Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Autoimmun ; 145: 103217, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38581915

RESUMO

The autoimmunity-promoting cytokine, Interleukin-15 (IL-15), is often claimed to be a key pathogenic cytokine in alopecia areata (AA). Yet, rhIL-15 promotes human hair follicle (HF) growth ex vivo. We have asked whether the expression of IL-15 and its receptor (IL-15R) isoforms is altered in human AA and how IL-15 impacts on human HF immune privilege (HF-IP) in the presence/absence of interferon-γ (IFNγ), the well-documented key AA-pathogenic cytokine, as well as on hair regrowth after experimental AA induction in vivo. Quantitative immunohistomorphometry showed the number of perifollicular IL-15+ T cells in AA skin biopsies to be significantly increased compared to healthy control skin, while IL-15, IL-15Rα, and IL-15Rγ protein expression within the hair bulb were significantly down-regulated in AA HFs. In organ-cultured human scalp HFs, rhIL-15 significantly reduced hair bulb expression of MICA, the key "danger" signal in AA pathogenesis, and increased production of the HF-IP guardian, α-MSH. Crucially, ex vivo, rhIL-15 prevented IFNγ-induced HF-IP collapse, restored a collapsed HF-IP by IL-15Rα-dependent signaling (as documented by IL-15Rα-silencing), and protected AA-preventive immunoinhibitory iNKT10 cells from IFNγ-induced apoptosis. rhIL-15 even promoted hair regrowth after experimental AA induction in human scalp skin xenotransplants on SCID/beige mice in vivo. Our data introduce IL-15 as a novel, functionally important HF-IP guardian whose signaling is constitutively defective in scalp HFs of AA patients. Our data suggest that selective stimulation of intrafollicular IL-15Rα signaling could become a novel therapeutic approach in AA management, while blocking it pharmacologically may hinder both HF-IP restoration and hair re-growth and may thus make HFs more vulnerable to AA relapse.


Assuntos
Alopecia em Áreas , Folículo Piloso , Privilégio Imunológico , Interferon gama , Interleucina-15 , Interleucina-15/metabolismo , Interleucina-15/imunologia , Folículo Piloso/imunologia , Folículo Piloso/metabolismo , Humanos , Animais , Alopecia em Áreas/imunologia , Alopecia em Áreas/metabolismo , Camundongos , Interferon gama/metabolismo , Feminino , Receptores de Interleucina-15/metabolismo , Receptores de Interleucina-15/imunologia , Masculino , Adulto , Pessoa de Meia-Idade , Subunidade alfa de Receptor de Interleucina-15/metabolismo , Subunidade alfa de Receptor de Interleucina-15/imunologia , Pele/imunologia , Pele/metabolismo , Pele/patologia , Modelos Animais de Doenças
2.
Allergy ; 78(6): 1538-1553, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36597714

RESUMO

BACKGROUND: The key signals that suffice to induce atopic dermatitis (AD) in human skin remain incompletely understood. Also, current mouse models reflect human AD only unsatisfactorily. Therefore, we have asked whether a humanized AD mouse model can be developed that reflects human AD more faithfully and permit to identify key signals that suffice to induce AD lesions in previously healthy human skin in vivo. METHODS: Healthy human skin from non-atopic donors was transplanted onto SCID/beige mice. After xenotransplant reinnervation by mouse sensory nerve fibers had occurred, mixed autologous human Th2 CD4+ and Tc2 CD8+ T cells that had been pretreated in vitro with IL-2, IL-4, and LPS were injected intradermally into the xenotransplants without skin barrier disruption. RESULTS: Injected non-atopic xenotransplants rapidly developed a morphological, functional, and immunological phenocopy of human AD lesions regarding skin barrier defects, immunopathology including intraepidermal eosinophils, mast cell activation, increase of thymic stromal lymphopoietin, eotaxin-1 and type 2 cytokine circuits, and even showed characteristic neuroimmunological abnormalities such as ß2-adrenergic receptor downregulation. The experimentally induced AD lesions in human skin responded to standard AD therapy (topical dexamethasone or tacrolimus; systemic anti-IL-4Rα antibody [dupilumab]), and relapsed when neurogenic skin inflammation was induced by exposing mice to perceived stress. CONCLUSIONS: This new animal model uniquely mimics the complexity of human AD and its clinical response to standard therapy and psychoemotional stressors in vivo, and shows that Th2-polarized lymphocytes associated with excessive IL-4Rα-mediated signaling suffice to induce human AD skin lesions, while atopy and epidermal barrier disruption are dispensable.


Assuntos
Dermatite Atópica , Humanos , Camundongos , Animais , Camundongos SCID , Pele/patologia , Citocinas/metabolismo , Linfócitos T CD8-Positivos
3.
Adv Skin Wound Care ; 36(10): 1-10, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37729169

RESUMO

OBJECTIVE: Skin adhesives offer many advantages over traditional wound-closure devices. Recently, the current research group reported on tissue adhesives composed of natural polymers (gelatin and alginate), which are biocompatible with mechanical properties suitable for tissue adhesion. The objective of the present study was to conduct clinical and histologic assessment of this hemostatic bioadhesive in the healing of long skin incisions (≥4 cm) in comparison with traditional and commercially available methods. METHODS: Researchers created 24 long incisions on the ventral side of two domestic pigs to compare four different treatment modalities: two topical bioadhesives based on gelatin and alginate combined with the hemostatic agent kaolin, nylon sutures, and commercial tissue adhesive N-butyl-2-cyanoacrylate. The bioadhesive compounds were spread on the incision surface and then mixed either manually or with a double-headed syringe. After 14 days, clinical and histologic measurements were performed to evaluate the healing phase of the wounds. RESULTS: The bioadhesive formulation that contained a relatively low crosslinker concentration demonstrated superior results to the formulation that contained a standard crosslinker concentration. However, no significant statistical differences were observed compared with the control incisions (sutures and commercial adhesive N-butyl-2-cyanoacrylate). This was verified by immunohistochemical analysis for epithelial integrity and scar formation as well as by clinical assessment. CONCLUSIONS: This newly developed bioadhesive demonstrated suitable properties for the closure of long incisions in a porcine skin model.


Assuntos
Embucrilato , Hemostáticos , Ferida Cirúrgica , Adesivos Teciduais , Suínos , Animais , Hemostáticos/farmacologia , Hemostáticos/uso terapêutico , Adesivos Teciduais/farmacologia , Adesivos Teciduais/uso terapêutico , Gelatina , Alginatos
4.
Exp Dermatol ; 31(4): 567-576, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34787924

RESUMO

BACKGROUND: Psoriasis is characterized by aberrant activation of several pro-inflammatory circuits as well as abnormal hyperproliferation and dysregulated apoptosis of keratinocytes (KCs). Most currently available therapeutic options primarily target psoriasis-associated immunological defects rather than epidermal abnormalities. OBJECTIVE: To investigate the efficacy of the histone deacetylase (HDAC) inhibitor, Vorinostat, in targeting hyperproliferation and impaired apoptosis in psoriatic skin. METHODS: Vorinostat effect was investigated in primary KCs cell cultures using cell cycle analysis by flow cytometry, apoptosis assays (Annexin V-FICH and caspase-3/7) and antibody arrays, qRT-PCR and immunohistochemistry. Vorinostat impact on clinical manifestations of psoriasis was investigated in a chimeric mouse model. RESULTS: Vorinostat was found to inhibit KCs proliferation and to induce their differentiation and apoptosis. Using a chimeric mouse model, vorinostat was found to result in marked attenuation of a psoriasiform phenotype with a significant decrease in epidermal thickness and inhibition of epidermal proliferation. CONCLUSIONS: Our results support the notion that vorinostat, a prototypic HDAC inhibitor, may be of potential use in the treatment of psoriasis and other hyperproliferative skin disorders.


Assuntos
Inibidores de Histona Desacetilases , Psoríase , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Camundongos , Psoríase/tratamento farmacológico , Vorinostat/farmacologia , Vorinostat/uso terapêutico
5.
Exp Dermatol ; 30(3): 319-336, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33368555

RESUMO

Mouse models for atopic dermatitis (AD) are an indispensable preclinical research tool for testing new candidate AD therapeutics and for interrogating AD pathobiology in vivo. In this Viewpoint, we delineate why, unfortunately, none of the currently available so-called "AD" mouse models satisfactorily reflect the clinical complexity of human AD, but imitate more "allergic" or "irriant" contact dermatitis conditions. This limits the predictive value of AD models for clinical outcomes of new tested candidate AD therapeutics and the instructiveness of mouse models for human AD pathophysiology research. Here, we propose to initiate a rational debate on the minimal criteria that a mouse model should meet in order to be considered relevant for human AD. We suggest that valid AD models should at least meet the following criteria: (a) an AD-like epidermal barrier defect with reduced filaggrin expression along with hyperproliferation, hyperplasia; (b) increased epidermal expression of thymic stromal lymphopoietin (TSLP), periostin and/or chemokines such as TARC (CCL17); (c) a characteristic dermal immune cell infiltrate with overexpression of some key cytokines such as IL-4, IL-13, IL-31 and IL-33; (d) distinctive "neurodermatitis" features (sensory skin hyperinnervation, defective beta-adrenergic signalling, neurogenic skin inflammation and triggering or aggravation of AD-like skin lesions by perceived stress); and (e) response of experimentally induced skin lesions to standard AD therapy. Finally, we delineate why humanized AD mouse models (human skin xenotransplants on SCID mice) offer a particularly promising preclinical research alternative to the currently available "AD" mouse models.


Assuntos
Dermatite Atópica/induzido quimicamente , Dermatite Atópica/fisiopatologia , Modelos Animais de Doenças , Animais , Biomarcadores , Calcitriol/análogos & derivados , Dermatite Atópica/genética , Dermatite Atópica/patologia , Haptenos , Humanos , Camundongos , Ovalbumina , Fenômenos Fisiológicos da Pele
6.
J Investig Dermatol Symp Proc ; 20(1): S11-S15, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33099377

RESUMO

The C3H/HeJ model has long dominated basic alopecia areata (AA) in vivo research and has been used as proof-of-principle that Jak inhibitors are suitable agents for AA management in vivo. However, its histologic features are not typical of human AA, and it is questionable whether it is sufficiently clinically predictive for evaluating the therapeutic effects of candidate AA agents. Instead, the humanized mouse model of AA has been used to functionally demonstrate the role of key immune cells in AA pathogenesis and to discover human-specific pharmacologic targets in AA management. Therefore, we advocate the use of both models in future preclinical AA research.


Assuntos
Alopecia em Áreas/tratamento farmacológico , Alopecia em Áreas/patologia , Modelos Animais de Doenças , Animais , Avaliação Pré-Clínica de Medicamentos , Humanos , Camundongos , Camundongos Endogâmicos C3H
7.
J Allergy Clin Immunol ; 144(6): 1478-1489, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31606262

RESUMO

This current review explores selected and as yet insufficiently investigated frontiers in current alopecia areata (AA) pathobiology research, with an emphasis on potential "new" players in AA pathobiology that deserve more systematic exploration and therapeutic targeting. Indeed, new evidence suggests that CD8+ T cells, which have long been thought to be the central players in AA pathobiology, are not the only drivers of disease. Instead, subsets of natural killer (NK) and so-called "unconventional" T cells (invariant NK T cells, γδ T cells, classic NK cells, and type 1 innate lymphoid cells), all of which can produce large amounts of IFN-γ, might also drive AA pathobiology independent of classical, autoantigen-dependent CD8+ T-cell functions. Another important new frontier is the role of regulatory lymphocyte subsets, such as regulatory T cells, γδ regulatory T cells, NKT10 cells, and perifollicular mast cells, in maintaining physiologic hair follicle immune privilege (IP); the extent to which these functions are defective in patients with AA; and how this IP-protective role could be restored therapeutically in patients with established AA. Broadening our AA research horizon along the lines suggested above promises not only to open the door to innovative and even more effective immunotherapy strategies for AA but will also likely be relevant for other autoimmune disorders in which pathobiology, ectopic MHC class I expression, and IP collapse play an important role.


Assuntos
Alopecia em Áreas/imunologia , Doenças Autoimunes/imunologia , Imunidade Inata , Alopecia em Áreas/patologia , Autoantígenos/imunologia , Doenças Autoimunes/patologia , Pesquisa Biomédica , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/patologia
8.
Immunology ; 158(3): 171-193, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31424569

RESUMO

Activated T cells are pathological in various autoimmune and inflammatory diseases including Psoriasis, and also in graft rejection and graft-versus-host-disease. In these pathological conditions, selective silencing of activated T cells through physiological receptors they express remains a clinical challenge. In our previous studies we found that activation of dopamine receptors (DRs) in resting human T cells activates these cells, and induces by itself many beneficial T cell functions. In this study, we found that normal human T cells express all types of DRs, and that expression of D1R, D4R and D5R increases profoundly after T cell receptor (TCR) activation. Interestingly, DR agonists shift the membrane potential (Vm ) of both resting and activated human T cells, and induces instantaneous T cell depolarization within 15 seconds only. Thus, activation of DRs in T cells depolarize these immune cells, alike activation of DRs in neural cells. The skin of Psoriasis patients contains 20-fold more D1R+ T cells than healthy human skin. In line with that, 25-fold more D1R+ T cells are present in Psoriasis humanized mouse model. Highly selective D1-like receptor agonists, primarily Fenoldopam (Corlopam) - a D1-like receptor agonist and a drug used in hypertension, induced the following suppressive effects on activated T cells of Psoriasis patients: reduced chemotactic migration towards the chemokine SDF-1/CXCL12; reduced dramatically the secretion of eight cytokines: tumor necrosis factor-α, interferon-γ, interleukin-1ß (IL-1ß), IL-2, IL-4, IL-6, IL-8 and IL-10; and reduced three T cell activation proteins/markers: CD69, CD28 and IL-2. Next, we invented a novel topical/dermal Fenoldopam formulation, allowing it to be spread on, and providing prolonged and regulated release in, diseased skin. Our novel topical/dermal Fenoldopam: reduced secretion of the eight cytokines by activated human T cells; reduced IL-1ß and IL-6 secretion by human lipopolysaccharide-inflamed skin; eliminated preferentially >90% of live and large/proliferating human T cells. Together, our findings show for the first time that both resting and activated T cells are depolarized instantaneously via DRs, and that targeting D1-like receptors in activated T cells and inflamed human skin by Fenoldopam, in Psoriasis, and potentially in other T cell-mediated diseases, could be therapeutic. Validation in vivo is required.


Assuntos
Fenoldopam/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Psoríase/imunologia , Receptores Dopaminérgicos/imunologia , Pele/imunologia , Linfócitos T/imunologia , Adulto , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos CD28/imunologia , Citocinas/imunologia , Feminino , Humanos , Lectinas Tipo C/imunologia , Masculino , Pessoa de Meia-Idade , Psoríase/patologia , Pele/patologia , Linfócitos T/patologia
10.
J Autoimmun ; 91: 61-72, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29680372

RESUMO

Alopecia areata (AA) is understood to be a CD8+/NKG2D+ T cell-dependent autoimmune disease. Here, we demonstrate that human AA pathogenesis of is also affected by iNKT10 cells, an unconventional T cell subtype whose number is significantly increased in AA compared to healthy human skin. AA lesions can be rapidly induced in healthy human scalp skin xenotransplants on Beige-SCID mice by intradermal injections of autologous healthy-donor PBMCs pre-activated with IL-2. We show that in this in vivo model, the development of AA lesions is prevented by recognized the iNKT cell activator, α-galactosylceramide (α-GalCer), which stimulates iNKT cells to expand and produce IL-10. Moreover, in pre-established humanized mouse AA lesions, hair regrowth is promoted by α-GalCer treatment through a process requiring both effector-memory iNKT cells, which can interact directly with CD8+/NKG2D+ T cells, and IL-10. This provides the first in vivo evidence in a humanized model of autoimmune disease that iNKT10 cells are key disease-protective lymphocytes. Since these regulatory NKT cells can both prevent the development of AA lesions and promote hair re-growth in established AA lesions, targeting iNKT10 cells may have preventive and therapeutic potential also in other autoimmune disorders related to AA.


Assuntos
Alopecia em Áreas/imunologia , Imunoterapia Adotiva/métodos , Células T Matadoras Naturais/imunologia , Transplante de Pele , Pele/patologia , Adulto , Animais , Autoimunidade , Células Cultivadas , Modelos Animais de Doenças , Feminino , Galactosilceramidas/imunologia , Humanos , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Ativação Linfocitária , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Células T Matadoras Naturais/transplante , Transplante Heterólogo
14.
Lancet ; 393(10169): 318-319, 2019 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-30696569
15.
Exp Dermatol ; 23(7): 464-5, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24673207

RESUMO

In a recently published issue of the journal, Bracke et al. demonstrate an impressive improvement in psoriasiform features in allogeneic human skin grafts transplanted onto immune-deficient mice. This improvement was achieved using a novel nanosome (SECosome) as a vehicle for delivering topically applied siRNA to human epidermis. Targeting the gene DFB4 with this delivery system, they prevented translation of the antimicrobial peptide, human ß defensin-2(hBD2), thus normalizing the psoriasiform epidermal phenotype of siRNA/SECosome-treated human skin grafts. This study encourages the exploration of topical gene silencing strategies in dermatology and refocuses our attention on both, the role of hBD2 in psoriasis pathogenesis and the thorny question which animal model reflects human psoriasis most faithfully.


Assuntos
Nanomedicina/métodos , Nanopartículas/química , Psoríase/genética , Psoríase/terapia , RNA Interferente Pequeno/uso terapêutico , beta-Defensinas/genética , Acetilcisteína/metabolismo , Animais , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Epiderme/metabolismo , Inativação Gênica , Humanos , Camundongos , Fenótipo , Transplante de Pele , beta-Defensinas/metabolismo
17.
J Investig Dermatol Symp Proc ; 16(1): S37-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24326548

RESUMO

Although alopecia areata (AA) is not life threatening, it may lead to severe psychological disturbances, reducing the quality of life in all ages. Thus, a new animal model is needed for shedding more light onto the pathogenesis of this cell-mediated, organ-specific autoimmune disease to identify more effective therapeutic strategies. Recently, we succeeded in developing a new humanized mouse model of AA, which includes transplantation of healthy human scalp skin obtained from normal volunteers on to severe-combined immunodeficient mice. This is followed by intradermal injection of either autologous or allogeneic peripheral blood mononuclear cells, which had been cultured with high dose of IL-2 and enriched for natural killer group 2D-positive (NKG2D+) and CD56+ cells. This protocol leads to rapid and predictable development of focal hair loss, with all the characteristic clinical, histological, and immunohistochemical features of AA. This humanized mouse AA model underscores the functional importance of NKG2D+ and CD56+ cells in AA pathogenesis and promises to be instrumental for identifying novel AA treatment strategies.


Assuntos
Alopecia em Áreas/imunologia , Modelos Animais de Doenças , Células Matadoras Naturais/imunologia , Animais , Antígeno CD56/análise , Humanos , Células Matadoras Naturais/química , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/análise , Transplante de Pele , Transplante Heterólogo
18.
Elife ; 122023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36930216

RESUMO

Here, we have explored the involvement of innate lymphoid cells-type 1 (ILC1) in the pathogenesis of alopecia areata (AA), because we found them to be significantly increased around lesional and non-lesional HFs of AA patients. To further explore these unexpected findings, we first co-cultured autologous circulating ILC1-like cells (ILC1lc) with healthy, but stressed, organ-cultured human scalp hair follicles (HFs). ILClc induced all hallmarks of AA ex vivo: they significantly promoted premature, apoptosis-driven HF regression (catagen), HF cytotoxicity/dystrophy, and most important for AA pathogenesis, the collapse of the HFs physiological immune privilege. NKG2D-blocking or IFNγ-neutralizing antibodies antagonized this. In vivo, intradermal injection of autologous activated, NKG2D+/IFNγ-secreting ILC1lc into healthy human scalp skin xenotransplanted onto SCID/beige mice sufficed to rapidly induce characteristic AA lesions. This provides the first evidence that ILC1lc, which are positive for the ILC1 phenotype and negative for the classical NK markers, suffice to induce AA in previously healthy human HFs ex vivo and in vivo, and further questions the conventional wisdom that AA is always an autoantigen-dependent, CD8 +T cell-driven autoimmune disease.


Assuntos
Alopecia em Áreas , Camundongos , Animais , Humanos , Alopecia em Áreas/patologia , Autoimunidade , Imunidade Inata , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Linfócitos/patologia , Camundongos SCID , Folículo Piloso
19.
Sci Adv ; 8(25): eabm6756, 2022 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-35749494

RESUMO

Transplanting aged human skin onto young SCID/beige mice morphologically rejuvenates the xenotransplants. This is accompanied by angiogenesis, epidermal repigmentation, and substantial improvements in key aging-associated biomarkers, including ß-galactosidase, p16ink4a, SIRT1, PGC1α, collagen 17A, and MMP1. Angiogenesis- and hypoxia-related pathways, namely, vascular endothelial growth factor A (VEGF-A) and HIF1A, are most up-regulated in rejuvenated human skin. This rejuvenation cascade, which can be prevented by VEGF-A-neutralizing antibodies, appears to be initiated by murine VEGF-A, which then up-regulates VEGF-A expression/secretion within aged human skin. While intradermally injected VEGF-loaded nanoparticles suffice to induce a molecular rejuvenation signature in aged human skin on old mice, VEGF-A treatment improves key aging parameters also in isolated, organ-cultured aged human skin, i.e., in the absence of functional skin vasculature, neural, or murine host inputs. This identifies VEGF-A as the first pharmacologically pliable master pathway for human organ rejuvenation in vivo and demonstrates the potential of our humanized mouse model for clinically relevant aging research.

20.
Dev Biol ; 335(2): 374-84, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19765572

RESUMO

Vertebrate heart development is derived from paired primordia of anterior dorsolateral mesoderm expressing Nkx2.5 and GATA4 transcription factors. Yet growth factors and intracellular pathways specifying heart precursor gene expression are poorly understood. In the present work, we investigated the signaling events initiating Nkx2.5 expression in Xenopus laevis. We describe here that fibroblast growth factor (FGF) initiates the expression of Nkx2.5 without affecting GATA4. At gastrula, FGF3 is expressed in anterior neural ectoderm, and results presented here indicate that this tissue is involved in the induction of Nkx2.5 expression in neighboring lateral tissues. Further studies indicate that the intracellular p38 MAPK and the CREB transcription factor function downstream of FGF to initiate Nkx2.5 expression. Activation of the p38 MAPK pathway and of the CREB protein is both necessary and sufficient for the initial expression of Nkx2.5. Therefore, we would like to suggest that FGF expressed in anterior neural ectoderm is a major inducer of Nkx2.5 expression in neighboring cells. In these cells, FGF activates an intracellular p38 MAPK signaling pathway and its downstream target, the CREB transcription factor, all participating in the expression of Nkx2.5 in cardiac progenitors.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ectoderma/metabolismo , Fatores de Crescimento de Fibroblastos/fisiologia , Proteínas de Homeodomínio/metabolismo , Miocárdio/metabolismo , Sistema Nervoso/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Fatores de Crescimento de Fibroblastos/metabolismo , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Hibridização In Situ , Microinjeções , Miocárdio/citologia , Miocárdio/enzimologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células-Tronco/enzimologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Proteínas de Xenopus/genética , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA