Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Stem Cells ; 41(3): 287-305, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36617947

RESUMO

Neural crest-like stem cells resembling embryonic neural crest cells (NCs) can be derived from adult human tissues such as the epidermis. However, these cells lose their multipotency rapidly in culture limiting their expansion for clinical use. Here, we show that the multipotency of keratinocyte-derived NCs (KC-NCs) can be preserved by activating the Wnt and BMP signaling axis, promoting expression of key NC-specifier genes and ultimately enhancing their differentiation potential. We also show that transcriptional changes leading to multipotency are linked to metabolic reprogramming of KC-NCs to a highly glycolytic state. Specifically, KC-NCs treated with CHIR and BMP2 rely almost exclusively on glycolysis for their energy needs, as seen by increased lactate production, glucose uptake, and glycolytic enzyme activities. This was accompanied by mitochondrial depolarization and decreased mitochondrial ATP production. Interestingly, the glycolytic end-product lactate stabilized ß-catenin and further augmented NC-gene expression. Taken together, our study shows that activation of the Wnt/BMP signaling coordinates the metabolic demands of neural crest-like stem cells governing decisions regarding multipotency and differentiation, with possible implications for regenerative medicine.


Assuntos
Crista Neural , Células-Tronco , Humanos , Diferenciação Celular , Via de Sinalização Wnt
2.
Int J Mol Sci ; 23(18)2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36142746

RESUMO

Our laboratory reported the derivation of neural crest stem cell (NCSC)-like cells from the interfollicular epidermis of the neonatal and adult epidermis. These keratinocyte (KC)-derived Neural Crest (NC)-like cells (KC-NC) could differentiate into functional neurons, Schwann cells (SC), melanocytes, and smooth muscle cells in vitro. Most notably, KC-NC migrated along stereotypical pathways and gave rise to multiple NC derivatives upon transplantation into chicken embryos, corroborating their NC phenotype. Here, we present an innovative design concept for developing anisotropically aligned scaffolds with chemically immobilized biological cues to promote differentiation of the KC-NC towards the SC. Specifically, we designed electrospun nanofibers and examined the effect of bioactive cues in guiding KC-NC differentiation into SC. KC-NC attached to nanofibers and adopted a spindle-like morphology, similar to the native extracellular matrix (ECM) microarchitecture of the peripheral nerves. Immobilization of biological cues, especially Neuregulin1 (NRG1) promoted the differentiation of KC-NC into the SC lineage. This study suggests that poly-ε-caprolactone (PCL) nanofibers decorated with topographical and cell-instructive cues may be a potential platform for enhancing KC-NC differentiation toward SC.


Assuntos
Nanofibras , Células-Tronco Neurais , Animais , Biomimética , Diferenciação Celular , Embrião de Galinha , Sinais (Psicologia) , Crista Neural/metabolismo , Células-Tronco Neurais/metabolismo , Células de Schwann/metabolismo
3.
FASEB J ; 34(3): 3792-3804, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31930567

RESUMO

Intercellular adhesion through homotypic interaction between cadherins regulates multiple cellular processes including cytoskeletal organization, proliferation, and survival. In this paper, we provide evidence that cadherin-11 (CDH11) binds to and promotes cell proliferation both in vitro and in vivo in synergy with the platelet-derived growth factor receptor beta (PDGFRß). Engagement of CDH11 increased the sensitivity of cells to PDGF-BB by 10- to 100-fold, resulting in rapid and sustained phosphorylation of AKT, ultimately promoting and cell proliferation and tissue regeneration. Indeed, wound healing experiments showed that healing was severely compromised in Cdh11-/- mice, as evidenced by significantly decreased proliferation, AKT phosphorylation, and extracellular matrix (ECM) synthesis of dermal cells. Our results shed light into understanding how intercellular adhesion can promote cell proliferation and may have implications for tissue regeneration and cancer progression.


Assuntos
Caderinas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Western Blotting , Caderinas/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Células Cultivadas , Imunofluorescência , Humanos , Imunoprecipitação , Camundongos , Fosforilação/genética , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
4.
Adv Funct Mater ; 30(48)2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33551712

RESUMO

Cell-free small diameter vascular grafts, based on small intestinal submucosa (SIS) functionalized with heparin and vascular endothelial growth factor (VEGF) manufactured and implanted successfully into the arterial system of neonatal lambs, where they remained patent and grew in size with the host to a similar extent and with similar rate as native arteries. Acellular tissue engineered vessels (A-TEV) integrated seamlessly into the native vasculature and developed confluent, functional endothelium that afforded patency. The medial layer was infiltrated by smooth muscle cells, showed no signs of calcification and developed contractile function. The vascular wall underwent remarkable extracellular matrix remodeling exhibiting elastin fibers and even inner elastic lamina within six months. Taken together, our results suggest that VEGF-based A-TEVs may be suitable for treatment of congenital heart disorders to alleviate the need for repeated surgeries, which are currently standard practice.

5.
FASEB J ; 33(4): 5089-5100, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30629890

RESUMO

Recently, our group demonstrated that immobilized VEGF can capture flowing endothelial cells (ECs) from the blood in vitro and promote endothelialization and patency of acellular tissue-engineered vessels (A-TEVs) into the arterial system of an ovine animal model. Here, we demonstrate implantability of submillimeter diameter heparin and VEGF-decorated A-TEVs in a mouse model and discuss the cellular and immunologic response. At 1 mo postimplantation, the graft lumen was fully endothelialized, as shown by expression of EC markers such as CD144, eNOS, CD31, and VEGFR2. Interestingly, the same cells coexpressed leukocyte/macrophage (Mϕ) markers CD14, CD16, VEGFR1, CD38, and EGR2. Notably, there was a stark difference in the cellular makeup between grafts containing VEGF and those containing heparin alone. In VEGF-containing grafts, infiltrating monocytes (MCs) converted into anti-inflammatory M2-Mϕs, and the grafts developed well-demarcated luminal and medial layers resembling those of native arteries. In contrast, in grafts containing only heparin, MCs converted primarily into M1-Mϕs, and the endothelial and smooth muscle layers were not well defined. Our results indicate that VEGF may play an important role in regulating A-TEV patency and regeneration, possibly by regulating the inflammatory response to the implants.-Smith, R. J., Jr., Yi, T., Nasiri, B., Breuer, C. K., Andreadis, S. T. Implantation of VEGF-functionalized cell-free vascular grafts: regenerative and immunological response.


Assuntos
Macrófagos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Endotélio/metabolismo , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo
6.
FASEB J ; 33(10): 10954-10965, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31287964

RESUMO

Collagen type III (COL3) is one of the 3 major collagens in the body, and loss of expression or mutations in the COL3 gene have been associated with the onset of vascular diseases such the Ehlers-Danlos syndrome. Previous work reported a significant reduction of COL3 in tissues such as skin and vessels with aging. In agreement, we found that COL3 was significantly reduced in senescent human mesenchymal stem cells and myofibroblasts derived from patients with Hutchinson-Gilford progeria syndrome, a premature aging syndrome. Most notably, we discovered that ectopic expression of the embryonic transcription factor Nanog homeobox (NANOG) restored COL3 expression by restoring the activity of the TGF-ß pathway that was impaired in senescent cells. RNA sequencing analysis showed that genes associated with the activation of the TGF-ß pathway were up-regulated, whereas negative regulators of the pathway were down-regulated upon NANOG expression. Chromatin immunoprecipitation sequencing and immunoprecipitation experiments revealed that NANOG bound to the mothers against decapentaplegic (SMAD)2 and SMAD3 promoters, in agreement with increased expression and phosphorylation levels of both proteins. Using chemical inhibition, short hairpin RNA knockdown, and gain of function approaches, we established that both SMAD2 and SMAD3 were necessary to mediate the effects of NANOG, but SMAD3 overexpression was also sufficient for COL3 production. In summary, NANOG restored production of COL3, which was impaired by cellular aging, suggesting novel strategies to restore the impaired extracellular matrix production and biomechanical function of aged tissues, with potential implications for regenerative medicine and anti-aging treatments.-Rong, N., Mistriotis, P., Wang, X., Tseropoulos, G., Rajabian, N., Zhang, Y., Wang, J., Liu, S., Andreadis, S. T. Restoring extracellular matrix synthesis in senescent stem cells.


Assuntos
Senescência Celular , Matriz Extracelular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Progéria/metabolismo , Idoso , Células Cultivadas , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Matriz Extracelular/genética , Humanos , Lactente , Células-Tronco Mesenquimais/fisiologia , Miofibroblastos/metabolismo , Miofibroblastos/fisiologia , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
7.
Prog Polym Sci ; 982019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36467305

RESUMO

Native tissues orchestrate their functions by complex interdependent cascades of biochemical and biophysical cues that vary spatially and temporally during cellular processes. Scaffolds with well-tuned structural, mechanical, and biochemical properties have been developed to guide cell behavior and provide insight on cell-matrix interaction. However, static scaffolds very often fail to mimic the dynamicity of native extracellular matrices. Stimuli-responsive scaffolds have emerged as powerful platforms that capture vital features of native tissues owing to their ability to change chemical and physical properties in response to cytocompatible stimuli, thus enabling on-demand manipulation of cell microenvironment. The vast expansion in biorthogonal chemistries and stimuli-responsive functionalities has fuelled further the development of new smart scaffolds that can permit multiple irreversible or reversible spatiotemporal modulation of cell-directing cues, thereby prompting in-depth studies to interpret the decisive elements that regulate cell behavior. Integration of stimuli-responsive hydrogels with current biofabrication technologies has allowed the development of dynamic scaffolds with organizational features and hierarchical architectures similar to native tissues. This review highlights the progress achieved using stimuli-responsive hydrogels in fundamental cell biology studies, with particular emphasis on the interplay between chemistry, biomaterials design, and biofabrication technologies for manipulation of cell microenvironment.

8.
J Cell Sci ; 129(15): 2950-61, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27311482

RESUMO

We discovered that Cadherin-11 (CDH11) regulates collagen and elastin synthesis, both affecting the mechanical properties and contractile function of animal tissues. Using a Cdh11-null mouse model, we observed a significant reduction in the mechanical properties [Youngs' modulus and ultimate tensile strength (UTS)] of Cdh11(-/-) as compared to wild-type (WT) mouse tissues, such as the aorta, bladder and skin. The deterioration of mechanical properties (Youngs' modulus and UTS) was accompanied by reduced collagen and elastin content in Cdh11(-/-) mouse tissues as well as in cells in culture. Similarly, knocking down CDH11 abolished collagen and elastin synthesis in human cells, and consequently reduced their ability to generate force. Conversely, engagement of CDH11 through homophilic interactions, led to swift activation of the TGF-ß and ROCK pathways as evidenced by phosphorylation of downstream effectors. Subsequently, activation of the key transcription factors, MRTF-A (also known as MKL1) and MYOCD led to significant upregulation of collagen and elastin genes. Taken together, our results demonstrate a novel role of adherens junctions in regulating extracellular matrix (ECM) synthesis with implications for many important biological processes, including maintenance of tissue integrity, wound healing and tissue regeneration.


Assuntos
Caderinas/metabolismo , Matriz Extracelular/metabolismo , Animais , Fenômenos Biomecânicos , Caderinas/deficiência , Colágeno/genética , Colágeno/metabolismo , Derme/citologia , Módulo de Elasticidade , Elastina/genética , Elastina/metabolismo , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Modelos Biológicos , Proteínas Nucleares/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Resistência à Tração , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Quinases Associadas a rho/metabolismo
9.
Stem Cells ; 35(5): 1402-1415, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28142205

RESUMO

During development, neural crest (NC) cells are induced by signaling events at the neural plate border of all vertebrate embryos. Initially arising within the central nervous system, NC cells subsequently undergo an epithelial to mesenchymal transition to migrate into the periphery, where they differentiate into diverse cell types. Here we provide evidence that postnatal human epidermal keratinocytes (KC), in response to fibroblast growth factor 2 and insulin like growth factor 1 signals, can be reprogrammed toward a NC fate. Genome-wide transcriptome analyses show that keratinocyte-derived NC cells are similar to those derived from human embryonic stem cells. Moreover, they give rise in vitro and in vivo to NC derivatives such as peripheral neurons, melanocytes, Schwann cells and mesenchymal cells (osteocytes, chondrocytes, adipocytes, and smooth muscle cells). By demonstrating that human keratin-14+ KC can form NC cells, even from clones of single cells, our results have important implications in stem cell biology and regenerative medicine. Stem Cells 2017;35:1402-1415.


Assuntos
Linhagem da Célula , Reprogramação Celular , Células Epidérmicas , Queratinócitos/citologia , Crista Neural/citologia , Idoso , Idoso de 80 Anos ou mais , Diferenciação Celular , Movimento Celular , Reprogramação Celular/genética , Células Clonais , Perfilação da Expressão Gênica , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Recém-Nascido , Queratinócitos/metabolismo , Pessoa de Meia-Idade , Células-Tronco Multipotentes/citologia , Placa Neural/citologia , Transcrição Gênica
10.
Stem Cells ; 35(1): 207-221, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27350449

RESUMO

Cellular senescence as a result of organismal aging or progeroid diseases leads to stem cell pool exhaustion hindering tissue regeneration and contributing to the progression of age related disorders. Here we discovered that ectopic expression of the pluripotent factor NANOG in senescent or progeroid myogenic progenitors reversed cellular aging and restored completely the ability to generate contractile force. To elicit its effects, NANOG enabled reactivation of the ROCK and Transforming Growth Factor (TGF)-ß pathways-both of which were impaired in senescent cells-leading to ACTIN polymerization, MRTF-A translocation into the nucleus and serum response factor (SRF)-dependent myogenic gene expression. Collectively our data reveal that cellular senescence can be reversed and provide a novel strategy to regain the lost function of aged stem cells without reprogramming to the pluripotent state. Stem Cells 2017;35:207-221.


Assuntos
Actinas/metabolismo , Diferenciação Celular , Senescência Celular , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteína Homeobox Nanog/metabolismo , Fator de Resposta Sérica/metabolismo , Idoso , Diferenciação Celular/genética , Senescência Celular/genética , Genoma Humano , Humanos , Modelos Biológicos , Desenvolvimento Muscular/genética , Miofibroblastos/metabolismo , Fenótipo , Progéria/genética , Progéria/patologia , Transdução de Sinais , Transativadores/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Quinases Associadas a rho/metabolismo
11.
Exp Cell Res ; 359(2): 327-336, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28803065

RESUMO

Adherens junctions (AJs) are a key structural component for tissue organization and function. Under fluid shear stress, AJs exhibit dynamic assembly/disassembly, but how shear stress couples to AJs is unclear. In MDCK cells we measured simultaneously the forces in cytoskeletal α-actinin and the density and length of AJs using a genetically coded optical force sensor, actinin-sstFRET, and fluorescently labeled E-cadherin (E-cad). We found that shear stress of 0.74dyn/cm2 for 3h significantly enhanced E-cad expression at cell-cell contacts and this phenomenon has two phases. The initial formation of segregated AJ plaques coincided with a decrease in cytoskeletal tension, but an increase in tension was necessary for expansion of the plaques and the formation of continuous AJs in the later phase. The changes in cytoskeletal tension and reorganization appear to be an upstream process in response to flow since it occurred in both wild type and dominant negative E-cad cells. Disruption of F-actin with a Rho-ROCK inhibitor eliminated AJ growth under flow. These results delineate the shear stress transduction paths in cultured cells, which helps to understand pathology of a range of diseases that involve dysfunction of E-cadherin.


Assuntos
Citoesqueleto de Actina/metabolismo , Junções Aderentes/metabolismo , Mecanotransdução Celular , Estresse Mecânico , Citoesqueleto de Actina/ultraestrutura , Actinina/genética , Actinina/metabolismo , Actinas/genética , Actinas/metabolismo , Junções Aderentes/ultraestrutura , Amidas/farmacologia , Animais , Fenômenos Biomecânicos , Técnicas Biossensoriais , Caderinas/genética , Caderinas/metabolismo , Cães , Transferência Ressonante de Energia de Fluorescência , Regulação da Expressão Gênica , Células Madin Darby de Rim Canino , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Reologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-30505334

RESUMO

The last two decades have seen many advances in regenerative medicine, including the development of tissue engineered vessels (TEVs) for replacement of damaged or diseased arteries or veins. Biomaterials from natural sources as well as synthetic polymeric materials have been employed in engineering vascular grafts. Recently, cell-free grafts have become available opening new possibilities for the next generation, off-the-shelf products. These TEVs are first tested in small or large animal models, which are usually young and healthy. However, the majority of patients in need of vascular grafts are elderly and suffer from comorbidities that may complicate their response to the implants. Therefore, it is important to evaluate TEVs in animal models of vascular disease in order to increase their predictive value and learn how the disease microenvironment may affect the patency and remodeling of vascular grafts. Small animals with various disease phenotypes are readily available due to the availability of transgenic or gene knockout technologies and can be used to address mechanistic questions related to vascular grafting. On the other hand, large animal models with similar anatomy, hematology and thrombotic responses to humans have been utilized in a preclinical setting. We propose that large animal models with certain pathologies or age range may provide more clinically relevant platforms for testing TEVs and facilitate the clinical translation of tissue engineering technologies by increasing the likelihood of success in clinical trials.

13.
J Cell Sci ; 127(Pt 12): 2627-38, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24741067

RESUMO

Although soluble factors, such as transforming growth factor ß1 (TGF-ß1), induce mesenchymal stem cell (MSC) differentiation towards the smooth muscle cell (SMC) lineage, the role of adherens junctions in this process is not well understood. In this study, we found that cadherin-11 but not cadherin-2 was necessary for MSC differentiation into SMCs. Cadherin-11 regulated the expression of TGF-ß1 and affected SMC differentiation through a pathway that was dependent on TGF-ß receptor II (TGFßRII) but independent of SMAD2 or SMAD3. In addition, cadherin-11 activated the expression of serum response factor (SRF) and SMC proteins through the Rho-associated protein kinase (ROCK) pathway. Engagement of cadherin-11 increased its own expression through SRF, indicative of the presence of an autoregulatory feedback loop that committed MSCs to the SMC fate. Notably, SMC-containing tissues (such as aorta and bladder) from cadherin-11-null (Cdh11(-/-)) mice showed significantly reduced levels of SMC proteins and exhibited diminished contractility compared with controls. This is the first report implicating cadherin-11 in SMC differentiation and contractile function in vitro as well as in vivo.


Assuntos
Caderinas/fisiologia , Diferenciação Celular , Células-Tronco Mesenquimais/fisiologia , Miócitos de Músculo Liso/fisiologia , Animais , Caderinas/metabolismo , Adesão Celular , Células Cultivadas , Ativação Enzimática , Retroalimentação Fisiológica , Expressão Gênica , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular , Desenvolvimento Muscular , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Bexiga Urinária/fisiologia , Quinases Associadas a rho/metabolismo
14.
Biomacromolecules ; 17(6): 2293-301, 2016 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-27151393

RESUMO

Previous studies showed that mouse submandibular gland cells form three-dimensional structures when grown on Laminin-111 gels. The use of Laminin-111 for tissue bioengineering is complicated due to its lack of purity. By contrast, the use of synthetic peptides derived from Laminin-111 is beneficial due to their high purity and easy manipulation. Two Laminin-111 peptides have been identified for salivary cells: the A99 peptide corresponding to the α1 chain from Laminin-111 and the YIGSR peptide corresponding to the ß1 chain from Laminin-111, which are important for cell adhesion and migration. We created three-dimensional salivary cell clusters using a modified fibrin hydrogel matrix containing immobilized Laminin-111 peptides. Results indicate that the YIGSR peptide improved morphology and lumen formation in rat parotid Par-C10 cells as compared to cells grown on unmodified fibrin hydrogel. Moreover, a combination of both peptides not only allowed the formation of functional three-dimensional salivary cell clusters but also increased attachment and number of cell clusters. In summary, we demonstrated that fibrin hydrogel decorated with Laminin-111 peptides supports attachment and differentiation of salivary gland cell clusters with mature lumens.


Assuntos
Adesão Celular/fisiologia , Fibrina/química , Hidrogéis/química , Laminina/química , Glândula Parótida/citologia , Fragmentos de Peptídeos/química , Animais , Células Cultivadas , Laminina/metabolismo , Glândula Parótida/efeitos dos fármacos , Glândula Parótida/metabolismo , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Ratos
15.
J Cell Sci ; 126(Pt 12): 2718-29, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23591817

RESUMO

We demonstrate that c-Jun N-terminal kinase (JNK) responds to substrate stiffness and regulates adherens junction (AJ) formation in epithelial cells in 2D cultures and in 3D tissues in vitro and in vivo. Rigid substrates led to JNK activation and AJ disassembly, whereas soft matrices suppressed JNK activity leading to AJ formation. Expression of constitutively active JNK (MKK7-JNK1) induced AJ dissolution even on soft substrates, whereas JNK knockdown (using shJNK) induced AJ formation even on hard substrates. In human epidermis, basal cells expressed phosphorylated JNK but lacked AJ, whereas suprabasal keratinocytes contained strong AJ but lacked phosphorylated JNK. AJ formation was significantly impaired even in the upper suprabasal layers of bioengineered epidermis when prepared with stiffer scaffold or keratinocytes expressing MKK7-JNK1. By contrast, shJNK1 or shJNK2 epidermis exhibited strong AJ even in the basal layer. The results with bioengineered epidermis were in full agreement with the epidermis of jnk1(-/-) or jnk2(-/-) mice. In conclusion, we propose that JNK mediates the effects of substrate stiffness on AJ formation in 2D and 3D contexts in vitro as well as in vivo.


Assuntos
Junções Aderentes/metabolismo , Células Epiteliais/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Animais , Células Cultivadas , Células Epidérmicas , Epiderme/metabolismo , Células Epiteliais/citologia , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , MAP Quinase Quinase 7/metabolismo , Camundongos , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Fosforilação
16.
Annu Rev Biomed Eng ; 16: 1-28, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24819474

RESUMO

Heart disease is the leading cause of morbidity and mortality worldwide, and regenerative therapies that replace damaged myocardium could benefit millions of patients annually. The many cell types in the heart, including cardiomyocytes, endothelial cells, vascular smooth muscle cells, pericytes, and cardiac fibroblasts, communicate via intercellular signaling and modulate each other's function. Although much progress has been made in generating cells of the cardiovascular lineage from human pluripotent stem cells, a major challenge now is creating the tissue architecture to integrate a microvascular circulation and afferent arterioles into such an engineered tissue. Recent advances in cardiac and vascular tissue engineering will move us closer to the goal of generating functionally mature tissue. Using the biology of the myocardium as the foundation for designing engineered tissue and addressing the challenges to implantation and integration, we can bridge the gap from bench to bedside for a clinically tractable engineered cardiac tissue.


Assuntos
Coração/fisiologia , Miocárdio/patologia , Regeneração/fisiologia , Engenharia Tecidual/métodos , Animais , Comunicação Celular , Linhagem da Célula , Técnicas de Cocultura , Células Endoteliais/citologia , Fibroblastos/citologia , Cardiopatias/metabolismo , Humanos , Camundongos , Microcirculação , Músculo Liso Vascular/citologia , Neovascularização Patológica , Células-Tronco Pluripotentes/citologia , Ratos , Células Estromais/citologia
17.
Bioconjug Chem ; 26(7): 1314-27, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25685943

RESUMO

We used magnetofection (MF) to achieve high transfection efficiency into human mesenchymal stem cells (MSCs). A custom-made magnet array, matching well-to-well to a 24-well plate, was generated and characterized. Theoretical predictions of magnetic force distribution within each well demonstrated that there was no magnetic field interference among magnets in adjacent wells. An optimized protocol for efficient gene delivery to human hair follicle derived MSCs (hHF-MSCs) was established using an egfp-encoding plasmid, reaching approximately ∼50% transfection efficiency without significant cytotoxicity. Then we applied the optimized MF protocol to express the pluripotency-associated transcription factor NANOG, which was previously shown to reverse the effects of organismal aging on MSC proliferation and myogenic differentiation capacity. Indeed, MF-mediated NANOG delivery increased proliferation and enhanced the differentiation of hHF-MSCs into smooth muscle cells (SMCs). Collectively, our results show that MF can achieve high levels of gene delivery to MSCs and, therefore, may be employed to moderate or reverse the effects of cellular senescence or reprogram cells to the pluripotent state without permanent genetic modification.


Assuntos
Folículo Piloso/citologia , Proteínas de Homeodomínio/genética , Magnetismo/métodos , Células-Tronco Mesenquimais/metabolismo , Miócitos de Músculo Liso/citologia , Transfecção/métodos , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , DNA/administração & dosagem , DNA/genética , Humanos , Campos Magnéticos , Células-Tronco Mesenquimais/citologia , Desenvolvimento Muscular , Miócitos de Músculo Liso/metabolismo , Proteína Homeobox Nanog , Regulação para Cima
18.
Mol Ther ; 22(11): 1971-82, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24895998

RESUMO

Uncovering the complexity of mesenchymal stem cell (MSC) differentiation requires novel methods to capture the dynamics of the process in a quantitative and high-throughput manner. To this end, we developed a lentiviral array (LVA) of reporters to capture the dynamics of gene and pathway activity during MSC differentiation into adipogenic, chondrogenic, and osteogenic lineages. Our results identified signature promoters and pathways with unique activation profile for each MSC lineage. In combination with chemical inhibitors, lineage-specific reporters predicted the effects of signaling pathway perturbations on MSC differentiation. Interestingly, some pathways were critical for differentiation into all lineages, while others had differential effects on each lineage. Our study suggests that when combined with large chemical or siRNA libraries, the reporter LVA can be used to uncover novel genes and signaling pathways affecting complex biological processes such as stem cell differentiation or reprogramming.


Assuntos
Células da Medula Óssea/citologia , Genes Reporter , Folículo Piloso/citologia , Lentivirus/genética , Células-Tronco Mesenquimais/citologia , Adipogenia , Adulto , Idoso , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Condrogênese , Perfilação da Expressão Gênica , Vetores Genéticos , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Osteogênese , Transdução de Sinais
19.
FASEB J ; 27(12): 4853-65, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23995291

RESUMO

Functional smooth muscle engineering requires isolation and expansion of smooth muscle cells (SMCs), and this process is particularly challenging for visceral smooth muscle tissue where progenitor cells have not been clearly identified. Herein we showed for the first time that efficient SMCs can be obtained from human amniotic fluid stem cells (hAFSCs). Clonal lines were generated from c-kit(+) hAFSCs. Differentiation toward SM lineage (SMhAFSCs) was obtained using a medium conditioned by PDGF-BB and TGF-ß1. Molecular assays revealed higher level of α smooth muscle actin (α-SMA), desmin, calponin, and smoothelin in SMhAFSCs when compared to hAFSCs. Ultrastructural analysis demonstrated that SMhAFSCs also presented in the cytoplasm increased intermediate filaments, dense bodies, and glycogen deposits like SMCs. SMhAFSC metabolism evaluated via mass spectrometry showed higher glucose oxidation and an enhanced response to mitogenic stimuli in comparison to hAFSCs. Patch clamp of transduced hAFSCs with lentiviral vectors encoding ZsGreen under the control of the α-SMA promoter was performed demonstrating that SMhAFSCs retained a smooth muscle cell-like electrophysiological fingerprint. Eventually SMhAFSCs contractility was evident both at single cell level and on a collagen gel. In conclusion, we showed here that hAFSCs under selective culture conditions are able to give rise to functional SMCs.


Assuntos
Líquido Amniótico/citologia , Diferenciação Celular , Linhagem da Célula , Células-Tronco Fetais/citologia , Células-Tronco Multipotentes/citologia , Miócitos de Músculo Liso/citologia , Actinas/genética , Actinas/metabolismo , Potenciais de Ação , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Meios de Cultivo Condicionados/farmacologia , Citoplasma/metabolismo , Citoplasma/ultraestrutura , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Desmina/genética , Desmina/metabolismo , Células-Tronco Fetais/efeitos dos fármacos , Células-Tronco Fetais/metabolismo , Células-Tronco Fetais/fisiologia , Glucose/metabolismo , Glicogênio/metabolismo , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/metabolismo , Células-Tronco Multipotentes/fisiologia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Calponinas
20.
Cardiovasc Res ; 120(8): 839-854, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38742656

RESUMO

Cell-free tissue-engineered vascular grafts provide a promising alternative to treat cardiovascular disease, but timely endothelialization is essential for ensuring patency and proper functioning post-implantation. Recent studies from our lab showed that blood cells like monocytes (MCs) and macrophages (Mϕ) may contribute directly to cellularization and regeneration of bioengineered arteries in small and large animal models. While MCs and Mϕ are leucocytes that are part of the innate immune response, they share common developmental origins with endothelial cells (ECs) and are known to play crucial roles during vessel formation (angiogenesis) and vessel repair after inflammation/injury. They are highly plastic cells that polarize into pro-inflammatory and anti-inflammatory phenotypes upon exposure to cytokines and differentiate into other cell types, including EC-like cells, in the presence of appropriate chemical and mechanical stimuli. This review focuses on the developmental origins of MCs and ECs; the role of MCs and Mϕ in vessel repair/regeneration during inflammation/injury; and the role of chemical signalling and mechanical forces in Mϕ inflammation that mediates vascular graft regeneration. We postulate that comprehensive understanding of these mechanisms will better inform the development of strategies to coax MCs/Mϕ into endothelializing the lumen and regenerate the smooth muscle layers of cell-free bioengineered arteries and veins that are designed to treat cardiovascular diseases and perhaps the native vasculature as well.


Assuntos
Prótese Vascular , Macrófagos , Monócitos , Regeneração , Engenharia Tecidual , Humanos , Monócitos/metabolismo , Monócitos/transplante , Engenharia Tecidual/métodos , Animais , Macrófagos/metabolismo , Neovascularização Fisiológica , Fenótipo , Implante de Prótese Vascular/instrumentação , Implante de Prótese Vascular/efeitos adversos , Células Endoteliais/metabolismo , Células Endoteliais/transplante , Desenho de Prótese , Mecanotransdução Celular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA