Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomater Sci ; 10(21): 6217-6229, 2022 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-36102692

RESUMO

Adjuvants play a critical role in enhancing vaccine efficacy; however, there is a need to develop new immunomodulatory compounds to address emerging pathogens and to expand the use of immunotherapies. Multidomain peptides (MDPs) are materials composed of canonical amino acids that form injectable supramolecular hydrogels under physiological salt and pH conditions. MDP hydrogels are rapidly infiltrated by immune cells in vivo and have previously been shown to influence cytokine production. Therefore, we hypothesized that these immunostimulatory characteristics would allow MDPs to function as vaccine adjuvants. Herein, we demonstrate that loading antigen into MDP hydrogels does not interfere with their rheological properties and that positively charged MDPs can act as antigen depots, as demonstrated by their ability to release ovalbumin (OVA) over a period of 7-9 days in vivo. Mice vaccinated with MDP-adjuvanted antigen generated significantly higher IgG titers than mice treated with the unadjuvanted control, suggesting that these hydrogels potentiate humoral immunity. Interestingly, MDP hydrogels did not elicit a robust cellular immune response, as indicated by the lower production of IgG2c and smaller populations of tetramer-positive CD8+ T splenocytes compared to mice vaccinated alum-adjuvanted OVA. Together, the data suggest that MDP hydrogel adjuvants strongly bias the immune response towards humoral immunity while evoking a very limited cellular immune response. As a result, MDPs may have the potential to serve as adjuvants for applications that benefit exclusively from humoral immunity.


Assuntos
Hidrogéis , Imunidade Humoral , Camundongos , Animais , Ovalbumina , Adjuvantes Imunológicos/química , Antígenos , Peptídeos , Adjuvantes Farmacêuticos , Imunoglobulina G , Aminoácidos , Citocinas
2.
Cells ; 12(1)2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36611823

RESUMO

Uncultured, unmodified, autologous, adipose-derived regenerative cells (UA-ADRCs) are a safe and effective treatment option for various musculoskeletal pathologies. However, it is unknown whether the composition of the final cell suspension systematically varies with the subject's individual age, sex, body mass index and ethnicity. UA-ADRCs were isolated from lipoaspirate from n = 232 subjects undergoing elective lipoplasty using the Transpose RT system (InGeneron, Inc.; Houston, TX, USA). The UA-ADRCs were assessed for the number of nucleated cells, cell viability and the number of viable nucleated cells per gram of adipose tissue harvested. Cells from n = 37 subjects were further characterized using four-channel flow cytometry. The present study shows, for the first time, that key characteristics of UA-ADRCs can be independent of the subject's age, sex, BMI and ethnicity. This result has important implications for the general applicability of UA-ADRCs in regeneration of musculoskeletal tissue. Future studies must determine whether the independence of key characteristics of UA-ADRCs of the subject's individual age, sex, BMI and ethnicity only applies to the system used in the present study, or also to others of the more than 25 different experimental methods and commercially available systems used to isolate UA-ADRCs from lipoaspirate that have been described in the literature.


Assuntos
Etnicidade , Lipectomia , Humanos , Índice de Massa Corporal , Sistemas Automatizados de Assistência Junto ao Leito , Tecido Adiposo
3.
Genes (Basel) ; 13(1)2021 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-35052435

RESUMO

The molecular and cellular processes leading to aortic aneurysm development in Marfan syndrome (MFS) remain poorly understood. In this study, we examined the changes of aortic cell populations and gene expression in MFS by performing single-cell RNA sequencing (scRNA seq) on ascending aortic aneurysm tissues from patients with MFS (n = 3) and age-matched non-aneurysmal control tissues from cardiac donors and recipients (n = 4). The expression of key molecules was confirmed by immunostaining. We detected diverse populations of smooth muscle cells (SMCs), fibroblasts, and endothelial cells (ECs) in the aortic wall. Aortic tissues from MFS showed alterations of cell populations with increased de-differentiated proliferative SMCs compared to controls. Furthermore, there was a downregulation of MYOCD and MYH11 in SMCs, and an upregulation of COL1A1/2 in fibroblasts in MFS samples compared to controls. We also examined TGF-ß signaling, an important pathway in aortic homeostasis. We found that TGFB1 was significantly upregulated in two fibroblast clusters in MFS tissues. However, TGF-ß receptor genes (predominantly TGFBR2) and SMAD genes were downregulated in SMCs, fibroblasts, and ECs in MFS, indicating impairment in TGF-ß signaling. In conclusion, despite upregulation of TGFB1, the rest of the canonical TGF-ß pathway and mature SMCs were consistently downregulated in MFS, indicating a potential compromise of TGF-ß signaling and lack of stimulus for SMC differentiation.


Assuntos
Aneurisma da Aorta Torácica/diagnóstico , Síndrome de Marfan/complicações , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adulto , Aneurisma da Aorta Torácica/etiologia , Aneurisma da Aorta Torácica/metabolismo , Estudos de Casos e Controles , Diferenciação Celular , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais , Análise de Célula Única , Fator de Crescimento Transformador beta/genética , Adulto Jovem
4.
Circulation ; 141(1): 42-66, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31887080

RESUMO

BACKGROUND: Sporadic aortic aneurysm and dissection (AAD), caused by progressive aortic smooth muscle cell (SMC) loss and extracellular matrix degradation, is a highly lethal condition. Identifying mechanisms that drive aortic degeneration is a crucial step in developing an effective pharmacologic treatment to prevent disease progression. Recent evidence has indicated that cytosolic DNA and abnormal activation of the cytosolic DNA sensing adaptor STING (stimulator of interferon genes) play a critical role in vascular inflammation and destruction. Here, we examined the involvement of this mechanism in aortic degeneration and sporadic AAD formation. METHODS: The presence of cytosolic DNA in aortic cells and activation of the STING pathway were examined in aortic tissues from patients with sporadic ascending thoracic AAD. The role of STING in AAD development was evaluated in Sting-deficient (Stinggt/gt) mice in a sporadic AAD model induced by challenging mice with a combination of a high-fat diet and angiotensin II. We also examined the direct effects of STING on SMC death and macrophage activation in vitro. RESULTS: In human sporadic AAD tissues, we observed the presence of cytosolic DNA in SMCs and macrophages and significant activation of the STING pathway. In the sporadic AAD model, Stinggt/gt mice showed significant reductions in challenge-induced aortic enlargement, dissection, and rupture in both the thoracic and abdominal aortic regions. Single-cell transcriptome analysis revealed that aortic challenge in wild-type mice induced the DNA damage response, the inflammatory response, dedifferentiation and cell death in SMCs, and matrix metalloproteinase expression in macrophages. These changes were attenuated in challenged Stinggt/gt mice. Mechanistically, nuclear and mitochondrial DNA damage in SMCs and the subsequent leak of DNA to the cytosol activated STING signaling, which induced cell death through apoptosis and necroptosis. In addition, DNA from damaged SMCs was engulfed by macrophages in which it activated STING and its target interferon regulatory factor 3, which directly induced matrix metalloproteinase-9 expression. We also found that pharmacologically inhibiting STING activation partially prevented AAD development. CONCLUSIONS: Our findings indicate that the presence of cytosolic DNA and subsequent activation of cytosolic DNA sensing adaptor STING signaling represent a key mechanism in aortic degeneration and that targeting STING may prevent sporadic AAD development.


Assuntos
Dissecção Aórtica/metabolismo , Ruptura Aórtica/metabolismo , Citosol/metabolismo , DNA/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Dissecção Aórtica/genética , Dissecção Aórtica/patologia , Animais , Ruptura Aórtica/genética , Ruptura Aórtica/patologia , Citosol/patologia , DNA/genética , Feminino , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout
5.
J Surg Res ; 245: 1-12, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31394402

RESUMO

BACKGROUND: The process of aortic injury, repair, and remodeling during aortic aneurysm and dissection is poorly understood. We examined the activation of bone marrow (BM)-derived and resident aortic cells in response to aortic injury in a mouse model of sporadic aortic aneurysm and dissection. MATERIALS AND METHODS: Wild-type C57BL/6 mice were transplanted with green fluorescent protein (GFP)+ BM cells. For 4 wk, these mice were either unchallenged with chow diet and saline infusion or challenged with high-fat diet and angiotensin II infusion. We then examined the aortic recruitment of GFP+ BM-derived cells, growth factor production, and the differentiation potential of GFP+ BM-derived and GFP- resident aortic cells. RESULTS: Aortic challenge induced recruitment of GFP+ BM cells and activation of GFP- resident aortic cells, both of which produced growth factors. Although BM cells and resident aortic cells equally contributed to the fibroblast populations, we did not detect the differentiation of BM cells into smooth muscle cells. Interestingly, aortic macrophages were both of BM-derived (45%) and of non-BM-derived (55%) origin. We also observed a significant increase in stem cell antigen-1 (Sca-1)+ stem/progenitor cells and neural/glial antigen 2 (NG2+) cells in the aortic wall of challenged mice. Although some of the Sca-1+ cells and NG2+ cells were BM derived, most of these cells were resident aortic cells. Sca-1+ cells produced growth factors and differentiated into fibroblasts and NG2+ cells. CONCLUSIONS: BM-derived and resident aortic cells are activated in response to aortic injury and contribute to aortic inflammation, repair, and remodeling by producing growth factors and differentiating into fibroblasts and inflammatory cells.


Assuntos
Aorta/patologia , Aneurisma Aórtico/patologia , Dissecção Aórtica/patologia , Dissecção Aórtica/etiologia , Dissecção Aórtica/imunologia , Animais , Aorta/citologia , Aorta/imunologia , Aneurisma Aórtico/complicações , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Fibroblastos/imunologia , Fibroblastos/metabolismo , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/metabolismo
6.
Biomaterials ; 231: 119667, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31855625

RESUMO

Multidomain Peptide (MDP) hydrogels are nanofibrous materials with many potential biomedical applications. The peptide sequence design of these materials offers high versatility and allows for the incorporation of various chemical functionalities into the nanofibrous scaffold. It is known that host response to biomaterials is strongly affected by factors such as size, shape, stiffness, and chemistry. However, there is a lack of fundamental understanding of the host response to different MDP hydrogels. In particular, it is unknown what effect the chemical functionality displayed on the nanofiber has on biological activity. Here we evaluated the early inflammatory host response to four MDP hydrogels displaying amines, guanidinium ions, and carboxylates in a subcutaneous injection model. While all the studied peptide materials possess similar nanostructure and physical properties, they trigger markedly different inflammatory responses. These were characterized by immunophenotyping of the cellular infiltrate using multi-color flow cytometry. The negatively-charged peptides elicit minimal inflammation characterized by tissue-resident macrophage infiltration, fast remodeling, and no collagen deposition or blood vessel formation within the implants. In contrast, the positively-charged peptides are highly infiltrated by immune cells, are remodeled at a slower rate, promote angiogenesis, and result in a high degree of collagen deposition. The presence of dynamic cell phenotypes characterizes the inflammation caused by the lysine-based peptide, including inflammatory monocytes, macrophages, and lymphoid cells, which is seen to be resolving over time. The arginine-based hydrogel shows higher inflammatory response with a persistent and significant infiltration of polymorphonuclear myeloid-derived cells, even ten days after implantation. This understanding of the immune response to peptide biomaterials improves our ability to design effective materials and to tailor their use for specific biomedical applications.


Assuntos
Hidrogéis , Nanofibras , Materiais Biocompatíveis , Imunidade , Peptídeos
7.
JAMA Surg ; 153(9): e181804, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30046809

RESUMO

Importance: Fluoroquinolones are among the most commonly prescribed antibiotics. Recent clinical studies indicated an association between fluoroquinolone use and increased risk of aortic aneurysm and dissection (AAD). This alarming association has raised concern, especially in patients with AAD with risk of rupture and in individuals at risk for developing AAD. Objective: To examine the effect of ciprofloxacin on AAD development in mice. Design, Setting, and Participants: In a mouse model of moderate, sporadic AAD, 4-week-old male and female C57BL/6J mice were challenged with a high-fat diet and low-dose angiotensin infusion (1000 ng/min/kg). Control unchallenged mice were fed a normal diet and infused with saline. After randomization, challenged and unchallenged mice received ciprofloxacin (100 mg/kg/d) or vehicle through daily gavage during angiotensin or saline infusion. Aortic aneurysm and dissection development and aortic destruction were compared between mice. The direct effects of ciprofloxacin on aortic smooth muscle cells were examined in cultured cells. Results: No notable aortic destruction was observed in unchallenged mice that received ciprofloxacin alone. Aortic challenge induced moderate aortic destruction with development of AAD in 17 of 38 mice (45%) and severe AAD in 9 (24%) but no rupture or death. However, challenged mice that received ciprofloxacin had severe aortic destruction and a significantly increased incidence of AAD (38 of 48 [79%]; P = .001; χ2 = 10.9), severe AAD (32 of 48 [67%]; P < .001; χ2 = 15.7), and rupture and premature death (7 of 48 [15%]; P = .01; χ2 = 6.0). The increased AAD incidence was observed in different aortic segments and was similar between male and female mice. Compared with aortic tissues from challenged control mice, those from challenged mice that received ciprofloxacin showed decreased expression of lysyl oxidase, an enzyme that is critical in the assembly and stabilization of elastic fibers and collagen. These aortas also showed increased matrix metalloproteinase levels and activity, elastic fiber fragmentation, and aortic cell injury. In cultured smooth muscle cells, ciprofloxacin treatment significantly reduced lysyl oxidase expression and activity, increased matrix metalloproteinase expression and activity, suppressed cell proliferation, and induced cell death. Furthermore, ciprofloxacin-a DNA topoisomerase inhibitor-caused nuclear and mitochondrial DNA damage and the release of DNA into the cytosol, subsequently inducing mitochondrial dysfunction, reactive oxygen species production, and activation of the cytosolic DNA sensor STING, which we further showed was involved in the suppression of lysyl oxidase expression and induction of matrix metalloproteinase expression. Conclusions and Relevance: Ciprofloxacin increases susceptibility to aortic dissection and rupture in a mouse model of moderate, sporadic AAD. Ciprofloxacin should be used with caution in patients with aortic dilatation, as well as in those at high risk for AAD.


Assuntos
Aorta Torácica , Aneurisma da Aorta Torácica , Dissecção Aórtica , Ciprofloxacina , Animais , Feminino , Masculino , Camundongos , Antibacterianos/efeitos adversos , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/patologia , Aneurisma da Aorta Torácica/induzido quimicamente , Aneurisma da Aorta Torácica/diagnóstico , Dissecção Aórtica/induzido quimicamente , Dissecção Aórtica/diagnóstico por imagem , Células Cultivadas , Ciprofloxacina/efeitos adversos , Modelos Animais de Doenças , Seguimentos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Estudos Retrospectivos
8.
Sci Rep ; 6: 31457, 2016 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-27538477

RESUMO

Mesp1 directs multipotential cardiovascular cell fates, even though it's transiently induced prior to the appearance of the cardiac progenitor program. Tracing Mesp1-expressing cells and their progeny allows isolation and characterization of the earliest cardiovascular progenitor cells. Studying the biology of Mesp1-CPCs in cell culture and ischemic disease models is an important initial step toward using them for heart disease treatment. Because of Mesp1's transitory nature, Mesp1-CPC lineages were traced by following EYFP expression in murine Mesp1(Cre/+); Rosa26(EYFP/+) ES cells. We captured EYFP+ cells that strongly expressed cardiac mesoderm markers and cardiac transcription factors, but not pluripotent or nascent mesoderm markers. BMP2/4 treatment led to the expansion of EYFP+ cells, while Wnt3a and Activin were marginally effective. BMP2/4 exposure readily led EYFP+ cells to endothelial and smooth muscle cells, but inhibition of the canonical Wnt signaling was required to enter the cardiomyocyte fate. Injected mouse pre-contractile Mesp1-EYFP+ CPCs improved the survivability of injured mice and restored the functional performance of infarcted hearts for at least 3 months. Mesp1-EYFP+ cells are bona fide CPCs and they integrated well in infarcted hearts and emerged de novo into terminally differentiated cardiac myocytes, smooth muscle and vascular endothelial cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Infarto do Miocárdio/terapia , Transplante de Células-Tronco , Células-Tronco/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Coração/diagnóstico por imagem , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos SCID , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Transcriptoma , Proteína Wnt3A/metabolismo
9.
Proc Natl Acad Sci U S A ; 113(34): 9551-6, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27512039

RESUMO

Understanding the mechanisms of early cardiac fate determination may lead to better approaches in promoting heart regeneration. We used a mesoderm posterior 1 (Mesp1)-Cre/Rosa26-EYFP reporter system to identify microRNAs (miRNAs) enriched in early cardiac progenitor cells. Most of these miRNA genes bear MESP1-binding sites and active histone signatures. In a calcium transient-based screening assay, we identified miRNAs that may promote the cardiomyocyte program. An X-chromosome miRNA cluster, miR-322/-503, is the most enriched in the Mesp1 lineage and is the most potent in the screening assay. It is specifically expressed in the looping heart. Ectopic miR-322/-503 mimicking the endogenous temporal patterns specifically drives a cardiomyocyte program while inhibiting neural lineages, likely by targeting the RNA-binding protein CUG-binding protein Elav-like family member 1 (Celf1). Thus, early miRNAs in lineage-committed cells may play powerful roles in cell-fate determination by cross-suppressing other lineages. miRNAs identified in this study, especially miR-322/-503, are potent regulators of early cardiac fate.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , Células-Tronco Embrionárias Murinas/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas CELF1/genética , Proteínas CELF1/metabolismo , Diferenciação Celular , Linhagem da Célula/genética , Embrião de Mamíferos , Perfilação da Expressão Gênica , Genes Reporter , Integrases/genética , Integrases/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mesoderma/citologia , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , MicroRNAs/metabolismo , Morfogênese/genética , Células-Tronco Embrionárias Murinas/citologia , Miócitos Cardíacos/citologia , Cultura Primária de Células , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Transdução de Sinais
10.
Biomaterials ; 98: 113-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27182813

RESUMO

For a proangiogenic therapy to be successful, it must promote the development of mature vasculature for rapid reperfusion of ischemic tissue. Whole growth factor, stem cell, and gene therapies have yet to achieve the clinical success needed to become FDA-approved revascularization therapies. Herein, we characterize a biodegradable peptide-based scaffold engineered to mimic VEGF and self-assemble into a nanofibrous, thixotropic hydrogel, SLanc. We found that this injectable hydrogel was rapidly infiltrated by host cells and could be degraded while promoting the generation of neovessels. In mice with induced hind limb ischemia, this synthetic peptide scaffold promoted angiogenesis and ischemic tissue recovery, as shown by Doppler-quantified limb perfusion and a treadmill endurance test. Thirteen-month-old mice showed significant recovery within 7 days of treatment. Biodistribution studies in healthy mice showed that the hydrogel is safe when administered intramuscularly, subcutaneously, or intravenously. These preclinical studies help establish the efficacy of this treatment for peripheral artery disease due to diminished microvascular perfusion, a necessary step before clinical translation. This peptide-based approach eliminates the need for cell transplantation or viral gene transfection (therapies currently being assessed in clinical trials) and could be a more effective regenerative medicine approach to microvascular tissue engineering.


Assuntos
Membro Posterior/irrigação sanguínea , Isquemia/tratamento farmacológico , Nanofibras/uso terapêutico , Neovascularização Fisiológica , Peptídeos/uso terapêutico , Animais , Células Endoteliais da Veia Umbilical Humana , Humanos , Isquemia/patologia , Camundongos Endogâmicos C57BL , Músculos/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Peptídeos/farmacologia , Reperfusão , Distribuição Tecidual/efeitos dos fármacos
11.
Ann Thorac Surg ; 101(6): 2085-96, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27090732

RESUMO

BACKGROUND: Insufficient aortic protection and repair may contribute to the development of aortic aneurysms and dissections (AAD). However, mechanisms of aortic protection and repair are poorly understood. We have shown that the multifunctional kinase AKT2 plays an important role in protecting the aortic wall. Here, we examined whether AKT2 protects against AAD by promoting bone marrow cell (BMC)-mediated aortic protection. METHODS: Irradiated wild-type mice received green fluorescent protein-expressing BMCs from wild-type mice or Akt2(-/-) mice, followed by challenge with angiotensin II (1000 ng/kg/min) infusion for 4 weeks. We compared BMC recruitment, aortic destruction, and AAD development between groups. The direct effects of wild-type and Akt2(-/-) BMCs on smooth muscle cell survival were examined in coculture experiments. RESULTS: After angiotensin II infusion, no (0 of 14) wild-type BMC recipients had AAD; in contrast, 64% (9 of 14) of Akt2(-/-) BMC recipients had AAD (p = 0.002) with severe aortic destruction. Compared with aortas from challenged wild-type BMC recipients, aortas from challenged Akt2(-/-) BMC recipients showed significantly less BMC recruitment, NG2 (neuron-glial antigen 2) progenitor activation, and FSP1 (fibroblast-specific protein 1) fibroblast activation. In addition, aortas from challenged Akt2(-/-) BMC recipients showed increased apoptosis and inflammation. In coculture experiments, wild-type but not Akt2(-/-) BMCs prevented smooth muscle cells from undergoing oxidative stress-induced apoptosis. CONCLUSIONS: After aortic challenge, BMCs are recruited to the aortic wall and provide protection by activating progenitors and fibroblasts and by promoting aortic cell survival. Our findings indicate that AKT2 is involved in these processes and that defects in this pathway may promote progressive degeneration during AAD development.


Assuntos
Aneurisma Aórtico/fisiopatologia , Dissecção Aórtica/fisiopatologia , Células da Medula Óssea/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Dissecção Aórtica/enzimologia , Angiotensina II/toxicidade , Animais , Aneurisma Aórtico/induzido quimicamente , Aneurisma Aórtico/enzimologia , Apoptose , Células da Medula Óssea/enzimologia , Movimento Celular , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/patologia , Genes Reporter , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Pericitos/patologia , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/genética , Quimera por Radiação , Transdução de Sinais , Células-Tronco/enzimologia , Células-Tronco/patologia
12.
Stem Cells ; 33(11): 3254-65, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26205879

RESUMO

MESP1 is considered the first sign of the nascent cardiac mesoderm and plays a critical role in the appearance of cardiac progenitors, while exhibiting a transient expression in the developing embryo. We profiled the transcriptome of a pure population of differentiating MESP1-marked cells and found that they chiefly contribute to the mesendoderm lineage. High-throughput sequencing of endogenous MESP1-bound DNA revealed that MESP1 preferentially binds to two variants of E-box sequences and activates critical mesendoderm modulators, including Eomes, Gata4, Wnt5a, Wnt5b, Mixl1, T, Gsc, and Wnt3. These mesendoderm markers were enriched in the MESP1 marked population before the appearance of cardiac progenitors and myocytes. Further, MESP1-binding is globally associated with H(3)K(27) acetylation, supporting a novel pivotal role of it in regulating target gene epigenetics. Therefore, MESP1, the pioneer cardiac factor, primarily directs the appearance of mesendoderm, the intermediary of the earliest progenitors of mesoderm and endoderm organogenesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Endoderma/fisiologia , Marcação de Genes , Estudo de Associação Genômica Ampla/métodos , Mesoderma/fisiologia , Ativação Transcricional/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/fisiologia , Linhagem Celular Transformada , Linhagem da Célula/fisiologia , Células-Tronco Embrionárias/fisiologia , Endoderma/embriologia , Marcação de Genes/métodos , Masculino , Mesoderma/embriologia , Camundongos , Miócitos Cardíacos/fisiologia , Ligação Proteica/fisiologia
13.
Biochim Biophys Acta ; 1852(7): 1490-7, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25887157

RESUMO

Myotonic dystrophy is a neuromuscular disease of RNA toxicity. The disease gene DMPK harbors expanded CTG trinucleotide repeats on its 3'-UTR. The transcripts of this mutant DMPK led to misregulation of RNA-binding proteins including MBNL1 and Celf1. In myoblasts, CUG-expansion impaired terminal differentiation. In this study, we formally tested how the abundance of Celf1 regulates normal myocyte differentiation, and how Celf1 expression level mediates CUG-expansion RNA toxicity-triggered impairment of myocyte differentiation. As the results, overexpression of Celf1 largely recapitulated the defects of myocytes with CUG-expansion, by increasing myocyte cycling. Knockdown of endogenous Celf1 level led to precocious myotube formation, supporting a negative connection between Celf1 abundance and myocyte terminal differentiation. Finally, knockdown of Celf1 in myocyte with CUG-expansion led to partial rescue, by promoting cell cycle exit. Our results suggest that Celf1 plays a distinctive and negative role in terminal myocyte differentiation, which partially contribute to DM1 RNA toxicity. Targeting Celf1 may be a valid strategy in correcting DM1 muscle phenotypes, especially for congenital cases.


Assuntos
Ciclo Celular , Diferenciação Celular , Mioblastos/metabolismo , Distrofia Miotônica/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas CELF1 , Linhagem Celular , Camundongos , Mioblastos/citologia , Miotonina Proteína Quinase/genética , Miotonina Proteína Quinase/metabolismo , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...