Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Immunity ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38772365

RESUMO

Inflammatory epithelial diseases are spurred by the concomitant dysregulation of immune and epithelial cells. How these two dysregulated cellular compartments simultaneously sustain their heightened metabolic demands is unclear. Single-cell and spatial transcriptomics (ST), along with immunofluorescence, revealed that hypoxia-inducible factor 1α (HIF1α), downstream of IL-17 signaling, drove psoriatic epithelial remodeling. Blocking HIF1α in human psoriatic lesions ex vivo impaired glycolysis and phenocopied anti-IL-17 therapy. In a murine model of skin inflammation, epidermal-specific loss of HIF1α or its target gene, glucose transporter 1, ameliorated epidermal, immune, vascular, and neuronal pathology. Mechanistically, glycolysis autonomously fueled epithelial pathology and enhanced lactate production, which augmented the γδ T17 cell response. RORγt-driven genetic deletion or pharmacological inhibition of either lactate-producing enzymes or lactate transporters attenuated epithelial pathology and IL-17A expression in vivo. Our findings identify a metabolic hierarchy between epithelial and immune compartments and the consequent coordination of metabolic processes that sustain inflammatory disease.

2.
Cell ; 167(5): 1323-1338.e14, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27863246

RESUMO

Aged skin heals wounds poorly, increasing susceptibility to infections. Restoring homeostasis after wounding requires the coordinated actions of epidermal and immune cells. Here we find that both intrinsic defects and communication with immune cells are impaired in aged keratinocytes, diminishing their efficiency in restoring the skin barrier after wounding. At the wound-edge, aged keratinocytes display reduced proliferation and migration. They also exhibit a dampened ability to transcriptionally activate epithelial-immune crosstalk regulators, including a failure to properly activate/maintain dendritic epithelial T cells (DETCs), which promote re-epithelialization following injury. Probing mechanism, we find that aged keratinocytes near the wound edge don't efficiently upregulate Skints or activate STAT3. Notably, when epidermal Stat3, Skints, or DETCs are silenced in young skin, re-epithelialization following wounding is perturbed. These findings underscore epithelial-immune crosstalk perturbations in general, and Skints in particular, as critical mediators in the age-related decline in wound-repair.


Assuntos
Envelhecimento/fisiologia , Subpopulações de Linfócitos/citologia , Transdução de Sinais , Cicatrização , Animais , Interleucina-6/administração & dosagem , Queratinócitos/metabolismo , Camundongos , Pele/citologia , Fenômenos Fisiológicos da Pele , Cicatrização/efeitos dos fármacos
3.
Cell ; 150(1): 136-50, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22770217

RESUMO

Sweat glands are abundant in the body and essential for thermoregulation. Like mammary glands, they originate from epidermal progenitors. However, they display few signs of cellular turnover, and whether they have stem cells and tissue-regenerative capacity remains largely unexplored. Using lineage tracing, we here identify in sweat ducts multipotent progenitors that transition to unipotency after developing the sweat gland. In characterizing four adult stem cell populations of glandular skin, we show that they display distinct regenerative capabilities and remain unipotent when healing epidermal, myoepithelial-specific, and lumenal-specific injuries. We devise purification schemes and isolate and transcriptionally profile progenitors. Exploiting molecular differences between sweat and mammary glands, we show that only some progenitors regain multipotency to produce de novo ductal and glandular structures, but that these can retain their identity even within certain foreign microenvironments. Our findings provide insight into glandular stem cells and a framework for the further study of sweat gland biology.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Homeostase , Glândulas Sudoríparas/citologia , Cicatrização , Células-Tronco Adultas/classificação , Animais , Células Epidérmicas , Epiderme/fisiologia , Feminino , Humanos , Glândulas Mamárias Animais/citologia , Camundongos , Morfogênese , Células-Tronco Multipotentes/fisiologia , Análise de Componente Principal , Transplante de Células-Tronco , Glândulas Sudoríparas/embriologia , Glândulas Sudoríparas/fisiologia
4.
Cell ; 144(3): 341-52, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21295697

RESUMO

Homeostasis and wound healing rely on stem cells (SCs) whose activity and directed migration are often governed by Wnt signaling. In dissecting how this pathway integrates with the necessary downstream cytoskeletal dynamics, we discovered that GSK3ß, a kinase inhibited by Wnt signaling, directly phosphorylates ACF7, a > 500 kDa microtubule-actin crosslinking protein abundant in hair follicle stem cells (HF-SCs). We map ACF7's GSK3ß sites to the microtubule-binding domain and show that phosphorylation uncouples ACF7 from microtubules. Phosphorylation-refractile ACF7 rescues overall microtubule architecture, but phosphorylation-constitutive mutants do not. Neither mutant rescues polarized movement, revealing that phospho-regulation must be dynamic. This circuitry is physiologically relevant and depends upon polarized GSK3ß inhibition at the migrating front of SCs/progeny streaming from HFs during wound repair. Moreover, only ACF7 and not GSKß-refractile-ACF7 restore polarized microtubule-growth and SC-migration to ACF7 null skin. Our findings provide insights into how this conserved spectraplakin integrates signaling, cytoskeletal dynamics, and polarized locomotion of somatic SCs.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas dos Microfilamentos/metabolismo , Microtúbulos/metabolismo , Pele/metabolismo , Células-Tronco/metabolismo , Cicatrização , Animais , Movimento Celular , Células Cultivadas , Glicogênio Sintase Quinase 3 beta , Camundongos , Camundongos Transgênicos , Fosforilação , Estrutura Terciária de Proteína , Pele/citologia , Células-Tronco/citologia
5.
Am J Clin Dermatol ; 25(3): 473-484, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38337127

RESUMO

BACKGROUND: Hidradenitis suppurativa (HS) is a debilitating cutaneous disease characterized by severe painful inflammatory nodules/abscesses. At present, data regarding the epidemiology and pathophysiology of this disease are limited. OBJECTIVE: To define the prevalence and comorbidity associations of HS. METHODS: This was a cross-sectional study of EPICTM Cosmos© examining over 180 million US patients. Prevalences were calculated by demographic and odds ratios (OR) and identified comorbidity correlations. RESULTS: All examined metabolism-related, psychological, and autoimmune/autoinflammatory (AI) diseases correlated with HS. The strongest associations were with pyoderma gangrenosum [OR 26.56; confidence interval (CI): 24.98-28.23], Down syndrome (OR 11.31; CI 10.93-11.70), and polycystic ovarian syndrome (OR 11.24; CI 11.09-11.38). Novel AI associations were found between HS and lupus (OR 6.60; CI 6.26-6.94) and multiple sclerosis (MS; OR 2.38; CI 2.29-2.48). Cutaneous malignancies were largely not associated in the unsegmented cohort; however, among Black patients, novel associations with melanoma (OR 2.39; CI 1.86-3.08) and basal cell carcinoma (OR 2.69; CI 2.15-3.36) were identified. LIMITATIONS: International Classification of Diseases (ICD)-based disease identification relies on coding fidelity and diagnostic accuracy. CONCLUSION: This is the first study to identify correlations between HS with melanoma and basal cell carcinoma (BCC) among Black patients as well as MS and lupus in all patients with HS.


Assuntos
Doenças Autoimunes , Comorbidade , Hidradenite Supurativa , Neoplasias Cutâneas , Humanos , Hidradenite Supurativa/epidemiologia , Hidradenite Supurativa/imunologia , Hidradenite Supurativa/complicações , Estudos Transversais , Feminino , Masculino , Prevalência , Neoplasias Cutâneas/epidemiologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Adulto , Doenças Autoimunes/epidemiologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/complicações , Pessoa de Meia-Idade , Estados Unidos/epidemiologia , Adulto Jovem , Idoso , Pioderma Gangrenoso/epidemiologia , Adolescente
6.
bioRxiv ; 2023 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-37745514

RESUMO

Sympathetic nerves co-develop with their target organs and release neurotransmitters to stimulate their functions after maturation. Here, we provide the molecular mechanism that during sweat gland morphogenesis, neurotransmitters released from sympathetic nerves act first to promote sweat duct elongation via norepinephrine and followed by acetylcholine to specify sweat gland stem cell fate, which matches the sequence of neurotransmitter switch. Without neuronal signals during development, the basal cells switch to exhibit suprabasal (luminal) cell features. Sarcolipin (SLN), a key regulator of sarcoendoplasmic reticulum (SR) Ca 2+ -ATPase (SERCA), expression is significantly down-regulated in the sweat gland myoepithelial cells upon denervation. Loss of SLN in sweat gland myoepithelial cells leads to decreased intracellular Ca 2+ over time in response to ACh stimulation, as well as upregulation of luminal cell features. In cell culture experiments, we showed that contrary to the paradigm that elevation of Ca 2+ promote epidermal differentiation, specification of the glandular myoepithelial (basal) cells requires high Ca 2+ while lowering Ca 2+ level promotes luminal (suprabasal) cell fate. Our work highlights that neuronal signals not only act transiently for mature sweat glands to function, but also exert long-term impact on glandular stem cell specification through regulating intracellular Ca 2+ dynamics.

7.
Transplant Rev (Orlando) ; 37(4): 100790, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37625211

RESUMO

OBJECTIVE: Rejection is common and pernicious following Vascularized Composite Allotransplantation (VCA). Current monitoring and diagnostic modalities include the clinical exam which is subjective and biopsy with dermatohistopathologic Banff grading, which is subjective and invasive. We reviewed literature exploring non- and minimally invasive modalities for diagnosing and monitoring rejection (NIMMs) in VCA. METHODS: PubMed, Cochrane, and Embase databases were queried, 3125 unique articles were reviewed, yielding 26 included studies exploring 17 distinct NIMMs. Broadly, NIMMs involved Imaging, Liquid Biomarkers, Epidermal Sampling, Clinical Grading Scales, and Introduction of Additional Donor Tissue. RESULTS: Serum biomarkers including MMP3 and donor-derived microparticles rose with rejection onset. Epidermal sampling non-invasively enabled measurement of cytokine & gene expression profiles implicated in rejection. Both hold promise for monitoring. Clinical grading scales were useful diagnostically as was reflection confocal microscopy. Introducing additional donor tissue showed promise for preemptively identifying rejection but requires additional allograft tissue burden for the recipient. CONCLUSION: NIMMs have the potential to dramatically improve monitoring and diagnosis in VCA. Many modalities show promise however, additional research is needed and a multimodal algorithmic approach should be explored.


Assuntos
Rejeição de Enxerto , Alotransplante de Tecidos Compostos Vascularizados , Humanos , Rejeição de Enxerto/diagnóstico , Alotransplante de Tecidos Compostos Vascularizados/métodos , Transplante Homólogo , Biomarcadores
8.
Nat Struct Mol Biol ; 13(11): 1010-5, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17028591

RESUMO

The Rag proteins carry out V(D)J recombination through a process mechanistically similar to cut-and-paste transposition. Specifically, Rag complexes form DNA hairpins through direct transesterification, using a catalytic Asp-Asp-Glu (DDE) triad in Rag1. How is sufficient DNA distortion introduced to allow hairpin formation? We hypothesized that, like certain transposases, the Rag proteins might use aromatic amino acid residues to stabilize a flipped-out base. Through in vivo and in vitro experiments and structural predictions, we identified residues in Rag1 crucial for hairpin formation. One of these, a conserved tryptophan (Trp893), probably participates in base-stacking interactions near the cleavage site, as do Trp298, Trp265 and Trp319 in the Tn5, Tn10 and Hermes transposases, respectively. Other residues surrounding the catalytic glutamate (YKEFRK) may share functional similarities with the YREK motif in IS4 family transposases.


Assuntos
Aminoácidos Aromáticos/metabolismo , DNA/química , DNA/metabolismo , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Sequência de Aminoácidos , Aminoácidos Aromáticos/análise , Aminoácidos Aromáticos/genética , Animais , Células CHO , Domínio Catalítico , Sequência Conservada , Cricetinae , Proteínas de Homeodomínio/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Alinhamento de Sequência , Relação Estrutura-Atividade , Transposases/química , Transposases/metabolismo , VDJ Recombinases/química , VDJ Recombinases/genética , VDJ Recombinases/metabolismo
9.
J Immunol ; 181(6): 4124-30, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768869

RESUMO

Hypomorphic RAG mutants with severely reduced V(D)J recombination activity cause Omenn Syndrome (OS), an immunodeficiency with features of immune dysregulation and a restricted TCR repertoire. Precisely how RAG mutants produce autoimmune and allergic symptoms has been unclear. Current models posit that the severe recombination defect restricts the number of lymphocyte clones, a few of which are selected upon Ag exposure. We show that murine RAG1 R972Q, corresponding to an OS mutation, renders the recombinase hypersensitive to selected coding sequences at the hairpin formation step. Other RAG1 OS mutants tested do not manifest this sequence sensitivity. These new data support a novel mechanism for OS: by selectively impairing recombination at certain coding flanks, a RAG mutant can cause primary repertoire restriction, as opposed to a more random, limited repertoire that develops secondary to severely diminished recombination activity.


Assuntos
Proteínas de Homeodomínio/genética , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Motivos de Aminoácidos/genética , Substituição de Aminoácidos/genética , Substituição de Aminoácidos/imunologia , Animais , Arginina/genética , Células CHO , Domínio Catalítico/genética , Cricetinae , Cricetulus , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T/genética , Glutamina/genética , Proteínas de Homeodomínio/fisiologia , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Imunodeficiência Combinada Severa/enzimologia , VDJ Recombinases/genética , VDJ Recombinases/fisiologia
10.
Nucleic Acids Res ; 36(9): 2864-73, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18375979

RESUMO

The Rag1 and Rag2 proteins initiate V(D)J recombination by introducing site-specific DNA double-strand breaks. Cleavage occurs by nicking one DNA strand, followed by a one-step transesterification reaction that forms a DNA hairpin structure. A similar reaction allows Rag transposition, in which the 3'-OH groups produced by Rag cleavage are joined to target DNA. The Rag1 active site DDE triad clearly plays a catalytic role in both cleavage and transposition, but no other residues in Rag1 responsible for transesterification have been identified. Furthermore, although Rag2 is essential for both cleavage and transposition, the nature of its involvement is unknown. Here, we identify basic amino acids in the catalytic core of Rag1 specifically important for transesterification. We also show that some Rag1 mutants with severe defects in hairpin formation nonetheless catalyze substantial levels of transposition. Lastly, we show that a catalytically defective Rag2 mutant is impaired in target capture and displays a novel form of coding flank sensitivity. These findings provide the first identification of components of Rag1 that are specifically required for transesterification and suggest an unexpected role for Rag2 in DNA cleavage and transposition.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Rearranjo Gênico , Proteínas de Homeodomínio/metabolismo , VDJ Recombinases/metabolismo , Animais , Células CHO , Catálise , Cricetinae , Cricetulus , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/genética , Esterificação , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Camundongos , Mutação
11.
Nat Cell Biol ; 22(6): 640-650, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32393888

RESUMO

Tissue homeostasis and regeneration rely on resident stem cells (SCs), whose behaviour is regulated through niche-dependent crosstalk. The mechanisms underlying SC identity are still unfolding. Here, using spatiotemporal gene ablation in murine hair follicles, we uncover a critical role for the transcription factors (TFs) nuclear factor IB (NFIB) and IX (NFIX) in maintaining SC identity. Without NFI TFs, SCs lose their hair-regenerating capability, and produce skin bearing striking resemblance to irreversible human alopecia, which also displays reduced NFIs. Through single-cell transcriptomics, ATAC-Seq and ChIP-Seq profiling, we expose a key role for NFIB and NFIX in governing super-enhancer maintenance of the key hair follicle SC-specific TF genes. When NFIB and NFIX are genetically removed, the stemness epigenetic landscape is lost. Super-enhancers driving SC identity are decommissioned, while unwanted lineages are de-repressed ectopically. Together, our findings expose NFIB and NFIX as crucial rheostats of tissue homeostasis, functioning to safeguard the SC epigenome from a breach in lineage confinement that otherwise triggers irreversible tissue degeneration.


Assuntos
Alopecia/patologia , Diferenciação Celular , Cromatina/metabolismo , Folículo Piloso/citologia , Fatores de Transcrição NFI/fisiologia , Células-Tronco/citologia , Alopecia/genética , Alopecia/metabolismo , Animais , Células Cultivadas , Cromatina/genética , Feminino , Folículo Piloso/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regeneração , Células-Tronco/metabolismo
12.
Elife ; 82019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30794158

RESUMO

Touch sensation is initiated by mechanosensory neurons that innervate distinct skin structures; however, little is known about how these neurons are patterned during mammalian skin development. We explored the cellular basis of touch-receptor patterning in mouse touch domes, which contain mechanosensory Merkel cell-neurite complexes and abut primary hair follicles. At embryonic stage 16.5 (E16.5), touch domes emerge as patches of Merkel cells and keratinocytes clustered with a previously unsuspected population of Bmp4-expressing dermal cells. Epidermal Noggin overexpression at E14.5 disrupted touch-dome formation but not hair-follicle specification, demonstrating a temporally distinct requirement for BMP signaling in placode-derived structures. Surprisingly, two neuronal populations preferentially targeted touch domes during development but only one persisted in mature touch domes. Finally, Keratin-17-expressing keratinocytes but not Merkel cells were necessary to establish innervation patterns during development. These findings identify key cell types and signaling pathways required for targeting Merkel-cell afferents to discrete mechanosensory compartments.


Assuntos
Padronização Corporal , Células de Merkel/fisiologia , Nervos Periféricos/embriologia , Pele/embriologia , Animais , Proteína Morfogenética Óssea 4/análise , Células Epidérmicas/fisiologia , Queratinócitos/fisiologia , Queratinas/análise , Camundongos
13.
Science ; 354(6319)2016 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-28008008

RESUMO

The gain of eccrine sweat glands in hairy body skin has empowered humans to run marathons and tolerate temperature extremes. Epithelial-mesenchymal cross-talk is integral to the diverse patterning of skin appendages, but the molecular events underlying their specification remain largely unknown. Using genome-wide analyses and functional studies, we show that sweat glands are specified by mesenchymal-derived bone morphogenetic proteins (BMPs) and fibroblast growth factors that signal to epithelial buds and suppress epithelial-derived sonic hedgehog (SHH) production. Conversely, hair follicles are specified when mesenchymal BMP signaling is blocked, permitting SHH production. Fate determination is confined to a critical developmental window and is regionally specified in mice. In contrast, a shift from hair to gland fates is achieved in humans when a spike in BMP silences SHH during the final embryonic wave(s) of bud morphogenesis.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Glândulas Écrinas/embriologia , Folículo Piloso/embriologia , Proteínas Hedgehog/metabolismo , Mesoderma/embriologia , Suor , Animais , Glândulas Écrinas/metabolismo , Epitélio/embriologia , Epitélio/metabolismo , Humanos , Mesoderma/metabolismo , Camundongos , Morfogênese , Transdução de Sinais , Via de Sinalização Wnt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA