension that disproportionately impact socioeconomically disadvantaged communities. Iron plays essential roles in many biological processes including oxygen transport, mitochondrial function, cell proliferation, and regeneration. However, excess iron induces the generation and propagation of reactive oxygen species, which lead to oxidative stress, cellular damage, and ferroptosis. Iron homeostasis is regulated in part by the kidney through iron resorption from the glomerular filtrate and exports into the plasma by ferroportin (FPN). Yet, the impact of iron overload in the kidney has not been addressed. To test more directly whether excess iron accumulation is toxic to kidneys, we generated a kidney proximal tubule-specific knockout of FPN. Despite significant intracellular iron accumulation in FPN mutant tubules, basal kidney function was not measurably different from wild type kidneys. However, upon induction of acute kidney injury (AKI), FPN mutant kidneys exhibited significantly more damage and failed recovery, evidence for ferroptosis, and increased fibrosis. Thus, disruption of iron export in proximal tubules, leading to iron overload, can significantly impair recovery from AKI and can contribute to progressive renal damage indicative of chronic kidney disease. Understanding the mechanisms that regulate iron homeostasis in the kidney may provide new therapeutic strategies for progressive kidney disease and other ferroptosis-associated disorders.NEW & NOTEWORTHY Physiological iron homeostasis depends in part on renal resorption and export into the plasma. We show that specific deletion of iron exporters in the proximal tubules sensitizes cells to injury and inhibits recovery. This can promote a chronic kidney disease phenotype. Our paper demonstrates the need for iron balance in the proximal tubules to maintain and promote healthy recovery after acute kidney injury.


Assuntos
Injúria Renal Aguda , Proteínas de Transporte de Cátions , Sobrecarga de Ferro , Insuficiência Renal Crônica , Humanos , Rim/metabolismo , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Ferro/metabolismo , Sobrecarga de Ferro/metabolismo , Homeostase/fisiologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo
3.
Kidney Int ; 105(2): 312-327, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37977366

RESUMO

Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part, this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Identifying the molecular and genetic regulators unique to nephron segments that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. Here, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI and found them upregulated after severe AKI and correlated with chronic injury. Surprisingly, proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to pre-conditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of proximal tubule cells in the S3 segment that displayed features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic pre-conditioning, and female sex. Thus, our results identified a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both the injury response and protection from ischemic AKI.


Assuntos
Injúria Renal Aguda , Túbulos Renais Proximais , Fator de Transcrição PAX2 , Fator de Transcrição PAX8 , Insuficiência Renal Crônica , Animais , Feminino , Camundongos , Injúria Renal Aguda/complicações , Injúria Renal Aguda/genética , Isquemia/complicações , Túbulos Renais Proximais/patologia , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/genética , Traumatismo por Reperfusão/genética , Fator de Transcrição PAX8/genética , Fator de Transcrição PAX8/metabolismo , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX2/metabolismo
4.
Dev Biol ; 472: 18-29, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33428890

RESUMO

The vertebrate eye anlage grows out of the brain and folds into bilayered optic cups. The eye is patterned along multiple axes, precisely controlled by genetic programs, to delineate neural retina, pigment epithelium, and optic stalk tissues. Pax genes encode developmental regulators of key morphogenetic events, with Pax2 being essential for interpreting inductive signals, including in the eye. PAX2 mutations cause ocular coloboma, when the ventral optic fissure fails to close. Previous studies established that Pax2 is necessary for fissure closure and to maintain the neural retina -- glial optic stalk boundary. Using a Pax2GFP/+ knock-in allele we discovered that the mutant optic nerve head (ONH) lacks molecular boundaries with the retina and RPE, rendering the ONH larger than normal. This was preceded by ventronasal cup mispatterning, a burst of overproliferation and followed by optic cup apoptosis. Our findings support the hypothesis that ONH cells are tripotential, requiring Pax2 to remain committed to glial fates. This work extends current models of ocular development, contributes to broader understanding of tissue boundary formation and informs the underlying mechanisms of human coloboma.


Assuntos
Olho/embriologia , Olho/metabolismo , Disco Óptico/embriologia , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX2/metabolismo , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Proliferação de Células/genética , Coloboma/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Disco Óptico/anormalidades , Disco Óptico/citologia , Retina/embriologia , Células-Tronco/metabolismo
5.
J Am Soc Nephrol ; 31(6): 1212-1225, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32381599

RESUMO

BACKGROUND: As the glomerular filtrate passes through the nephron and into the renal medulla, electrolytes, water, and urea are reabsorbed through the concerted actions of solute carrier channels and aquaporins at various positions along the nephron and in the outer and inner medulla. Proliferating stem cells expressing the nuclear transcription factor Pax2 give rise to renal epithelial cells. Pax2 expression ends once the epithelial cells differentiate into mature proximal and distal tubules, whereas expression of the related Pax8 protein continues. The collecting tubules and renal medulla are derived from Pax2-positive ureteric bud epithelia that continue to express Pax2 and Pax8 in adult kidneys. Despite the crucial role of Pax2 in renal development, functions for Pax2 or Pax8 in adult renal epithelia have not been established. METHODS: To examine the roles of Pax2 and Pax8 in the adult mouse kidney, we deleted either Pax2, Pax8, or both genes in adult mice and examined the resulting phenotypes and changes in gene expression patterns. We also explored the mechanism of Pax8-mediated activation of potential target genes in inner medullary collecting duct cells. RESULTS: Mice with induced deletions of both Pax2 and Pax8 exhibit severe polyuria that can be attributed to significant changes in the expression of solute carriers, such as the urea transporters encoded by Slc14a2, as well as aquaporins within the inner and outer medulla. Furthermore, Pax8 expression is induced by high-salt levels in collecting duct cells and activates the Slc14a2 gene by recruiting a histone methyltransferase complex to the promoter. CONCLUSIONS: These data reveal novel functions for Pax proteins in adult renal epithelia that are essential for retaining water and concentrating urine.


Assuntos
Aquaporinas/fisiologia , Capacidade de Concentração Renal/fisiologia , Rim/fisiologia , Proteínas de Membrana Transportadoras/fisiologia , Fator de Transcrição PAX2/fisiologia , Fator de Transcrição PAX8/fisiologia , Animais , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Osmorregulação , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX8/genética , Transportadores de Ureia
6.
J Biol Chem ; 294(9): 3125-3136, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30602563

RESUMO

Bone morphogenetic protein (BMP) signaling is critical in renal development and disease. In animal models of chronic kidney disease (CKD), re-activation of BMP signaling is reported to be protective by promoting renal repair and regeneration. Clinical use of recombinant BMPs, however, requires harmful doses to achieve efficacy and is costly because of BMPs' complex synthesis. Therefore, alternative strategies are needed to harness the beneficial effects of BMP signaling in CKD. Key aspects of the BMP signaling pathway can be regulated by both extracellular and intracellular molecules. In particular, secreted proteins like noggin and chordin inhibit BMP activity, whereas kielin/chordin-like proteins (KCP) enhance it and attenuate kidney fibrosis or CKD. Clinical development of KCP, however, is precluded by its size and complexity. Therefore, we propose an alternative strategy to enhance BMP signaling by using small molecules, which are simpler to synthesize and more cost-effective. To address our objective, here we developed a small-molecule high-throughput screen (HTS) with human renal cells having an integrated luciferase construct highly responsive to BMPs. We demonstrate the activity of a potent benzoxazole compound, sb4, that rapidly stimulated BMP signaling in these cells. Activation of BMP signaling by sb4 increased the phosphorylation of key second messengers (SMAD-1/5/9) and also increased expression of direct target genes (inhibitors of DNA binding, Id1 and Id3) in canonical BMP signaling. Our results underscore the feasibility of utilizing HTS to identify compounds that mimic key downstream events of BMP signaling in renal cells and have yielded a lead BMP agonist.


Assuntos
Benzoxazóis/farmacologia , Proteínas Morfogenéticas Ósseas/agonistas , Proteínas Morfogenéticas Ósseas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Fosfoproteínas/metabolismo , Proteínas Smad/metabolismo
7.
Mol Cell ; 45(2): 185-95, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22169276

RESUMO

The repression of transcription, through the concerted actions of tissue specific DNA binding proteins, Polycomb repressor complexes, and DNA methylation, is essential for maintaining stem cell pluripotency and for cell fate specification in development. In this report, we show that recruitment of the co-repressor protein Grg4 to a Pax DNA-binding site displaces the adaptor protein PTIP and a histone H3K4me complex. Grg4 recruits the arginine methyltransferase PRMT5 to chromatin resulting in symmetric H4R3 dimethylation. PRMT5 is essential for recruiting Polycomb proteins, in a Pax2/Grg4 dependent manner, which results in H3K27 methylation. These data define the early epigenetic events in response to Pax/Grg mediated gene repression and demonstrate that a single DNA binding protein can recruit either an activator or a repressor complex depending on the availability of Grg4. These data suggest a model for understanding the initiation of Groucho/Grg/TLE mediated gene silencing.


Assuntos
Epigênese Genética , Histonas/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Proteínas de Transporte/metabolismo , Cromatina/metabolismo , Proteínas de Ligação a DNA , Inativação Gênica , Células HEK293 , Humanos , Metilação , Modelos Genéticos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas do Grupo Polycomb , Proteínas Metiltransferases/metabolismo , Proteínas Metiltransferases/fisiologia , Transporte Proteico , Proteína-Arginina N-Metiltransferases , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
8.
J Am Soc Nephrol ; 30(1): 80-94, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30545984

RESUMO

BACKGROUND: Renal interstitial fibrosis results from activation and proliferation of fibroblasts to myofibroblasts, secretion and accumulation of extracellular matrix, and displacement of normal renal tubules. In contrast to chronic renal disease, acute injury may be repaired, a process that includes a decrease in the number of myofibroblasts in the interstitium and degradation of the accumulated extracellular matrix, leaving little evidence of prior injury. METHODS: To investigate whether activated fibroblasts demonstrate changes in gene expression that correspond with regression after acute injury but are not observed in chronic models of fibrosis, we used microarrays to analyze gene expression patterns among fibroblast populations at different stages of injury or repair. We then mined the data for signaling pathways in fibroblasts corresponding to the acute proliferative, regression, and chronic phases of renal injury. RESULTS: We identified multiple gene clusters with changes that correlate with the three phases of renal injury, including changes in levels of receptors for the antifibrotic factor PGE2. In adult renal fibroblast cultures, PGE2 was able to upregulate many genes that are suppressed by the profibrotic cytokine TGF-ß, whereas many PGE2-downregulated genes were activated by TGF-ß. High levels of TGF-ß suppressed expression of a subset of PG receptors in fibroblast cultures, making these cells resistant to any effects of PGE2. CONCLUSIONS: Inherent gene expression changes in activated fibroblasts accompany the transition from AKI to repair and regeneration. In chronic models, however, activated fibroblasts are resistant to the antifibrotic effects of PGE2 due to suppression of a subset of PGE receptors.


Assuntos
Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Dinoprostona/farmacologia , Regulação da Expressão Gênica , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Fibroblastos/citologia , Fibrose/genética , Fibrose/patologia , Perfilação da Expressão Gênica , Imuno-Histoquímica , Camundongos , Miofibroblastos/citologia , Reação em Cadeia da Polimerase em Tempo Real/métodos , Transdução de Sinais/genética
9.
J Biol Chem ; 292(22): 9051-9062, 2017 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-28424263

RESUMO

Obesity and its associated complications such as insulin resistance and non-alcoholic fatty liver disease are reaching epidemic proportions. In mice, the TGF-ß superfamily is implicated in the regulation of white and brown adipose tissue differentiation. The kielin/chordin-like protein (KCP) is a secreted regulator of the TGF-ß superfamily pathways that can inhibit both TGF-ß and activin signals while enhancing bone morphogenetic protein (BMP) signaling. However, KCP's effects on metabolism and obesity have not been studied in animal models. Therefore, we examined the effects of KCP loss or gain of function in mice that were maintained on either a regular or a high-fat diet. KCP loss sensitized the mice to obesity and associated complications such as glucose intolerance and adipose tissue inflammation and fibrosis. In contrast, transgenic mice that expressed KCP in the kidney, liver, and adipose tissues were resistant to developing high-fat diet-induced obesity and had significantly reduced white adipose tissue. Moreover, KCP overexpression shifted the pattern of SMAD signaling in vivo, increasing the levels of phospho (P)-SMAD1 and decreasing P-SMAD3. Adipocytes in culture showed a cell-autonomous effect in response to added TGF-ß1 or BMP7. Metabolic profiling indicated increased energy expenditure in KCP-overexpressing mice and reduced expenditure in the KCP mutants with no effect on food intake or activity. These findings demonstrate that shifting the TGF-ß superfamily signaling with a secreted protein can alter the physiology and thermogenic properties of adipose tissue to reduce obesity even when mice are fed a high-fat diet.


Assuntos
Adipócitos/metabolismo , Proteínas de Transporte/metabolismo , Gorduras na Dieta/efeitos adversos , Síndrome Metabólica/metabolismo , Obesidade/metabolismo , Transdução de Sinais , Adipócitos/patologia , Animais , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/metabolismo , Proteínas de Transporte/genética , Gorduras na Dieta/farmacologia , Síndrome Metabólica/induzido quimicamente , Síndrome Metabólica/genética , Síndrome Metabólica/patologia , Camundongos , Camundongos Knockout , Obesidade/induzido quimicamente , Obesidade/genética , Obesidade/patologia , Especificidade de Órgãos/genética , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
10.
Kidney Int ; 94(2): 259-267, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29685496

RESUMO

Pax genes encode developmental regulators that are expressed in a variety of tissues and control critical events in morphogenesis. In the kidney, Pax2 and Pax8 are expressed in embryonic development and in specific renal diseases associated with aberrant epithelial cell proliferation. Prior genetic and cell biological studies suggest that reducing the activity of Pax proteins in renal cancer or in polycystic kidney disease can slow the progression of these conditions. The Pax proteins may be critical for providing tissue and locus specificity to recruit epigenetic modifiers that control gene expression and chromatin structure. Although they are nuclear, targeting Pax proteins to inhibit function may be feasible with small molecules. Such inhibition of Pax protein function may provide novel therapies for subsets of renal disorders that are tissue- and cell type-specific and avoid systemic effects on non-Pax-expressing cells and tissues. Given the paucity of effective treatments for renal cancer and cystic disease, the Pax family of proteins represents new pharmaceutical targets that merit exploration and further development.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Renais/tratamento farmacológico , Fator de Transcrição PAX2/antagonistas & inibidores , Fator de Transcrição PAX8/antagonistas & inibidores , Doenças Renais Policísticas/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Progressão da Doença , Epigênese Genética/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Rim/citologia , Rim/crescimento & desenvolvimento , Rim/metabolismo , Neoplasias Renais/genética , Neoplasias Renais/patologia , Terapia de Alvo Molecular/métodos , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX2/metabolismo , Fator de Transcrição PAX8/genética , Fator de Transcrição PAX8/metabolismo , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Domínios Proteicos/efeitos dos fármacos , Urotélio/citologia , Urotélio/efeitos dos fármacos , Urotélio/metabolismo , Urotélio/patologia
11.
Dev Biol ; 399(2): 296-305, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25617721

RESUMO

Activation of the Pax2 gene marks the intermediate mesoderm shortly after gastrulation, as the mesoderm becomes compartmentalized into paraxial, intermediate, and lateral plate. Using an EGFP knock-in allele of Pax2 to identify and sort cells of the intermediate mesodermal lineage, we compared gene expression patterns in EGFP positive cells that were heterozygous or homozygous null for Pax2. Thus, we identified critical regulators of intermediate mesoderm and kidney development whose expression depended on Pax2 function. In cell culture models, Pax2 is thought to recruit epigenetic modifying complex to imprint activating histone methylation marks through interactions with the adaptor protein PTIP. In kidney organ culture, conditional PTIP deletion showed that many Pax2 target genes, which were activated early in renal progenitor cells, remained on once activated, whereas Pax2 target genes expressed later in kidney development were unable to be fully activated without PTIP. In Pax2 mutants, we also identified a set of genes whose expression was up-regulated in EGFP positive cells and whose expression was consistent with a cell fate transformation to paraxial mesoderm and its derivatives. These data provide evidence that Pax2 specifies the intermediate mesoderm and renal epithelial cells through epigenetic mechanisms and in part by repressing paraxial mesodermal fate.


Assuntos
Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica/fisiologia , Rim/embriologia , Mesoderma/embriologia , Proteínas Nucleares/metabolismo , Fator de Transcrição PAX2/metabolismo , Células-Tronco/metabolismo , Animais , Western Blotting , Primers do DNA/genética , Proteínas de Ligação a DNA , Citometria de Fluxo , Regulação da Expressão Gênica/genética , Técnicas de Introdução de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hibridização In Situ , Rim/citologia , Mesoderma/citologia , Camundongos , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
J Biol Chem ; 290(11): 7185-94, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25631048

RESUMO

Pax genes encode developmental regulatory proteins that specify cell lineages and tissues in metazoans. Upon binding to DNA through the conserved paired domain, Pax proteins can recruit both activating and repressing complexes that imprint distinct patterns of histone methylation associated with either gene activation or silencing. How the switch from Pax-mediated activation to repression is regulated remains poorly understood. In this report, we identify the phosphatase PPM1B as an essential component of the Groucho4 repressor complex that is recruited by Pax2 to chromatin. PPM1B can dephosphorylate the Pax2 activation domain and displace the adaptor protein PTIP, thus inhibiting H3K4 methylation and gene activation. Loss of PPM1B prevents Groucho-mediated gene repression. Thus, PPM1B helps switch Pax2 from a transcriptional activator to a repressor protein. This can have profound implications for developmental regulation by Pax proteins and suggests a model for imprinting specific epigenetic marks depending on the availability of co-factors.


Assuntos
Proteínas de Transporte/metabolismo , Inativação Gênica , Proteínas Nucleares/metabolismo , Fator de Transcrição PAX2/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteínas Repressoras/metabolismo , Ativação Transcricional , Proteínas de Transporte/análise , Cromatina/metabolismo , Proteínas de Ligação a DNA , Células HEK293 , Humanos , Proteínas Nucleares/análise , Fator de Transcrição PAX2/análise , Fosfoproteínas Fosfatases/análise , Mapas de Interação de Proteínas , Proteína Fosfatase 2C , Proteínas Repressoras/análise
13.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G587-G598, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27514479

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is a common cause of chronic liver disease and is increasing with the rising rate of obesity in the developed world. Signaling pathways known to influence the rate of lipid deposition in liver, known as hepatic steatosis, include the transforming growth factor (TGF) superfamily, which function through the SMAD second messengers. The kielin/chordin-like protein (KCP) is a large secreted protein that can enhance bone morphogenetic protein signaling while suppressing TGF-ß signaling in cells and in genetically modified mice. In this report, we show that aging KCP mutant (Kcp-/-) mice are increasingly susceptible to developing hepatic steatosis and liver fibrosis. When young mice are put on a high-fat diet, Kcp-/- mice are also more susceptible to developing liver pathology, compared with their wild-type littermates. Furthermore, mice that express a Pepck-KCP transgene (KcpTg) in the liver are resistant to developing liver pathology even when fed a high-fat diet. Analyses of liver tissues reveal a significant reduction of P-Smad3, consistent with a role for KCP in suppressing TGF-ß signaling. Transcriptome analyses show that livers from Kcp-/- mice fed a normal diet are more like wild-type livers from mice fed a high-fat diet. However, the KCP transgene can suppress many of the changes in liver gene expression that are due to a high-fat diet. These data demonstrate that shifting the TGF-ß signaling paradigm with the secreted regulatory protein KCP can significantly alter the liver pathology in aging mice and in diet-induced NAFLD.


Assuntos
Proteínas de Transporte/metabolismo , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Transdução de Sinais/fisiologia , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Proteínas de Transporte/genética , Dieta Hiperlipídica , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Fígado/patologia , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Fosforilação , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo
14.
Dev Biol ; 373(1): 64-71, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23063797

RESUMO

The differentiation of mature sperm from male germ cells requires both chromatin remodeling and compaction as well as DNA double stranded break repair of sister chromatids. We examined the function of PTIP, a protein implicated in both DNA repair and histone methylation, during spermatogenesis by using a conditional, inducible mutation in adult male mice. Loss of PTIP led to the developmental arrest of spermatocytes, testicular atrophy, and infertility. By immunostaining with specific markers for different stages of spermatogenesis and for proteins involved in DNA damage and repair mechanisms, we conclude that the lack of PTIP results in genomic instability and DNA damage resulting in the cessation of spermatogenesis in meiosis I. These data underscore the importance of PTIP in the DNA repair process associated with the development of mature spermatozoa.


Assuntos
Proteínas de Transporte/genética , Dano ao DNA/genética , Reparo do DNA/genética , Instabilidade Genômica/genética , Meiose/fisiologia , Proteínas Nucleares/genética , Espermatogênese/genética , Testículo/metabolismo , Animais , Western Blotting , Primers do DNA/genética , Proteínas de Ligação a DNA , Perfilação da Expressão Gênica , Masculino , Meiose/genética , Camundongos , Microscopia de Fluorescência , Testosterona/sangue
15.
Hum Mol Genet ; 21(19): 4225-36, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22736030

RESUMO

Glutamine (Q) expansion diseases are a family of degenerative disorders caused by the lengthening of CAG triplet repeats present in the coding sequences of seemingly unrelated genes whose mutant proteins drive pathogenesis. Despite all the molecular evidence for the genetic basis of these diseases, how mutant poly-Q proteins promote cell death and drive pathogenesis remains controversial. In this report, we show a specific interaction between the mutant androgen receptor (AR), a protein associated with spinal and bulbar muscular atrophy (SBMA), and the nuclear protein PTIP (Pax Transactivation-domain Interacting Protein), a protein with an unusually long Q-rich domain that functions in DNA repair. Upon exposure to ionizing radiation, PTIP localizes to nuclear foci that are sites of DNA damage and repair. However, the expression of poly-Q AR sequesters PTIP away from radiation-induced nuclear foci. This results in sensitivity to DNA-damaging agents and chromosomal instabilities. In a mouse model of SBMA, evidence for DNA damage is detected in muscle cell nuclei and muscular atrophy is accelerated when one copy of the gene encoding PTIP is removed. These data provide a new paradigm for understanding the mechanisms of cellular degeneration observed in poly-Q expansion diseases.


Assuntos
Atrofia Bulboespinal Ligada ao X/genética , Atrofia Bulboespinal Ligada ao X/metabolismo , Proteínas de Transporte/metabolismo , Reparo do DNA , Instabilidade Genômica , Proteínas Nucleares/metabolismo , Peptídeos/genética , Receptores Androgênicos/metabolismo , Expansão das Repetições de Trinucleotídeos , Animais , Proteínas de Transporte/genética , Proteínas de Ligação a DNA , Humanos , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Androgênicos/genética
16.
Pediatr Nephrol ; 29(4): 589-95, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23996452

RESUMO

During embryonic development, DNA binding proteins help specify and restrict the fates of pluripotent stem cells. In the developing kidney, Pax2 proteins are among the earliest markers for the renal epithelial cell lineage, with expression in the mesenchyme and in proliferating epithelia. The Pax2 protein is essential for interpreting inductive signals emanating from the ureteric bud such that the kidney mesenchyme can convert to epithelia. The biochemistry of Pax protein function is being studied in a variety of model systems. Through interactions with the adaptor Pax transactivation-domain interacting protein (PTIP), Pax proteins can recruit members of the Trithorax family of histone methyltransferases to imprint activating epigenetic marks on chromatin. However, interactions with the corepressor Groucho-related gene-4 (Grg4) protein can inhibit activation and instead recruit Polycomb repressor complexes to promote target-gene silencing. We present a model whereby the regulated interactions of Pax proteins with available cofactor-mediated activation or gene silencing at different stages of development. The implications for establishing and maintaining the epigenome are discussed.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Rim/embriologia , Fatores de Transcrição Box Pareados/metabolismo , Animais , Humanos
17.
J Am Soc Nephrol ; 24(6): 897-905, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23539757

RESUMO

The secreted kielin/chordin-like (KCP) protein, one of a family of cysteine-rich proteins, suppresses TGF-ß signaling by sequestering the ligand from its receptor, but it enhances bone morphogenetic protein (BMP) signaling by promoting ligand-receptor interactions. Given the critical roles for TGF-ß and BMP proteins in enhancing or suppressing renal interstitial fibrosis, respectively, we examined whether secreted KCP could attenuate renal fibrosis in mouse models of chronic and acute disease. Transgenic mice that express KCP in adult kidneys showed significantly less expression of collagen IV, α-smooth muscle actin, and other markers of disease progression in the unilateral ureteral obstruction model of renal interstitial fibrosis. In the folic acid nephrotoxicity model of acute tubular necrosis, mice expressing KCP survived high doses of folic acid that were lethal for wild-type mice. With a lower dose of folic acid, mice expressing KCP exhibited improved renal recovery compared with wild-type mice. Thus, these data suggest that extracellular regulation of the TGF-ß/BMP signaling axis by KCP, and by extension possibly other cysteine-rich domain proteins, can attenuate both acute and chronic renal injury.


Assuntos
Injúria Renal Aguda/metabolismo , Proteínas de Transporte/metabolismo , Necrose Tubular Aguda/metabolismo , Insuficiência Renal Crônica/metabolismo , Transdução de Sinais/fisiologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Progressão da Doença , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Fibrose , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Necrose Tubular Aguda/patologia , Necrose Tubular Aguda/fisiopatologia , Camundongos , Camundongos Transgênicos , Cultura Primária de Células , Estrutura Terciária de Proteína , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/fisiopatologia , Fator de Crescimento Transformador beta/metabolismo , Transgenes/fisiologia
18.
Dev Biol ; 365(1): 241-50, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22410172

RESUMO

The Pax2 gene encodes a DNA binding protein with multiple functions in the developing intermediate mesoderm and urogenital tract. Loss of Pax2 in mice results in the complete absence of kidneys, ureters, and sex specific epithelial structures derived from the intermediate mesoderm in both males and females. In this report, we describe two new alleles of Pax2 created by inserting the enhanced green fluorescent protein coding region into the 5' untranslated leader sequence. One allele is a hypomorph that generates less protein and exhibits structural defects in kidneys and ureters upon homozygosity. A second allele is a true null that can be used to image Pax2 expressing cells in a mutant background. Organ culture and embryo analyses point to a loss of epithelial cell polarity and increased mobility in cells that have deleted Pax2 function. These experiments provide new insight into the role of Pax2 protein levels in determining correct renal architecture and cell fate. These new Pax2 alleles are valuable genetic reagents for in vivo studies of urogenital development.


Assuntos
Alelos , Regulação da Expressão Gênica no Desenvolvimento , Rim/metabolismo , Fator de Transcrição PAX2/genética , Fatores Etários , Envelhecimento/fisiologia , Animais , Feminino , Proteínas de Fluorescência Verde , Rim/embriologia , Masculino , Camundongos , Técnicas de Cultura de Órgãos , Fator de Transcrição PAX2/metabolismo
19.
J Biol Chem ; 287(25): 21290-302, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22556418

RESUMO

Transforming growth factor ß1 (TGF-ß) promotes renal interstitial fibrosis in vivo and the expression of mesenchymal genes in vitro; however, most of its direct targets in epithelial cells are still elusive. In a screen for genes directly activated by TGF-ß, we found that components of the Wnt signaling pathway, especially Wnt11, were targets of activation by TGF-ß and Smad3 in primary renal epithelial cells. In gain and loss of function experiments, Wnt11 mediated the actions of TGF-ß through enhanced activation of mesenchymal marker genes, such as Zeb1, Snail1, Pai1, and αSMA, without affecting Smad3 phosphorylation. Inhibition of Wnt11 by receptor knockdown or treatment with Wnt inhibitors limited the effects of TGF-ß on gene expression. We found no evidence that Wnt11 activated the canonical Wnt signaling pathway in renal epithelial cells; rather, the function of Wnt11 was mediated by the c-Jun N-terminal kinase (JNK) pathway. Consistent with the in vitro results, all the TGF-ß, Wnt11, and JNK targets were activated in a unilateral ureteral obstruction (UUO) model of renal fibrosis in vivo. Our findings demonstrated cooperativity among the TGF-ß, Wnt11, and JNK signaling pathways and suggest new targets for anti-fibrotic therapy in renal tissue.


Assuntos
Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Nefropatias/metabolismo , Rim/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/patologia , Fibrose , Técnicas de Silenciamento de Genes , Humanos , Rim/patologia , Nefropatias/genética , Nefropatias/patologia , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Fator de Crescimento Transformador beta1/genética , Obstrução Ureteral/genética , Obstrução Ureteral/patologia , Proteínas Wnt/genética
20.
Biochem Biophys Res Commun ; 440(3): 454-9, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-24099773

RESUMO

Groucho related genes encode transcriptional repressor proteins critical for normal developmental processes. The bone morphogenetic proteins belong to the transforming growth factor-ß (TGF-ß) superfamily and play important signaling roles in development and disease. However, the regulation of BMP signaling, especially within cells, is largely unknown. In this report, we show that expression of the Groucho related gene Grg4 robustly activates the expression of a BMP reporter gene, as well as enhancing and sustaining the upregulation of the endogenous Id1 gene induced by BMP7. BMP7 administration did not affect the endogenous level of Grg4 nor did it enhance the phosphorylation of receptor activated Smad proteins. Rather, Grg4 expression reduced the levels of the endogenous inhibitory Smad7, thus increasing the transcriptional responses mediated by BMP responsive sequences. The data point to a novel mechanisms for attenuating BMP signaling through altering the ratio of activating versus inhibitory Smad proteins.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Regulação da Expressão Gênica , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Proteína Smad7/metabolismo , Proteína Morfogenética Óssea 7/metabolismo , Genes Reporter , Células HEK293 , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Proteína Smad7/genética , Supressão Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA