Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.562
Filtrar
Mais filtros

Eixos temáticos
Intervalo de ano de publicação
1.
Cell ; 182(3): 578-593.e19, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32679029

RESUMO

Piloerection (goosebumps) requires concerted actions of the hair follicle, the arrector pili muscle (APM), and the sympathetic nerve, providing a model to study interactions across epithelium, mesenchyme, and nerves. Here, we show that APMs and sympathetic nerves form a dual-component niche to modulate hair follicle stem cell (HFSC) activity. Sympathetic nerves form synapse-like structures with HFSCs and regulate HFSCs through norepinephrine, whereas APMs maintain sympathetic innervation to HFSCs. Without norepinephrine signaling, HFSCs enter deep quiescence by down-regulating the cell cycle and metabolism while up-regulating quiescence regulators Foxp1 and Fgf18. During development, HFSC progeny secretes Sonic Hedgehog (SHH) to direct the formation of this APM-sympathetic nerve niche, which in turn controls hair follicle regeneration in adults. Our results reveal a reciprocal interdependence between a regenerative tissue and its niche at different stages and demonstrate sympathetic nerves can modulate stem cells through synapse-like connections and neurotransmitters to couple tissue production with demands.


Assuntos
Nervo Acessório/fisiologia , Folículo Piloso/citologia , Cabelo/crescimento & desenvolvimento , Proteínas Hedgehog/metabolismo , Norepinefrina/metabolismo , Transdução de Sinais/genética , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Nervo Acessório/citologia , Animais , Ciclo Celular/genética , Temperatura Baixa , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Cabelo/citologia , Cabelo/fisiologia , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piloereção , RNA-Seq , Receptores Adrenérgicos beta 2/deficiência , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Nicho de Células-Tronco , Células-Tronco/citologia , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/fisiologia , Sinapses/fisiologia
2.
Immunity ; 57(5): 1071-1086.e7, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38677291

RESUMO

Following tissue damage, epithelial stem cells (SCs) are mobilized to enter the wound, where they confront harsh inflammatory environments that can impede their ability to repair the injury. Here, we investigated the mechanisms that protect skin SCs within this inflammatory environment. Characterization of gene expression profiles of hair follicle SCs (HFSCs) that migrated into the wound site revealed activation of an immune-modulatory program, including expression of CD80, major histocompatibility complex class II (MHCII), and CXC motif chemokine ligand 5 (CXCL5). Deletion of CD80 in HFSCs impaired re-epithelialization, reduced accumulation of peripherally generated Treg (pTreg) cells, and increased infiltration of neutrophils in wounded skin. Importantly, similar wound healing defects were also observed in mice lacking pTreg cells. Our findings suggest that upon skin injury, HFSCs establish a temporary protective network by promoting local expansion of Treg cells, thereby enabling re-epithelialization while still kindling inflammation outside this niche until the barrier is restored.


Assuntos
Antígeno B7-1 , Folículo Piloso , Inflamação , Pele , Células-Tronco , Linfócitos T Reguladores , Cicatrização , Animais , Linfócitos T Reguladores/imunologia , Camundongos , Cicatrização/imunologia , Pele/imunologia , Pele/lesões , Pele/patologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Inflamação/imunologia , Folículo Piloso/imunologia , Antígeno B7-1/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reepitelização/imunologia , Movimento Celular/imunologia , Proliferação de Células
3.
Cell ; 169(6): 1119-1129.e11, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28552347

RESUMO

The maintenance of tissue homeostasis is critically dependent on the function of tissue-resident immune cells and the differentiation capacity of tissue-resident stem cells (SCs). How immune cells influence the function of SCs is largely unknown. Regulatory T cells (Tregs) in skin preferentially localize to hair follicles (HFs), which house a major subset of skin SCs (HFSCs). Here, we mechanistically dissect the role of Tregs in HF and HFSC biology. Lineage-specific cell depletion revealed that Tregs promote HF regeneration by augmenting HFSC proliferation and differentiation. Transcriptional and phenotypic profiling of Tregs and HFSCs revealed that skin-resident Tregs preferentially express high levels of the Notch ligand family member, Jagged 1 (Jag1). Expression of Jag1 on Tregs facilitated HFSC function and efficient HF regeneration. Taken together, our work demonstrates that Tregs in skin play a major role in HF biology by promoting the function of HFSCs.


Assuntos
Folículo Piloso/citologia , Células-Tronco/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Células Epiteliais/metabolismo , Folículo Piloso/metabolismo , Humanos , Inflamação/metabolismo , Proteína Jagged-1/metabolismo , Camundongos
4.
Immunity ; 55(10): 1891-1908.e12, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36044899

RESUMO

Demodex mites are commensal parasites of hair follicles (HFs). Normally asymptomatic, inflammatory outgrowth of mites can accompany malnutrition, immune dysfunction, and aging, but mechanisms restricting Demodex outgrowth are not defined. Here, we show that control of mite HF colonization in mice required group 2 innate lymphoid cells (ILC2s), interleukin-13 (IL-13), and its receptor, IL-4Ra-IL-13Ra1. HF-associated ILC2s elaborated IL-13 that attenuated HFs and epithelial proliferation at anagen onset; in their absence, Demodex colonization led to increased epithelial proliferation and replacement of gene programs for repair by aberrant inflammation, leading to the loss of barrier function and HF exhaustion. Humans with rhinophymatous acne rosacea, an inflammatory condition associated with Demodex, had increased HF inflammation with decreased type 2 cytokines, consistent with the inverse relationship seen in mice. Our studies uncover a key role for skin ILC2s and IL-13, which comprise an immune checkpoint that sustains cutaneous integrity and restricts pathologic infestation by colonizing HF mites.


Assuntos
Infestações por Ácaros , Ácaros , Animais , Citocinas , Folículo Piloso/patologia , Humanos , Imunidade Inata , Inflamação , Interleucina-13 , Linfócitos/patologia , Camundongos , Infestações por Ácaros/complicações , Infestações por Ácaros/parasitologia , Infestações por Ácaros/patologia , Simbiose
5.
Immunity ; 50(3): 655-667.e4, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30893588

RESUMO

Restoration of barrier-tissue integrity after injury is dependent on the function of immune cells and stem cells (SCs) residing in the tissue. In response to skin injury, hair-follicle stem cells (HFSCs), normally poised for hair generation, are recruited to the site of injury and differentiate into cells that repair damaged epithelium. We used a SC fate-mapping approach to examine the contribution of regulatory T (Treg) cells to epidermal-barrier repair after injury. Depletion of Treg cells impaired skin-barrier regeneration and was associated with a Th17 inflammatory response and failed HFSC differentiation. In this setting, damaged epithelial cells preferentially expressed the neutrophil chemoattractant CXCL5, and blockade of CXCL5 or neutrophil depletion restored barrier function and SC differentiation after epidermal injury. Thus, Treg-cell regulation of localized inflammation enables HFSC differentiation and, thereby, skin-barrier regeneration, with implications for the maintenance and repair of other barrier tissues.


Assuntos
Diferenciação Celular/fisiologia , Quimiocina CXCL5/metabolismo , Epiderme/metabolismo , Folículo Piloso/metabolismo , Interleucina-17/metabolismo , Regeneração/fisiologia , Linfócitos T Reguladores/metabolismo , Animais , Células Epidérmicas/metabolismo , Células Epiteliais/metabolismo , Cabelo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/metabolismo
6.
EMBO J ; 42(10): e112196, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36994549

RESUMO

Blood vessels can play dual roles in tissue growth by transporting gases and nutrients and by regulating tissue stem cell activity via signaling. Correlative evidence implicates skin endothelial cells (ECs) as signaling niches of hair follicle stem cells (HFSCs), but functional demonstration from gene depletion of signaling molecules in ECs is missing to date. Here, we show that depletion of the vasculature-factor Alk1 increases BMP4 secretion from ECs, which delays HFSC activation. Furthermore, while previous evidence suggests a lymphatic vessel role in adult HFSC activation possibly through tissue drainage, a blood vessel role has not yet been addressed. Genetic perturbation of the ALK1-BMP4 axis in all ECs or the lymphatic ECs specifically unveils inhibition of HFSC activation by blood vessels. Our work suggests a broader relevance of blood vessels, adding adult HFSCs to the EC functional repertoire as signaling niches for the adult stem cells.


Assuntos
Receptores de Activinas Tipo II , Células-Tronco Adultas , Proteína Morfogenética Óssea 4 , Folículo Piloso , Animais , Camundongos , Células Endoteliais , Transdução de Sinais , Células-Tronco , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo
7.
Immunity ; 48(2): 271-285.e5, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29466757

RESUMO

Stem cells are critical for the maintenance of many tissues, but whether their integrity is maintained in the face of immunity is unclear. Here we found that cycling epithelial stem cells, including Lgr5+ intestinal stem cells, as well as ovary and mammary stem cells, were eliminated by activated T cells, but quiescent stem cells in the hair follicle and muscle were resistant to T cell killing. Immune evasion was an intrinsic property of the quiescent stem cells resulting from systemic downregulation of the antigen presentation machinery, including MHC class I and TAP proteins, and is mediated by the transactivator NLRC5. This process was reversed upon stem cell entry into the cell cycle. These studies identify a link between stem cell quiescence, antigen presentation, and immune evasion. As cancer-initiating cells can derive from stem cells, these findings may help explain how the earliest cancer cells evade immune surveillance.


Assuntos
Folículo Piloso/citologia , Evasão da Resposta Imune , Vigilância Imunológica , Células-Tronco/imunologia , Animais , Apresentação de Antígeno , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Músculos/citologia , Receptores Acoplados a Proteínas G/fisiologia , Evasão Tumoral
8.
Development ; 150(22)2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37982496

RESUMO

Tissue interactions are essential for guiding organ development and regeneration. Hair follicle formation relies on inductive signalling between two tissues, the embryonic surface epithelium and the adjacent mesenchyme. Although previous research has highlighted the hair-inducing potential of the mesenchymal component of the hair follicle - the dermal papilla and its precursor, the dermal condensate - the source and nature of the primary inductive signal before dermal condensate formation have remained elusive. Here, we performed epithelial-mesenchymal tissue recombination experiments using hair-forming back skin and glabrous plantar skin from mouse embryos to unveil that the back skin mesenchyme is inductive even before dermal condensate formation. Moreover, the naïve, unpatterned mesenchyme was sufficient to trigger hair follicle formation even in the oral epithelium. Building on previous knowledge, we explored the hair-inductive ability of the Wnt agonist R-spondin 1 and a Bmp receptor inhibitor in embryonic skin explants. Although R-spondin 1 instigated precocious placode-specific transcriptional responses, it was insufficient for hair follicle induction, either alone or in combination with Bmp receptor inhibition. Our findings pave the way for identifying the hair follicle-inducing cue.


Assuntos
Folículo Piloso , Cabelo , Camundongos , Animais , Folículo Piloso/fisiologia , Pele , Mesoderma/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas
9.
Development ; 150(17)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37622728

RESUMO

The planar cell polarity (PCP) pathway collectively orients cells with respect to a body axis. Hair follicles of the murine epidermis provide a striking readout of PCP activity in their uniform alignment across the skin. Here, we characterize, from the molecular to tissue-scale, PCP establishment in the rosette fancy mouse, a natural variant with posterior-specific whorls in its fur, to understand how epidermal polarity is coordinated across the tissue. We find that rosette hair follicles emerge with reversed orientations specifically in the posterior region, creating a mirror image of epidermal polarity. The rosette trait is associated with a missense mutation in the core PCP gene Fzd6, which alters a consensus site for N-linked glycosylation, inhibiting its membrane localization. Unexpectedly, the Fzd6 trafficking defect does not block asymmetric localization of the other PCP proteins. Rather, the normally uniform axis of PCP asymmetry rotates where the PCP-directed cell movements that orient follicles are reversed, suggesting the PCP axis rotates 180°. Collectively, our multiscale analysis of epidermal polarity reveals PCP patterning can be regionally decoupled to produce posterior whorls in the rosette fancy mouse.


Assuntos
Epiderme , Folículo Piloso , Animais , Camundongos , Pele , Células Epidérmicas , Movimento Celular
10.
Mol Cell ; 70(4): 573-587.e4, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29775577

RESUMO

Apoptosis culminates in the activation of caspase-3, which plays an important role in implementing the cell death program. Here, we reveal a non-apoptotic role of caspase-3 as a key regulator of cell proliferation and organ size. Caspase-3 is specifically activated in the proliferating cells of the sebaceous gland, but does not instruct cell elimination. Deletion or chemical inhibition of caspase-3 diminishes cell proliferation, decreases cell number and reduces sebaceous gland size in vivo. Exploring the underlying mechanism, we demonstrate that α-catenin is cleaved by caspase-3, thus facilitating the activation and nuclear translocation of yes-associated protein (YAP), a vital regulator of organ size. Accordingly, activation of caspase-3 leads to YAP-dependent organ size augmentation. Finally, we show that X-linked inhibitor of apoptosis protein (XIAP) serves as an endogenous feedback antagonist for the caspase-3/YAP signaling module. Taken together, we report here a molecular mechanism wherein the apoptotic machinery is refocused to regulate cell proliferation and orchestrate organ size.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caspase 3/fisiologia , Proliferação de Células , Retroalimentação Fisiológica , Proteínas Inibidoras de Apoptose/fisiologia , Fosfoproteínas/metabolismo , Fatores de Processamento de RNA/fisiologia , alfa Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Proteínas de Ciclo Celular , Feminino , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão , Fosfoproteínas/genética , Transporte Proteico , Proteínas de Sinalização YAP , alfa Catenina/genética
11.
Proc Natl Acad Sci U S A ; 120(29): e2305764120, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37428932

RESUMO

Alopecia areata (AA) is among the most prevalent autoimmune diseases, but the development of innovative therapeutic strategies has lagged due to an incomplete understanding of the immunological underpinnings of disease. Here, we performed single-cell RNA sequencing (scRNAseq) of skin-infiltrating immune cells from the graft-induced C3H/HeJ mouse model of AA, coupled with antibody-based depletion to interrogate the functional role of specific cell types in AA in vivo. Since AA is predominantly T cell-mediated, we focused on dissecting lymphocyte function in AA. Both our scRNAseq and functional studies established CD8+ T cells as the primary disease-driving cell type in AA. Only the depletion of CD8+ T cells, but not CD4+ T cells, NK, B, or γδ T cells, was sufficient to prevent and reverse AA. Selective depletion of regulatory T cells (Treg) showed that Treg are protective against AA in C3H/HeJ mice, suggesting that failure of Treg-mediated immunosuppression is not a major disease mechanism in AA. Focused analyses of CD8+ T cells revealed five subsets, whose heterogeneity is defined by an "effectorness gradient" of interrelated transcriptional states that culminate in increased effector function and tissue residency. scRNAseq of human AA skin showed that CD8+ T cells in human AA follow a similar trajectory, underscoring that shared mechanisms drive disease in both murine and human AA. Our study represents a comprehensive, systematic interrogation of lymphocyte heterogeneity in AA and uncovers a novel framework for AA-associated CD8+ T cells with implications for the design of future therapeutics.


Assuntos
Alopecia em Áreas , Camundongos , Humanos , Animais , Alopecia em Áreas/genética , Alopecia em Áreas/tratamento farmacológico , Camundongos Endogâmicos C3H , Subpopulações de Linfócitos , Análise de Sequência de RNA
12.
Proc Natl Acad Sci U S A ; 120(36): e2221982120, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37643215

RESUMO

Stem cells in organoids self-organize into tissue patterns with unknown mechanisms. Here, we use skin organoids to analyze this process. Cell behavior videos show that the morphological transformation from multiple spheroidal units with morphogenesis competence (CMU) to planar skin is characterized by two abrupt cell motility-increasing events before calming down. The self-organizing processes are controlled by a morphogenetic module composed of molecular sensors, modulators, and executers. Increasing dermal stiffness provides the initial driving force (driver) which activates Yap1 (sensor) in epidermal cysts. Notch signaling (modulator 1) in epidermal cyst tunes the threshold of Yap1 activation. Activated Yap1 induces Wnts and MMPs (epidermal executers) in basal cells to facilitate cellular flows, allowing epidermal cells to protrude out from the CMU. Dermal cell-expressed Rock (dermal executer) generates a stiff force bridge between two CMU and accelerates tissue mixing via activating Laminin and ß1-integrin. Thus, this self-organizing coalescence process is controlled by a mechano-chemical circuit. Beyond skin, self-organization in organoids may use similar mechano-chemical circuit structures.


Assuntos
Epiderme , Pele , Personalidade , Organoides , Emoções , Proteínas Adaptadoras de Transdução de Sinal
13.
Proc Natl Acad Sci U S A ; 120(22): e2220635120, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37216502

RESUMO

Stiffness and actomyosin contractility are intrinsic mechanical properties of animal cells required for the shaping of tissues. However, whether tissue stem cells (SCs) and progenitors located within SC niche have different mechanical properties that modulate their size and function remains unclear. Here, we show that hair follicle SCs in the bulge are stiff with high actomyosin contractility and resistant to size change, whereas hair germ (HG) progenitors are soft and periodically enlarge and contract during quiescence. During activation of hair follicle growth, HGs reduce contraction and more frequently enlarge, a process that is associated with weakening of the actomyosin network, nuclear YAP accumulation, and cell cycle reentry. Induction of miR-205, a novel regulator of the actomyosin cytoskeleton, reduces actomyosin contractility and activates hair regeneration in young and old mice. This study reveals the control of tissue SC size and activities by spatiotemporally compartmentalized mechanical properties and demonstrates the possibility to stimulate tissue regeneration by fine-tuning cell mechanics.


Assuntos
Folículo Piloso , MicroRNAs , Animais , Camundongos , Actomiosina/metabolismo , Cabelo , Folículo Piloso/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco/metabolismo
14.
Dev Biol ; 516: 20-34, 2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39059679

RESUMO

Ectodermal organs, such as hair follicles, originate from simple epithelial and mesenchymal sheets through a complex developmental process driven by interactions between these cell types. This process involves dermal condensation, placode formation, bud morphogenesis, and organogenesis, and all of these processes require intricate interactions among various tissues. Recent research has emphasized the crucial role of reciprocal and dynamic interactions between cells and the extracellular matrix (ECM), referred to as the "dynamic duo", in the development of ectodermal organs. These interactions provide spatially and temporally changing biophysical and biochemical cues within tissues. Using the hair follicle as an example, this review highlights two types of cell-ECM adhesion units-focal adhesion-type and hemidesmosome-type adhesion units-that facilitate communication between epithelial and mesenchymal cells. This review further explores how these adhesion units, along with other cell-ECM interactions, evolve during hair follicle development and regeneration, underscoring their importance in guiding both developmental and regenerative processes.

15.
EMBO J ; 40(11): e107135, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33880808

RESUMO

Stem cells are the essential source of building blocks for tissue homeostasis and regeneration. Their behavior is dictated by both cell-intrinsic cues and extrinsic cues from the microenvironment, known as the stem cell niche. Interestingly, recent work began to demonstrate that hair follicle stem cells (HFSCs) are not only passive recipients of signals from the surroundings, but also actively send out signals to modulate the organization and function of their own niches. Here, we discuss recent findings, and briefly refer to the old, on the interaction of HFSCs and their niches with the emphasis on the outwards signals from HFSCs toward their niches. We also highlight recent technology advancements that further promote our understanding of HFSC niches. Taken together, the HFSCs emerge as a skin-organizing center rich in signaling output for niche remodeling during various stages of adult skin homeostasis. The intricate crosstalk between HFSCs and their niches adds important insight to skin biology that will inform clinical and bioengineering fields aiming to build complete and functional 3D organotypic cultures for skin replacement therapies.


Assuntos
Células-Tronco Adultas/metabolismo , Folículo Piloso/citologia , Transdução de Sinais , Células-Tronco Adultas/citologia , Animais , Comunicação Celular , Folículo Piloso/metabolismo , Homeostase , Humanos , Nicho de Células-Tronco
16.
FASEB J ; 38(4): e23476, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38334392

RESUMO

The prevalence of alopecia has increased recently. Hair loss is often accompanied by the resting phase of hair follicles (HFs). Dermal papilla (DP) plays a crucial role in HF development, growth, and regeneration. Activating DP can revive resting HFs. Augmenting WNT/ß-catenin signaling stimulates HF growth. However, the factors responsible for activating resting HFs effectively are unclear. In this study, we investigated epidermal cytokines that can activate resting HFs effectively. We overexpressed ß-catenin in both in vivo and in vitro models to observe its effects on resting HFs. Then, we screened potential epidermal cytokines from GEO DATASETs and assessed their functions using mice models and skin-derived precursors (SKPs). Finally, we explored the molecular mechanism underlying the action of the identified cytokine. The results showed that activation of WNT/ß-catenin in the epidermis prompted telogen-anagen transition. Keratinocytes infected with Ctnnb1-overexpressing lentivirus enhanced SKP expansion. Subsequently, we identified endothelin 1 (ET-1) expressed higher in hair-growing epidermis and induced the proliferation of DP cells and activates telogen-phase HFs in vivo. Moreover, ET-1 promotes the proliferation and stemness of SKPs. Western blot analysis and in vivo experiments revealed that ET-1 induces the transition from telogen-to-anagen phase by upregulating the PI3K/AKT pathway. These findings highlight the potential of ET-1 as a promising cytokine for HF activation and the treatment of hair loss.


Assuntos
Folículo Piloso , Proteínas Proto-Oncogênicas c-akt , Animais , Camundongos , Folículo Piloso/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Endotelina-1/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Células Cultivadas , Proliferação de Células , Epiderme/metabolismo , Alopecia/metabolismo , Via de Sinalização Wnt , Derme/metabolismo , Citocinas/metabolismo
17.
Exp Cell Res ; 438(1): 114049, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38642790

RESUMO

BACKGROUND: Acellular nerve allografts (ANAs) have been successfully applied to bridge facial nerve defects, and transplantation of stem cells may enhance the regenerative results. Up to now, application of hair follicle epidermal neural crest stem cell-derived Schwann cell-like cells (EPI-NCSC-SCLCs) combined with ANAs for bridging facial nerve defects has not been reported. METHODS: The effect of ANAs laden with green fluorescent protein (GFP)-labeled EPI-NCSC-SCLCs (ANA + cells) on bridging rat facial nerve trunk defects (5-mm-long) was detected by functional and morphological examination, as compared with autografts and ANAs, respectively. RESULTS: (1) EPI-NCSC-SCLCs had good compatibility with ANAs in vitro. (2) In the ANA + cells group, the GFP signals were observed by in vivo imaging system for small animals within 8 weeks, and GFP-labeled EPI-NCSC-SCLCs were detected in the tissue slices at 16 weeks postoperatively. (3) The facial symmetry at rest after surgery in the ANA + cells group was better than that in the ANA group (p < 0.05), and similar to that in the autograft group (p > 0.05). The initial recovery time of vibrissal and eyelid movement in the ANA group was 2 weeks later than that in the other two groups. (4) The myelinated fibers, myelin sheath thickness and diameter of the axons of the buccal branches in the ANA group were significantly worse than those in the other two groups (P < 0.05), and the results in the ANA + cells group were similar to those in the autograft group (p > 0.05). CONCLUSIONS: EPI-NCSC-SCLCs could promote functional and morphological recovery of rat facial nerve defects, and GFP labeling could track the transplanted EPI-NCSC-SCLCs in vivo for a certain period of time. These may provide a novel choice for clinical treatment of peripheral nerve defects.


Assuntos
Aloenxertos , Nervo Facial , Proteínas de Fluorescência Verde , Folículo Piloso , Regeneração Nervosa , Crista Neural , Células de Schwann , Animais , Células de Schwann/transplante , Folículo Piloso/transplante , Folículo Piloso/citologia , Crista Neural/citologia , Crista Neural/transplante , Ratos , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Fluorescência Verde/genética , Regeneração Nervosa/fisiologia , Células-Tronco Neurais/transplante , Células-Tronco Neurais/citologia , Ratos Sprague-Dawley , Traumatismos do Nervo Facial/terapia , Traumatismos do Nervo Facial/patologia , Traumatismos do Nervo Facial/cirurgia , Masculino
18.
Genomics ; 116(2): 110818, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38431032

RESUMO

Sheep breeds with hair-shedding traits have many advantages over non-shedding sheep breeds, not only because of reduced shearing labor and feeding management costs but also because it reduces in vitro parasites and improves adaptability to summer heat stress. The wool of Dorper sheep naturally sheds in spring due to the periodic growth of hair follicles. CircRNAs primarily regulate the morphogenesis of hair follicles through the ceRNA mechanism. In this study, five 2-year-old Dorper ewes with extreme hair-shedding phenotype (S) and three Dorper ewes with non-shedding (N) phenotype were selected for subsequent analyses. For RNA extraction, skin tissues were collected on 27th September 2019 (S1, N1), 3rd January 2020 (S2, N2), and 17th March 2020 (S3, N3), which were then subjected to RNA-seq. RNA-seq technology revealed 20,185 novel circRNAs in the hair follicles of Dorper sheep. Among them, 1450 circRNAs were differentially expressed (DE). Clustering heatmap and expression pattern analyses were performed on DE circRNAs, which indicated 78 circRNAs with T pattern (Telogen, highly expressed in telogen), and the source genes for candidate circRNAs were further screened by functional enrichment analysis, which identified 13 crucial genes enriched in pathways associated with hair follicle development. Additionally, a ceRNA regulatory network comprising 4 circRNAs, 11 miRNAs, and 13 target genes was constructed. Overall, this study screened circRNAs that may be associated with the telogen phase of hair follicles in sheep, providing a relevant theoretical basis for wool shedding in sheep and for breeding Dorper sheep with automatic wool shedding.


Assuntos
MicroRNAs , RNA Circular , Ovinos/genética , Animais , Feminino , RNA Circular/metabolismo , RNA Endógeno Competitivo , Folículo Piloso/metabolismo , Carneiro Doméstico/genética , MicroRNAs/metabolismo
19.
Genes Dev ; 31(8): 721-723, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28512235

RESUMO

Melanocytes present in hair follicles are responsible for their pigmentation. Melanocyte differentiation and hair pigmentation depend on the stem cell factor (SCF)/c-Kit signaling pathway, but the niche that regulates melanocyte differentiation is not well characterized. In this issue of Genes & Development, Liao and colleagues (pp. 744-756) identify Krox20+-derived cells of the hair shaft as the niche and the essential source of SCF required for melanocyte maturation. This study delineates the niche factors regulating melanocyte differentiation and hair pigmentation and opens up new avenues to further characterize the cross-talk between the hair follicle and melanocytes that controls melanocyte maintenance and differentiation.


Assuntos
Diferenciação Celular , Folículo Piloso/citologia , Melanócitos/citologia , Animais , Melanócitos/metabolismo , Pigmentação/genética , Pigmentação/fisiologia , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo
20.
Genes Dev ; 31(8): 744-756, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28465357

RESUMO

Hair differentiates from follicle stem cells through progenitor cells in the matrix. In contrast to stem cells in the bulge, the identities of the progenitors and the mechanisms by which they regulate hair shaft components are poorly understood. Hair is also pigmented by melanocytes in the follicle. However, the niche that regulates follicular melanocytes is not well characterized. Here, we report the identification of hair shaft progenitors in the matrix that are differentiated from follicular epithelial cells expressing transcription factor KROX20. Depletion of Krox20 lineage cells results in arrest of hair growth, confirming the critical role of KROX20+ cells as antecedents of structural cells found in hair. Expression of stem cell factor (SCF) by these cells is necessary for the maintenance of differentiated melanocytes and for hair pigmentation. Our findings reveal the identities of hair matrix progenitors that regulate hair growth and pigmentation, partly by creating an SCF-dependent niche for follicular melanocytes.


Assuntos
Cabelo/citologia , Pigmentação/fisiologia , Fator de Células-Tronco/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Cabelo/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Melaninas/metabolismo , Camundongos , Pigmentação/genética , Fator de Células-Tronco/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa