Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(19): e2213696120, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37126682

RESUMO

To better understand the genetic basis of heart disease, we identified a variant in the Flightless-I homolog (FLII) gene that generates a R1243H missense change and predisposes to cardiac remodeling across multiple previous human genome-wide association studies (GWAS). Since this gene is of unknown function in the mammalian heart we generated gain- and loss-of-function genetically altered mice, as well as knock-in mice with the syntenic R1245H amino acid substitution, which showed that Flii protein binds the sarcomeric actin thin filament and influences its length. Deletion of Flii from the heart, or mice with the R1245H amino acid substitution, show cardiomyopathy due to shortening of the actin thin filaments. Mechanistically, Flii is a known actin binding protein that we show associates with tropomodulin-1 (TMOD1) to regulate sarcomere thin filament length. Indeed, overexpression of leiomodin-2 in the heart, which lengthens the actin-containing thin filaments, partially rescued disease due to heart-specific deletion of Flii. Collectively, the identified FLII human variant likely increases cardiomyopathy risk through an alteration in sarcomere structure and associated contractile dynamics, like other sarcomere gene-based familial cardiomyopathies.


Assuntos
Actinas , Cardiomiopatias , Humanos , Animais , Camundongos , Actinas/metabolismo , Sarcômeros/metabolismo , Estudo de Associação Genômica Ampla , Citoesqueleto de Actina/metabolismo , Cardiomiopatias/metabolismo , Mamíferos/genética , Proteínas dos Microfilamentos/metabolismo , Transativadores/metabolismo , Tropomodulina/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas Musculares/metabolismo
2.
J Biol Chem ; 299(12): 105426, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37926281

RESUMO

S-palmitoylation is a reversible lipid modification catalyzed by 23 S-acyltransferases with a conserved zinc finger aspartate-histidine-histidine-cysteine (zDHHC) domain that facilitates targeting of proteins to specific intracellular membranes. Here we performed a gain-of-function screen in the mouse and identified the Golgi-localized enzymes zDHHC3 and zDHHC7 as regulators of cardiac hypertrophy. Cardiomyocyte-specific transgenic mice overexpressing zDHHC3 show cardiac disease, and S-acyl proteomics identified the small GTPase Rac1 as a novel substrate of zDHHC3. Notably, cardiomyopathy and congestive heart failure in zDHHC3 transgenic mice is preceded by enhanced Rac1 S-palmitoylation, membrane localization, activity, downstream hypertrophic signaling, and concomitant induction of all Rho family small GTPases whereas mice overexpressing an enzymatically dead zDHHC3 mutant show no discernible effect. However, loss of Rac1 or other identified zDHHC3 targets Gαq/11 or galectin-1 does not diminish zDHHC3-induced cardiomyopathy, suggesting multiple effectors and pathways promoting decompensation with sustained zDHHC3 activity. Genetic deletion of Zdhhc3 in combination with Zdhhc7 reduces cardiac hypertrophy during the early response to pressure overload stimulation but not over longer time periods. Indeed, cardiac hypertrophy in response to 2 weeks of angiotensin-II infusion is not diminished by Zdhhc3/7 deletion, again suggesting other S-acyltransferases or signaling mechanisms compensate to promote hypertrophic signaling. Taken together, these data indicate that the activity of zDHHC3 and zDHHC7 at the cardiomyocyte Golgi promote Rac1 signaling and maladaptive cardiac remodeling, but redundant signaling effectors compensate to maintain cardiac hypertrophy with sustained pathological stimulation in the absence of zDHHC3/7.


Assuntos
Cardiomiopatias , Miócitos Cardíacos , Animais , Camundongos , Aciltransferases/genética , Aciltransferases/metabolismo , Cardiomegalia/metabolismo , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Histidina/metabolismo , Lipoilação , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo
3.
Circulation ; 147(9): 746-758, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36695175

RESUMO

BACKGROUND: Acute kidney injury (AKI) is a short-term life-threatening condition that, if survived, can lead to renal insufficiency and development of chronic kidney disease. The pathogenesis of AKI and chronic kidney disease involves direct effects on the heart and the development of hypertrophy and cardiomyopathy. METHODS: We used mouse models of ischemia/reperfusion AKI and unilateral ureteral obstruction to investigate the role of IL-33 (interleukin-33) and its receptor-encoding gene Il1rl1 (also called ST2L [suppression of tumorigenicity 2]) in cardiac remodeling after AKI. Mice with cell type-specific genetic disruption of the IL-33/ST2L axis were used, and IL-33 monoclonal antibody, adeno-associated virus encoding IL-33 or ST2L, and recombinant IL-33, as well. RESULTS: Mice deficient in Il33 were refractory to cardiomyopathy associated with 2 models of kidney injury. Treatment of mice with monoclonal IL-33 antibody also protected the heart after AKI. Moreover, overexpression of IL-33 or injection of recombinant IL-33 induced cardiac hypertrophy or cardiomyopathy, but not in mice lacking Il1rl1. AKI-induced cardiomyopathy was also reduced in mice with cardiac myocyte-specific deletion of Il1rl1 but not in endothelial cell- or fibroblast-specific deletion of Il1rl1. Last, overexpression of the ST2L receptor in cardiac myocytes recapitulated induction of cardiac hypertrophy. CONCLUSIONS: These results indicate that IL-33 released from the kidney during AKI underlies cardiorenal syndrome by directly signaling to cardiac myocytes, suggesting that antagonism of IL-33/ST2 axis would be cardioprotective in patients with kidney disease.


Assuntos
Injúria Renal Aguda , Cardiomiopatias , Interleucina-33 , Insuficiência Renal Crônica , Traumatismo por Reperfusão , Animais , Camundongos , Injúria Renal Aguda/etiologia , Cardiomegalia/patologia , Cardiomiopatias/genética , Cardiomiopatias/complicações , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Rim/patologia , Miócitos Cardíacos/patologia , Insuficiência Renal Crônica/complicações , Traumatismo por Reperfusão/patologia
4.
Nature ; 509(7500): 337-41, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24805242

RESUMO

If and how the heart regenerates after an injury event is highly debated. c-kit-expressing cardiac progenitor cells have been reported as the primary source for generation of new myocardium after injury. Here we generated two genetic approaches in mice to examine whether endogenous c-kit(+) cells contribute differentiated cardiomyocytes to the heart during development, with ageing or after injury in adulthood. A complementary DNA encoding either Cre recombinase or a tamoxifen-inducible MerCreMer chimaeric protein was targeted to the Kit locus in mice and then bred with reporter lines to permanently mark cell lineage. Endogenous c-kit(+) cells did produce new cardiomyocytes within the heart, although at a percentage of approximately 0.03 or less, and if a preponderance towards cellular fusion is considered, the percentage falls to below approximately 0.008. By contrast, c-kit(+) cells amply generated cardiac endothelial cells. Thus, endogenous c-kit(+) cells can generate cardiomyocytes within the heart, although probably at a functionally insignificant level.


Assuntos
Linhagem da Célula , Traumatismos Cardíacos/patologia , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Miocárdio/citologia , Miócitos Cardíacos/citologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Envelhecimento/fisiologia , Animais , Diferenciação Celular , Fusão Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Coração/crescimento & desenvolvimento , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos , Modelos Biológicos , Miócitos Cardíacos/metabolismo , Regeneração/fisiologia , Tamoxifeno/farmacologia
5.
Circulation ; 138(25): 2931-2939, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29991486

RESUMO

BACKGROUND: The adult mammalian heart displays a cardiomyocyte turnover rate of ≈1%/y throughout postnatal life and after injuries such as myocardial infarction (MI), but the question of which cell types drive this low level of new cardiomyocyte formation remains contentious. Cardiac-resident stem cells marked by stem cell antigen-1 (Sca-1, gene name Ly6a) have been proposed as an important source of cardiomyocyte renewal. However, the in vivo contribution of endogenous Sca-1+ cells to the heart at baseline or after MI has not been investigated. METHODS: Here we generated Ly6a gene-targeted mice containing either a constitutive or an inducible Cre recombinase to perform genetic lineage tracing of Sca-1+ cells in vivo. RESULTS: We observed that the contribution of endogenous Sca-1+ cells to the cardiomyocyte population in the heart was <0.005% throughout all of cardiac development, with aging, or after MI. In contrast, Sca-1+ cells abundantly contributed to the cardiac vasculature in mice during physiological growth and in the post-MI heart during cardiac remodeling. Specifically, Sca-1 lineage-traced endothelial cells expanded postnatally in the mouse heart after birth and into adulthood. Moreover, pulse labeling of Sca-1+ cells with an inducible Ly6a-MerCreMer allele also revealed a preferential expansion of Sca-1 lineage-traced endothelial cells after MI injury in the mouse. CONCLUSIONS: Cardiac-resident Sca-1+ cells are not significant contributors to cardiomyocyte renewal in vivo. However, cardiac Sca-1+ cells represent a subset of vascular endothelial cells that expand postnatally with enhanced responsiveness to pathological stress in vivo.


Assuntos
Células-Tronco Adultas/fisiologia , Envelhecimento/fisiologia , Antígenos Ly/metabolismo , Endotélio Vascular/fisiologia , Coração/fisiologia , Proteínas de Membrana/metabolismo , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/fisiologia , Animais , Antígenos Ly/genética , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Vasos Coronários/cirurgia , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Modelos Animais , Desenvolvimento Muscular , Infarto do Miocárdio/genética
7.
J Biol Chem ; 291(19): 9920-8, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26966179

RESUMO

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by mutations in the gene encoding dystrophin. Loss of dystrophin protein compromises the stability of the sarcolemma membrane surrounding each muscle cell fiber, leading to membrane ruptures and leakiness that induces myofiber necrosis, a subsequent inflammatory response, and progressive tissue fibrosis with loss of functional capacity. Cathepsin S (Ctss) is a cysteine protease that is actively secreted in areas of tissue injury and ongoing inflammation, where it participates in extracellular matrix remodeling and healing. Here we show significant induction of Ctss expression and proteolytic activity following acute muscle injury or in muscle from mdx mice, a model of DMD. To examine the functional ramifications associated with greater Ctss expression, the Ctss gene was deleted in the mdx genetic background, resulting in protection from muscular dystrophy pathogenesis that included reduced myofiber turnover and histopathology, reduced fibrosis, and improved running capacity. Mechanistically, deletion of the Ctss gene in the mdx background significantly increased myofiber sarcolemmal membrane stability with greater expression and membrane localization of utrophin, integrins, and ß-dystroglycan, which anchor the membrane to the basal lamina and underlying cytoskeletal proteins. Consistent with these results, skeletal muscle-specific transgenic mice overexpressing Ctss showed increased myofiber necrosis, muscle histopathology, and a functional deficit reminiscent of muscular dystrophy. Hence, Ctss induction during muscular dystrophy is a pathologic event that partially underlies disease pathogenesis, and its inhibition might serve as a new therapeutic strategy in DMD.


Assuntos
Catepsinas/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Fibras Musculares Esqueléticas/enzimologia , Distrofia Muscular Animal/enzimologia , Distrofia Muscular de Duchenne/enzimologia , Animais , Citoesqueleto/enzimologia , Citoesqueleto/genética , Citoesqueleto/patologia , Camundongos , Camundongos Endogâmicos mdx , Camundongos Knockout , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Necrose , Proteólise , Sarcolema/enzimologia , Sarcolema/genética , Sarcolema/patologia
8.
Cell Rep ; 43(5): 114149, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38678560

RESUMO

Loss of muscle mass is a feature of chronic illness and aging. Here, we report that skeletal muscle-specific thrombospondin-1 transgenic mice (Thbs1 Tg) have profound muscle atrophy with age-dependent decreases in exercise capacity and premature lethality. Mechanistically, Thbs1 activates transforming growth factor ß (TGFß)-Smad2/3 signaling, which also induces activating transcription factor 4 (ATF4) expression that together modulates the autophagy-lysosomal pathway (ALP) and ubiquitin-proteasome system (UPS) to facilitate muscle atrophy. Indeed, myofiber-specific inhibition of TGFß-receptor signaling represses the induction of ATF4, normalizes ALP and UPS, and partially restores muscle mass in Thbs1 Tg mice. Similarly, myofiber-specific deletion of Smad2 and Smad3 or the Atf4 gene antagonizes Thbs1-induced muscle atrophy. More importantly, Thbs1-/- mice show significantly reduced levels of denervation- and caloric restriction-mediated muscle atrophy, along with blunted TGFß-Smad3-ATF4 signaling. Thus, Thbs1-mediated TGFß-Smad3-ATF4 signaling in skeletal muscle regulates tissue rarefaction, suggesting a target for atrophy-based muscle diseases and sarcopenia with aging.


Assuntos
Fator 4 Ativador da Transcrição , Músculo Esquelético , Atrofia Muscular , Transdução de Sinais , Proteína Smad2 , Proteína Smad3 , Trombospondina 1 , Fator de Crescimento Transformador beta , Animais , Masculino , Camundongos , Fator 4 Ativador da Transcrição/metabolismo , Autofagia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Trombospondina 1/metabolismo , Trombospondina 1/genética , Fator de Crescimento Transformador beta/metabolismo
9.
Development ; 137(23): 3953-63, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20980403

RESUMO

Kruppel-like transcription factors (Klfs) are essential for the induction and maintenance of pluripotency of embryonic stem cells (ESCs), yet little is known about their roles in establishing the three lineages of the pre-implantation embryo. Here, we show that Klf5 is required for the formation of the trophectoderm (TE) and the inner cell mass (ICM), and for repressing primitive endoderm (PE) development. Although cell polarity appeared normal, Klf5 mutant embryos arrested at the blastocyst stage and failed to hatch due to defective TE development. Klf5 acted cell-autonomously in the TE, downstream of Fgf4 and upstream of Cdx2, Eomes and Krt8. In the ICM, loss of Klf5 resulted in reduced expression of pluripotency markers Oct4 and Nanog, but led to increased Sox17 expression in the PE, suggesting that Klf5 suppresses the PE lineage. Consistent with this, overexpression of Klf5 in transgenic embryos was sufficient to suppress the Sox17(+) PE lineage in the ICM. Klf5 overexpression led to a dose-dependent decrease in Sox17 promoter activity in reporter assays in cultured cells. Moreover, in chimeric embryos, Klf5(-/-) cells preferentially contributed to the Sox17(+) PE lineage and Cdx2 expression was not rescued in Klf5(-/-) outer cells. Finally, outgrowths from Klf5(-/-) embryos failed to form an ICM/pluripotent colony, had very few Oct4(+) or Cdx2(+) cells, but showed an increase in the percentage of Sox17(+) PE cells. These findings demonstrate that Klf5 is a dynamic regulator of all three lineages in the pre-implantation embryo by promoting the TE and epiblast lineages while suppressing the PE lineage.


Assuntos
Blastocisto/citologia , Blastocisto/metabolismo , Linhagem da Célula , Fatores de Transcrição Kruppel-Like/metabolismo , Animais , Biomarcadores/metabolismo , Fator de Transcrição CDX2 , Contagem de Células , Morte Celular , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Cruzamentos Genéticos , Ectoderma/citologia , Ectoderma/metabolismo , Endoderma/citologia , Endoderma/metabolismo , Feminino , Fator 4 de Crescimento de Fibroblastos/genética , Fator 4 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
10.
Cell Rep ; 42(11): 113465, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37976157

RESUMO

Mitochondria use the electron transport chain to generate high-energy phosphate from oxidative phosphorylation, a process also regulated by the mitochondrial Ca2+ uniporter (MCU) and Ca2+ levels. Here, we show that MCUb, an inhibitor of MCU-mediated Ca2+ influx, is induced by caloric restriction, where it increases mitochondrial fatty acid utilization. To mimic the fasted state with reduced mitochondrial Ca2+ influx, we generated genetically altered mice with skeletal muscle-specific MCUb expression that showed greater fatty acid usage, less fat accumulation, and lower body weight. In contrast, mice lacking Mcub in skeletal muscle showed increased pyruvate dehydrogenase activity, increased muscle malonyl coenzyme A (CoA), reduced fatty acid utilization, glucose intolerance, and increased adiposity. Mechanistically, pyruvate dehydrogenase kinase 4 (PDK4) overexpression in muscle of Mcub-deleted mice abolished altered substrate preference. Thus, MCUb is an inducible control point in regulating skeletal muscle mitochondrial Ca2+ levels and substrate utilization that impacts total metabolic balance.


Assuntos
Cálcio , Mitocôndrias , Animais , Camundongos , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Ácidos Graxos/metabolismo , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo
11.
Front Physiol ; 14: 1054169, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36733907

RESUMO

Introduction: The ribosomal protein L3-like (RPL3L) is a heart and skeletal muscle-specific ribosomal protein and paralogue of the more ubiquitously expressed RPL3 protein. Mutations in the human RPL3L gene are linked to childhood cardiomyopathy and age-related atrial fibrillation, yet the function of RPL3L in the mammalian heart remains unknown. Methods and Results: Here, we observed that mouse cardiac ventricles express RPL3 at birth, where it is gradually replaced by RPL3L in adulthood but re-expressed with induction of hypertrophy in adults. Rpl3l gene-deleted mice were generated to examine the role of this gene in the heart, although Rpl3l -/- mice showed no overt changes in cardiac structure or function at baseline or after pressure overload hypertrophy, likely because RPL3 expression was upregulated and maintained in adulthood. mRNA expression analysis and ribosome profiling failed to show differences between the hearts of Rpl3l null and wild type mice in adulthood. Moreover, ribosomes lacking RPL3L showed no differences in localization within cardiomyocytes compared to wild type controls, nor was there an alteration in cardiac tissue ultrastructure or mitochondrial function in adult Rpl3l -/- mice. Similarly, overexpression of either RPL3 or RPL3L with adeno-associated virus -9 in the hearts of mice did not cause discernable pathology. However, by 18 months of age Rpl3l -/- null mice had significantly smaller hearts compared to wild type littermates. Conclusion: Thus, deletion of Rpl3l forces maintenance of RPL3 expression within the heart that appears to fully compensate for the loss of RPL3L, although older Rpl3l -/- mice showed a mild but significant reduction in heart weight.

12.
PLoS One ; 16(7): e0254667, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34260645

RESUMO

The world is currently in a pandemic of COVID-19 (Coronavirus disease-2019) caused by a novel positive-sense, single-stranded RNA ß-coronavirus referred to as SARS-CoV-2. Here we investigated rates of SARS-CoV-2 infection in the greater Cincinnati, Ohio, USA metropolitan area from August 13 to December 8, 2020, just prior to initiation of the national vaccination program. Examination of 9,550 adult blood donor volunteers for serum IgG antibody positivity against the SARS-CoV-2 Spike protein showed an overall prevalence of 8.40%, measured as 7.56% in the first 58 days and 9.24% in the last 58 days, and 12.86% in December 2020, which we extrapolated to ~20% as of March, 2021. Males and females showed similar rates of past infection, and rates among Hispanic or Latinos, African Americans and Whites were also investigated. Donors under 30 years of age had the highest rates of past infection, while those over 60 had the lowest. Geographic analysis showed higher rates of infectivity on the West side of Cincinnati compared with the East side (split by I-75) and the lowest rates in the adjoining region of Kentucky (across the Ohio river). These results in regional seroprevalence will help inform efforts to best achieve herd immunity in conjunction with the national vaccination campaign.


Assuntos
Anticorpos Antivirais/sangue , Doadores de Sangue/estatística & dados numéricos , COVID-19/epidemiologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , COVID-19/imunologia , Feminino , Humanos , Imunoglobulina G/sangue , Masculino , Pessoa de Meia-Idade , Ohio/etnologia , Pandemias , Estudos Soroepidemiológicos , Adulto Jovem
13.
Cancer Res ; 64(10): 3525-32, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15150107

RESUMO

Approximately 70% of human breast cancers are estrogen receptor alpha (ERalpha)-positive, but the origins of ERalpha-positive and -negative tumors remain unclear. Hormonal regulation of mammary gland development in mice is similar to that in humans; however, most mouse models produce only ERalpha-negative tumors. In addition, these mouse tumors metastasize at a low rate relative to human breast tumors. We report here that somatic mutations of p53 in mouse mammary epithelial cells using the Cre/loxP system leads to ERalpha-positive and -negative tumors. p53 inactivation under a constitutive active WAPCre(c) in prepubertal/pubertal mice, but not under MMTVCre in adult mice, leads to the development of ERalpha-positive tumors, suggesting that target cells or developmental stages can determine ERalpha status in mammary tumors. Importantly, these tumors have a high rate of metastasis. An inverse relationship between the number of targeted cells and median tumor latency was also observed. Median tumor latency reaches a plateau when targeted cell numbers exceed 20%, implying the existence of saturation kinetics for breast carcinogenesis. Genetic alterations commonly observed in human breast cancer including c-myc amplification and Her2/Neu/erbB2 activation were seen in these mouse tumors. Thus, this tumor system reproduces many important features of human breast cancer and provides tools for the study of the origins of ERalpha-positive and -negative breast tumors in mice.


Assuntos
Genes p53/genética , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Mutação , Receptores de Estrogênio/biossíntese , Alelos , Animais , Modelos Animais de Doenças , Receptor alfa de Estrogênio , Feminino , Inativação Gênica , Integrases/genética , Neoplasias Mamárias Experimentais/metabolismo , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Metástase Neoplásica , Proteínas Virais/genética
14.
PLoS One ; 11(10): e0164897, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27764156

RESUMO

Nemo-like kinase (NLK) is an evolutionary conserved serine/threonine protein kinase implicated in development, proliferation and apoptosis regulation. Here we identified NLK as a gene product induced in the hearts of mice subjected to pressure overload or myocardial infarction injury, suggesting a potential regulatory role with pathological stimulation to this organ. To examine the potential functional consequences of increased NLK levels, cardiac-specific transgenic mice with inducible expression of this gene product were generated, as well as cardiac-specific Nlk gene-deleted mice. NLK transgenic mice demonstrated baseline cardiac hypertrophy, dilation, interstitial fibrosis, apoptosis and progression towards heart failure in response to two surgery-induced cardiac disease models. In contrast, cardiac-specific deletion of Nlk from the heart, achieved by crossing a Nlk-loxP allele containing mouse with either a mouse containing a ß-myosin heavy chain promoter driven Cre transgene or a tamoxifen inducible α-myosin heavy chain promoter containing transgene driving a MerCreMer cDNA, protected the mice from cardiac dysfunction following pathological stimuli. Mechanistically, NLK interacted with multiple proteins including the transcription factor Stat1, which was significantly increased in the hearts of NLK transgenic mice. These results indicate that NLK is a pathological effector in the heart.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/genética , Miocárdio/metabolismo , Animais , Cardiomiopatias/etiologia , Células Cultivadas , Ecocardiografia , Feminino , Células HEK293 , Coração/diagnóstico por imagem , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/deficiência , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Serina-Treonina Quinases , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
15.
Nat Commun ; 7: 12260, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27447449

RESUMO

Cardiac fibroblasts convert to myofibroblasts with injury to mediate healing after acute myocardial infarction (MI) and to mediate long-standing fibrosis with chronic disease. Myofibroblasts remain a poorly defined cell type in terms of their origins and functional effects in vivo. Here we generate Postn (periostin) gene-targeted mice containing a tamoxifen-inducible Cre for cellular lineage-tracing analysis. This Postn allele identifies essentially all myofibroblasts within the heart and multiple other tissues. Lineage tracing with four additional Cre-expressing mouse lines shows that periostin-expressing myofibroblasts in the heart derive from tissue-resident fibroblasts of the Tcf21 lineage, but not endothelial, immune/myeloid or smooth muscle cells. Deletion of periostin(+) myofibroblasts reduces collagen production and scar formation after MI. Periostin-traced myofibroblasts also revert back to a less-activated state upon injury resolution. Our results define the myofibroblast as a periostin-expressing cell type necessary for adaptive healing and fibrosis in the heart, which arises from Tcf21(+) tissue-resident fibroblasts.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Moléculas de Adesão Celular/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miofibroblastos/metabolismo , Animais , Biomarcadores/metabolismo , Moléculas de Adesão Celular/genética , Feminino , Integrases , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Tamoxifeno
16.
Nat Commun ; 6: 10084, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26670661

RESUMO

The differentiation of fibroblasts into myofibroblasts mediates tissue wound healing and fibrotic remodelling, although the molecular programme underlying this process remains poorly understood. Here we perform a genome-wide screen for genes that control myofibroblast transformation, and identify the RNA-binding protein muscleblind-like1 (MBNL1). MBNL1 overexpression promotes transformation of fibroblasts into myofibroblasts, whereas loss of Mbnl1 abrogates transformation and impairs the fibrotic phase of wound healing in mouse models of myocardial infarction and dermal injury. Mechanistically, MBNL1 directly binds to and regulates a network of differentiation-specific and cytoskeletal/matrix-assembly transcripts to promote myofibroblast differentiation. One of these transcripts is the nodal transcriptional regulator serum response factor (SRF), whereas another is calcineurin Aß. CRISPR-Cas9-mediated gene-editing of the MBNL1-binding site within the Srf 3'UTR impairs myofibroblast differentiation, whereas in vivo deletion of Srf in fibroblasts impairs wound healing and fibrosis. These data establish a new RNA-dependent paradigm for myofibroblast formation through MBNL1.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Fibrose/genética , Fibrose/metabolismo , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibrose/fisiopatologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , RNA/genética , RNA/metabolismo , Proteínas de Ligação a RNA/genética , Cicatrização
17.
Dev Cell ; 17(1): 62-74, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19619492

RESUMO

The ventral pancreas, biliary system, and liver arise from the posterior ventral foregut, but the cell-intrinsic pathway by which these organ lineages are separated is not known. Here we show that the extrahepatobiliary system shares a common origin with the ventral pancreas and not the liver, as previously thought. These pancreatobiliary progenitor cells coexpress the transcription factors PDX1 and SOX17 at E8.5 and their segregation into a PDX1+ ventral pancreas and a SOX17+ biliary primordium is Sox17-dependent. Deletion of Sox17 at E8.5 results in the loss of biliary structures and ectopic pancreatic tissue in the liver bud and common duct, while Sox17 overexpression suppresses pancreas development and promotes ectopic biliary-like tissue throughout the PDX1+ domain. Restricting SOX17+ biliary progenitor cells to the ventral region of the gut requires the notch effector Hes1. Our results highlight the role of Sox17 and Hes1 in patterning and morphogenetic segregation of ventral foregut lineages.


Assuntos
Sistema Biliar/embriologia , Fígado/embriologia , Organogênese/fisiologia , Pâncreas/embriologia , Células-Tronco/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Sistema Biliar/citologia , Biomarcadores/metabolismo , Linhagem da Célula , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Proteínas HMGB , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fígado/citologia , Camundongos , Camundongos Knockout , Morfogênese/fisiologia , Pâncreas/citologia , Fatores de Transcrição SOXF , Células-Tronco/citologia , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição HES-1 , Transgenes
18.
Dev Dyn ; 236(7): 1997-2003, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17576135

RESUMO

Little is known about how the endoderm germ layer is patterned along the anterior-posterior (A-P) axis before the formation of a gut tube (embryonic day [e] 7.5-8.5 in mouse), largely due to a paucity of molecular markers of endoderm. In particular, there are few genes that mark posterior domains of endoderm that give rise to the midgut and hindgut. We have identified 8 molecular markers that are expressed in discrete domains of the gastrula stage endoderm (e7.5), suggesting that a significant level of pattern exists in the endoderm before the formation of a gut tube. Three genes Tmprss2, NM_029639, and Dsp are expressed in a presumptive midgut domain overlying the node, a domain for which molecular markers have not previously been identified. Two genes, Klf5 and Epha2 are expressed in posterior endoderm associated with the primitive streak. Expression of these five genes persists in the midgut and/or hindgut at e8.5, 9.5 and 10.5, suggesting that these genes are markers of these domains throughout these stages of development. We have identified three genes Slc39a8, Amot, and Dp1l1, which are expressed in the visceral endoderm at e7.5. Starting at e9.5, Dp1l1 is expressed de novo in the liver, midgut, and hindgut. Our findings suggest that presumptive midgut and hindgut domains are being established at the molecular level by the end of gastrulation, earlier than previously thought, and emphasize the importance of endoderm patterning before the formation of the fetal gut.


Assuntos
Biomarcadores , Endoderma/metabolismo , Feto/metabolismo , Gástrula/metabolismo , Animais , Feminino , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Gravidez
19.
Mol Cell Biol ; 27(22): 7802-15, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17875931

RESUMO

The canonical Wnt pathway is necessary for gut epithelial cell proliferation, and aberrant activation of this pathway causes intestinal neoplasia. We report a novel mechanism by which the Sox family of transcription factors regulate the canonical Wnt signaling pathway. We found that some Sox proteins antagonize while others enhance beta-catenin/T-cell factor (TCF) activity. Sox17, which is expressed in the normal gut epithelium but exhibits reduced expression in intestinal neoplasia, is antagonistic to Wnt signaling. When overexpressed in SW480 colon carcinoma cells, Sox17 represses beta-catenin/TCF activity in a dose-dependent manner and inhibits proliferation. Sox17 and Sox4 are expressed in mutually exclusive domains in normal and neoplastic gut tissues, and gain- and loss-of-function studies demonstrate that Sox4 enhances beta-catenin/TCF activity and the proliferation of SW480 cells. In addition to binding beta-catenin, both Sox17 and Sox4 physically interact with TCF/lymphoid enhancer factor (LEF) family members via their respective high-mobility-group box domains. Results from gain- and loss-of-function experiments suggest that the interaction of Sox proteins with beta-catenin and TCF/LEF proteins regulates the stability of beta-catenin and TCF/LEF. In particular, Sox17 promotes the degradation of both beta-catenin and TCF proteins via a noncanonical, glycogen synthase kinase 3beta-independent mechanism that can be blocked by proteasome inhibitors. In contrast, Sox4 may function to stabilize beta-catenin protein. These findings indicate that Sox proteins can act as both antagonists and agonists of beta-catenin/TCF activity, and this mechanism may regulate Wnt signaling responses in many developmental and disease contexts.


Assuntos
Carcinoma/metabolismo , Proliferação de Células , Neoplasias do Colo/metabolismo , Proteínas HMGB/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Fatores de Transcrição TCF/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo , Animais , Linhagem Celular , Embrião não Mamífero/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Proteínas HMGB/genética , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Estrutura Terciária de Proteína , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição SOXC , Fatores de Transcrição SOXF , Transdução de Sinais/fisiologia , Fatores de Transcrição TCF/genética , Transativadores/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição , Fatores de Transcrição/genética , Transcrição Gênica , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas de Xenopus , Xenopus laevis , beta Catenina/genética
20.
Science ; 314(5804): 1467-70, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17138902

RESUMO

Women with mutations in the breast cancer susceptibility gene BRCA1 are predisposed to breast and ovarian cancers. Why the BRCA1 protein suppresses tumor development specifically in ovarian hormone-sensitive tissues remains unclear. We demonstrate that mammary glands of nulliparous Brca1/p53-deficient mice accumulate lateral branches and undergo extensive alveologenesis, a phenotype that occurs only during pregnancy in wild-type mice. Progesterone receptors, but not estrogen receptors, are overexpressed in the mutant mammary epithelial cells because of a defect in their degradation by the proteasome pathway. Treatment of Brca1/p53-deficient mice with the progesterone antagonist mifepristone (RU 486) prevented mammary tumorigenesis. These findings reveal a tissue-specific function for the BRCA1 protein and raise the possibility that antiprogesterone treatment may be useful for breast cancer prevention in individuals with BRCA1 mutations.


Assuntos
Genes BRCA1 , Antagonistas de Hormônios/farmacologia , Neoplasias Mamárias Animais/prevenção & controle , Mifepristona/farmacologia , Progesterona/antagonistas & inibidores , Receptores de Progesterona/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Estradiol/farmacologia , Ciclo Estral , Feminino , Genes p53 , Antagonistas de Hormônios/uso terapêutico , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/genética , Camundongos , Mifepristona/uso terapêutico , Mutação , Fosforilação , Progesterona/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Interferente Pequeno , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/genética , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA