Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
PLoS Genet ; 19(2): e1010614, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36745673

RESUMEN

Enhancers are context-specific regulators of expression that drive biological complexity and variation through the redeployment of conserved genes. An example of this is the enhancer-mediated control of Engrailed 1 (EN1), a pleiotropic gene whose expression is required for the formation of mammalian eccrine sweat glands. We previously identified the En1 candidate enhancer (ECE) 18 cis-regulatory element that has been highly and repeatedly derived on the human lineage to potentiate ectodermal EN1 and induce our species' uniquely high eccrine gland density. Intriguingly, ECE18 quantitative activity is negligible outside of primates and ECE18 is not required for En1 regulation and eccrine gland formation in mice, raising the possibility that distinct enhancers have evolved to modulate the same trait. Here we report the identification of the ECE20 enhancer and show it has conserved functionality in mouse and human developing skin ectoderm. Unlike ECE18, knock-out of ECE20 in mice reduces ectodermal En1 and eccrine gland number. Notably, we find ECE20, but not ECE18, is also required for En1 expression in the embryonic mouse brain, demonstrating that ECE20 is a pleiotropic En1 enhancer. Finally, that ECE18 deletion does not potentiate the eccrine phenotype of ECE20 knock-out mice supports the secondary incorporation of ECE18 into the regulation of this trait in primates. Our findings reveal that the mammalian En1 regulatory machinery diversified to incorporate both shared and lineage-restricted enhancers to regulate the same phenotype, and also have implications for understanding the forces that shape the robustness and evolvability of developmental traits.


Asunto(s)
Genes Homeobox , Proteínas de Homeodominio , Ratones , Animales , Humanos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Ratones Noqueados , Fenotipo , Glándulas Sudoríparas/metabolismo , Mamíferos/genética , Mamíferos/metabolismo
2.
Adv Healthc Mater ; 13(12): e2303256, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38207170

RESUMEN

Janus kinase (JAK) inhibitors are approved for many dermatologic disorders, but their use is limited by systemic toxicities including serious cardiovascular events and malignancy. To overcome these limitations, injectable hydrogels are engineered for the local and sustained delivery of baricitinib, a representative JAK inhibitor. Hydrogels are formed via disulfide crosslinking of thiolated hyaluronic acid macromers. Dynamic thioimidate bonds are introduced between the thiolated hyaluronic acid and nitrile-containing baricitinib for drug tethering, which is confirmed with 1H and 13C nuclear magnetic resonance (NMR). Release of baricitinib is tunable over six weeks in vitro and active in inhibiting JAK signaling in a cell line containing a luciferase reporter reflecting interferon signaling. For in vivo activity, baricitinib hydrogels or controls are injected intradermally into an imiquimod-induced mouse model of psoriasis. Imiquimod increases epidermal thickness in mice, which is unaffected when treated with baricitinib or hydrogel alone. Treatment with baricitinib hydrogels suppresses the increased epidermal thickness in mice treated with imiquimod, suggesting that the sustained and local release of baricitinib is important for a therapeutic outcome. This study is the first to utilize a thioimidate chemistry to deliver JAK inhibitors to the skin through injectable hydrogels, which has translational potential for treating inflammatory disorders.


Asunto(s)
Azetidinas , Hidrogeles , Purinas , Pirazoles , Piel , Sulfonamidas , Animales , Hidrogeles/química , Purinas/química , Purinas/farmacología , Sulfonamidas/química , Sulfonamidas/farmacología , Sulfonamidas/administración & dosificación , Ratones , Pirazoles/química , Pirazoles/farmacología , Azetidinas/química , Azetidinas/farmacología , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Humanos , Psoriasis/tratamiento farmacológico , Psoriasis/patología , Psoriasis/inducido químicamente , Imiquimod/química , Imiquimod/farmacología , Inhibidores de las Cinasas Janus/química , Inhibidores de las Cinasas Janus/farmacología , Femenino
3.
Dev Dyn ; 239(10): 2619-26, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21038446

RESUMEN

The hair follicle is considered to be a model system for studying organogenesis. In our initial study using mouse cells (Zheng et al., 2005) we found that new hair follicle formation always starts from an epithelial platform: the epidermal cells aggregate, the aggregates encyst, and from the periphery of the cysts, centrifugally, hair buds, pegs, and follicles form. In this report, we extend our initial study to four distantly related mammals: opossum, rat, dog and human. We find that in these four species, plus mouse, the most trichogenic cells are found in the earliest stages of hair follicle development and that the cellular mechanism of new hair follicle formation starting from dissociated cells is largely the same. These studies suggest that there is essentially one way by which dissociated mammalian skin cells form a new hair follicle in vivo and that this mechanism has been highly conserved.


Asunto(s)
Folículo Piloso/embriología , Organogénesis/fisiología , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Perros , Folículo Piloso/citología , Humanos , Zarigüeyas , Organogénesis/genética , Ratas , Piel/citología , Piel/metabolismo
4.
Science ; 373(6554)2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34326208

RESUMEN

Emerging studies indicate that the immune system can regulate systemic metabolism. Here, we show that thymic stromal lymphopoietin (TSLP) stimulates T cells to induce selective white adipose loss, which protects against obesity, improves glucose metabolism, and mitigates nonalcoholic steatohepatitis. Unexpectedly, adipose loss was not caused by alterations in food intake, absorption, or energy expenditure. Rather, it was induced by the excessive loss of lipids through the skin as sebum. TSLP and T cells regulated sebum release and sebum-associated antimicrobial peptide expression in the steady state. In human skin, TSLP expression correlated directly with sebum-associated gene expression. Thus, we establish a paradigm in which adipose loss can be achieved by means of sebum hypersecretion and uncover a role for adaptive immunity in skin barrier function through sebum secretion.


Asunto(s)
Tejido Adiposo Blanco/anatomía & histología , Citocinas/metabolismo , Sebo/metabolismo , Piel/metabolismo , Inmunidad Adaptativa , Animales , Citocinas/genética , Dieta , Glucosa/metabolismo , Homeostasis , Humanos , Inmunoglobulinas/metabolismo , Metabolismo de los Lípidos , Ratones , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/prevención & control , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores de Citocinas/metabolismo , Glándulas Sebáceas/metabolismo , Transducción de Señal , Piel/inmunología , Linfocitos T/fisiología , Pérdida de Peso , Linfopoyetina del Estroma Tímico
5.
Science ; 355(6326): 748-752, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28059714

RESUMEN

Although regeneration through the reprogramming of one cell lineage to another occurs in fish and amphibians, it has not been observed in mammals. We discovered in the mouse that during wound healing, adipocytes regenerate from myofibroblasts, a cell type thought to be differentiated and nonadipogenic. Myofibroblast reprogramming required neogenic hair follicles, which triggered bone morphogenetic protein (BMP) signaling and then activation of adipocyte transcription factors expressed during development. Overexpression of the BMP antagonist Noggin in hair follicles or deletion of the BMP receptor in myofibroblasts prevented adipocyte formation. Adipocytes formed from human keloid fibroblasts either when treated with BMP or when placed with human hair follicles in vitro. Thus, we identify the myofibroblast as a plastic cell type that may be manipulated to treat scars in humans.


Asunto(s)
Adipocitos/fisiología , Reprogramación Celular , Miofibroblastos/fisiología , Regeneración , Cicatrización de Heridas , Animales , Proteína Morfogenética Ósea 2/farmacología , Proteína Morfogenética Ósea 4/farmacología , Proteínas Morfogenéticas Óseas/metabolismo , Células Cultivadas , Cicatriz/patología , Proteínas de Unión al ADN/metabolismo , Fibroblastos/patología , Folículo Piloso/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/efectos de los fármacos , Proteínas Recombinantes/farmacología , Transducción de Señal , Factores de Transcripción/metabolismo
6.
Science ; 353(6295): 179-84, 2016 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-27365313

RESUMEN

Ideally, therapy for autoimmune diseases should eliminate pathogenic autoimmune cells while sparing protective immunity, but feasible strategies for such an approach have been elusive. Here, we show that in the antibody-mediated autoimmune disease pemphigus vulgaris (PV), autoantigen-based chimeric immunoreceptors can direct T cells to kill autoreactive B lymphocytes through the specificity of the B cell receptor (BCR). We engineered human T cells to express a chimeric autoantibody receptor (CAAR), consisting of the PV autoantigen, desmoglein (Dsg) 3, fused to CD137-CD3ζ signaling domains. Dsg3 CAAR-T cells exhibit specific cytotoxicity against cells expressing anti-Dsg3 BCRs in vitro and expand, persist, and specifically eliminate Dsg3-specific B cells in vivo. CAAR-T cells may provide an effective and universal strategy for specific targeting of autoreactive B cells in antibody-mediated autoimmune disease.


Asunto(s)
Autoanticuerpos/inmunología , Desmogleína 3/inmunología , Terapia de Inmunosupresión/métodos , Pénfigo/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/inmunología , Animales , Autoanticuerpos/sangre , Autoinmunidad , Linfocitos B/inmunología , Complejo CD3/química , Complejo CD3/inmunología , Ingeniería Celular , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Memoria Inmunológica , Ratones , Pénfigo/inmunología , Estructura Terciaria de Proteína , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/química , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
7.
Sci Transl Med ; 7(275): 275ra22, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25696001

RESUMEN

Chimeric antigen receptors (CARs) are synthetic molecules designed to redirect T cells to specific antigens. CAR-modified T cells can mediate long-term durable remissions in B cell malignancies, but expanding this platform to solid tumors requires the discovery of surface targets with limited expression in normal tissues. The variant III mutation of the epidermal growth factor receptor (EGFRvIII) results from an in-frame deletion of a portion of the extracellular domain, creating a neoepitope. We chose a vector backbone encoding a second-generation CAR based on efficacy of a murine scFv-based CAR in a xenograft model of glioblastoma. Next, we generated a panel of humanized scFvs and tested their specificity and function as soluble proteins and in the form of CAR-transduced T cells; a low-affinity scFv was selected on the basis of its specificity for EGFRvIII over wild-type EGFR. The lead candidate scFv was tested in vitro for its ability to direct CAR-transduced T cells to specifically lyse, proliferate, and secrete cytokines in response to antigen-bearing targets. We further evaluated the specificity of the lead CAR candidate in vitro against EGFR-expressing keratinocytes and in vivo in a model of mice grafted with normal human skin. EGFRvIII-directed CAR T cells were also able to control tumor growth in xenogeneic subcutaneous and orthotopic models of human EGFRvIII(+) glioblastoma. On the basis of these results, we have designed a phase 1 clinical study of CAR T cells transduced with humanized scFv directed to EGFRvIII in patients with either residual or recurrent glioblastoma (NCT02209376).


Asunto(s)
Neoplasias Encefálicas/terapia , Receptores ErbB/inmunología , Glioblastoma/terapia , Inmunoterapia , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones
8.
Nat Commun ; 5: 3071, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24468981

RESUMEN

Epithelial stem cells (EpSCs) in the hair follicle bulge are required for hair follicle growth and cycling. The isolation and propagation of human EpSCs for tissue engineering purposes remains a challenge. Here we develop a strategy to differentiate human iPSCs (hiPSCs) into CD200(+)/ITGA6(+) EpSCs that can reconstitute the epithelial components of the hair follicle and interfollicular epidermis. The hiPSC-derived CD200(+)/ITGA6(+) cells show a similar gene expression signature as EpSCs directly isolated from human hair follicles. Human iPSC-derived CD200(+)/ITGA6(+) cells are capable of generating all hair follicle lineages including the hair shaft, and the inner and outer root sheaths in skin reconstitution assays. The regenerated hair follicles possess a KRT15(+) stem cell population and produce hair shafts expressing hair-specific keratins. These results suggest an approach for generating large numbers of human EpSCs for tissue engineering and new treatments for hair loss, wound healing and other degenerative skin disorders.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/citología , Folículo Piloso/citología , Células Madre Pluripotentes/citología , Antígenos CD/metabolismo , Células Cultivadas , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/fisiología , Folículo Piloso/fisiología , Humanos , Técnicas In Vitro , Integrina alfa6/metabolismo , Células Madre Pluripotentes/fisiología , Piel/citología , Piel/fisiopatología , Ingeniería de Tejidos/métodos
9.
Nat Commun ; 5: 5807, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25510211

RESUMEN

Direct reprogramming provides a fundamentally new approach for the generation of patient-specific cells. Here, by screening a pool of candidate transcription factors, we identify that a combination of the three factors, MITF, SOX10 and PAX3, directly converts mouse and human fibroblasts to functional melanocytes. Induced melanocytes (iMels) activate melanocyte-specific networks, express components of pigment production and delivery system and produce melanosomes. Human iMels properly integrate into the dermal-epidermal junction and produce and deliver melanin pigment to surrounding keratinocytes in a 3D organotypic skin reconstruct. Human iMels generate pigmented epidermis and hair follicles in skin reconstitution assays in vivo. The generation of iMels has important implications for studies of melanocyte lineage commitment, pigmentation disorders and cell replacement therapies.


Asunto(s)
Reprogramación Celular/genética , Fibroblastos/metabolismo , Melaninas/biosíntesis , Melanocitos/metabolismo , Transgenes , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Dermis/citología , Dermis/metabolismo , Fibroblastos/citología , Expresión Génica , Vectores Genéticos , Folículo Piloso/citología , Folículo Piloso/metabolismo , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Melaninas/genética , Melanocitos/citología , Melanosomas/genética , Melanosomas/metabolismo , Ratones , Ratones Transgénicos , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Cultivo Primario de Células , Retroviridae/genética , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Transfección/métodos
10.
Nat Med ; 19(7): 916-23, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23727932

RESUMEN

Understanding molecular mechanisms for regeneration of hair follicles provides new opportunities for developing treatments for hair loss and other skin disorders. Here we show that fibroblast growth factor 9 (Fgf9), initially secreted by γδ T cells, modulates hair follicle regeneration after wounding the skin of adult mice. Reducing Fgf9 expression decreases this wound-induced hair neogenesis (WIHN). Conversely, overexpression of Fgf9 results in a two- to threefold increase in the number of neogenic hair follicles. We found that Fgf9 from γδ T cells triggers Wnt expression and subsequent Wnt activation in wound fibroblasts. Through a unique feedback mechanism, activated fibroblasts then express Fgf9, thus amplifying Wnt activity throughout the wound dermis during a crucial phase of skin regeneration. Notably, humans lack a robust population of resident dermal γδ T cells, potentially explaining their inability to regenerate hair after wounding. These findings highlight the essential relationship between the immune system and tissue regeneration. The importance of Fgf9 in hair follicle regeneration suggests that it could be used therapeutically in humans.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos/metabolismo , Factor 9 de Crecimiento de Fibroblastos/farmacología , Folículo Piloso/efectos de los fármacos , Linfocitos T/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Animales , Células Cultivadas , Dermis/citología , Dermis/inmunología , Dermis/metabolismo , Dermis/fisiología , Retroalimentación Fisiológica/efectos de los fármacos , Folículo Piloso/crecimiento & desarrollo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Regeneración/efectos de los fármacos , Regeneración/fisiología , Linfocitos T/fisiología , Proteínas Wnt/metabolismo , Cicatrización de Heridas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA