Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
2.
Technol Cult ; 65(1): 143-175, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38661797

RESUMEN

Archives and oral histories show that the Ming Tombs Reservoir was a showcase project in Communist China directed by and involving the country's top leaders. This was one of the first projects to rely on the mobilization of physical labor rather than specialized machinery, driven by a belief in self-reliance and the use of local resources. It argues that the focus on the "masses," rather than engineers or scientists, challenged established engineering procedures and technical traditions. Historical evidence suggests that adopting a "build while being designed" mindset and mobilizing the "masses," projects could be completed, but often in ways that ultimately proved less than optimal. The case study suggests that innovations fail when local enthusiasm and technical knowledge are not balanced. By focusing on the role of the "masses" in shaping a novel technological landscape, this article highlights "mass engineering" to better understand this model of native innovations and economic autarky.


Asunto(s)
Ingeniería , China , Humanos , Historia del Siglo XX , Ingeniería/historia , Comunismo/historia , Política
3.
Protein Cell ; 14(3): 159-161, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37063413
4.
Genes Dev ; 2022 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-36008138

RESUMEN

Stem cells are fundamental units of tissue remodeling whose functions are dictated by lineage-specific transcription factors. Home to epidermal stem cells and their upward-stratifying progenies, skin relies on its secretory functions to form the outermost protective barrier, of which a transcriptional orchestrator has been elusive. KLF5 is a Krüppel-like transcription factor broadly involved in development and regeneration whose lineage specificity, if any, remains unclear. Here we report KLF5 specifically marks the epidermis, and its deletion leads to skin barrier dysfunction in vivo. Lipid envelopes and secretory lamellar bodies are defective in KLF5-deficient skin, accompanied by preferential loss of complex sphingolipids. KLF5 binds to and transcriptionally regulates genes encoding rate-limiting sphingolipid metabolism enzymes. Remarkably, skin barrier defects elicited by KLF5 ablation can be rescued by dietary interventions. Finally, we found that KLF5 is widely suppressed in human diseases with disrupted epidermal secretion, and its regulation of sphingolipid metabolism is conserved in human skin. Altogether, we established KLF5 as a disease-relevant transcription factor governing sphingolipid metabolism and barrier function in the skin, likely representing a long-sought secretory lineage-defining factor across tissue types.

5.
Front Cell Dev Biol ; 10: 903904, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35663405

RESUMEN

Skin is the largest organ in human body, harboring a plethora of cell types and serving as the organismal barrier. Skin aging such as wrinkling and hair graying is graphically pronounced, and the molecular mechanisms behind these phenotypic manifestations are beginning to unfold. As in many other organs and tissues, epigenetic and metabolic deregulations have emerged as key aging drivers. Particularly in the context of the skin epithelium, the epigenome and metabolome coordinately shape lineage plasticity and orchestrate stem cell function during aging. Our review discusses recent studies that proposed molecular mechanisms that drive the degeneration of hair follicles, a major appendage of the skin. By focusing on skin while comparing it to model organisms and adult stem cells of other tissues, we summarize literature on genotoxic stress, nutritional sensing, metabolic rewiring, mitochondrial activity, and epigenetic regulations of stem cell plasticity. Finally, we speculate about the rejuvenation potential of rate-limiting upstream signals during aging and the dominant role of the tissue microenvironment in dictating aged epithelial stem cell function.

6.
EMBO J ; 41(7): e109470, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35212000

RESUMEN

Skin wound repair is essential for organismal survival and failure of which leads to non-healing wounds, a leading health issue worldwide. However, mechanistic understanding of chronic wounds remains a major challenge due to lack of appropriate genetic mouse models. αSMA+ myofibroblasts, a unique class of dermal fibroblasts, are associated with cutaneous wound healing but their precise function remains unknown. We demonstrate that genetic depletion of αSMA+ myofibroblasts leads to pleiotropic wound healing defects, including lack of reepithelialization and granulation, dampened angiogenesis, and heightened hypoxia, hallmarks of chronic non-healing wounds. Other wound-associated FAP+ and FSP1+ fibroblasts do not exhibit such dominant functions. While type I collagen (COL1) expressing cells play a role in the repair process, COL1 produced by αSMA+ myofibroblasts is surprisingly dispensable for wound repair. In contrast, we show that ß1 integrin from αSMA+ myofibroblasts, but not TGFßRII, is essential for wound healing, facilitating contractility, reepithelization, and vascularization. Collectively, our study provides evidence for the functions of myofibroblasts in ß1 integrin-mediated wound repair with potential implications for treating chronic non-healing wounds.


Asunto(s)
Colágeno Tipo I , Miofibroblastos , Cicatrización de Heridas , Animales , Colágeno Tipo I/genética , Fibroblastos , Integrina beta1/genética , Ratones , Piel
7.
Protein Cell ; 13(5): 309-312, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33665762
8.
Exp Dermatol ; 30(4): 529-545, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33249665

RESUMEN

The epidermis and skin appendages are maintained by their resident epithelial stem cells, which undergo long-term self-renewal and multilineage differentiation. Upon injury, stem cells are activated to mediate re-epithelialization and restore tissue function. During this process, they often mount lineage plasticity and expand their fates in response to damage signals. Stem cell function is tightly controlled by transcription machineries and signalling transductions, many of which derail in degenerative, inflammatory and malignant dermatologic diseases. Here, by describing both well-characterized and newly emerged pathways, we discuss the transcriptional and signalling mechanisms governing skin epithelial homeostasis, wound repair and squamous cancer. Throughout, we highlight common themes underscoring epithelial stem cell plasticity and tissue-level crosstalk in the context of skin physiology and pathology.


Asunto(s)
Células Epiteliales/metabolismo , Homeostasis , Neoplasias Cutáneas/fisiopatología , Piel/metabolismo , Células Madre/metabolismo , Cicatrización de Heridas/fisiología , Animales , Diferenciación Celular , Microambiente Celular , Folículo Piloso/metabolismo , Humanos , Ratones , Transducción de Señal
9.
Nat Cell Biol ; 22(11): 1396, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33046885

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Nat Cell Biol ; 22(6): 640-650, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32393888

RESUMEN

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.


Asunto(s)
Alopecia/patología , Diferenciación Celular , Cromatina/metabolismo , Folículo Piloso/citología , Factores de Transcripción NFI/fisiología , Células Madre/citología , Alopecia/genética , Alopecia/metabolismo , Animales , Células Cultivadas , Cromatina/genética , Femenino , Folículo Piloso/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regeneración , Células Madre/metabolismo
11.
Nat Cell Biol ; 22(7): 779-790, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32451440

RESUMEN

Tissue stem cells are the cell of origin for many malignancies. Metabolites regulate the balance between self-renewal and differentiation, but whether endogenous metabolic pathways or nutrient availability predispose stem cells towards transformation remains unknown. Here, we address this question in epidermal stem cells (EpdSCs), which are a cell of origin for squamous cell carcinoma. We find that oncogenic EpdSCs are serine auxotrophs whose growth and self-renewal require abundant exogenous serine. When extracellular serine is limited, EpdSCs activate de novo serine synthesis, which in turn stimulates α-ketoglutarate-dependent dioxygenases that remove the repressive histone modification H3K27me3 and activate differentiation programmes. Accordingly, serine starvation or enforced α-ketoglutarate production antagonizes squamous cell carcinoma growth. Conversely, blocking serine synthesis or repressing α-ketoglutarate-driven demethylation facilitates malignant progression. Together, these findings reveal that extracellular serine is a critical determinant of EpdSC fate and provide insight into how nutrient availability is integrated with stem cell fate decisions during tumour initiation.


Asunto(s)
Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica/patología , Células Epidérmicas/patología , Ácidos Cetoglutáricos/metabolismo , Serina/metabolismo , Células Madre/patología , Animales , Carcinoma de Células Escamosas/metabolismo , Diferenciación Celular , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Células Epidérmicas/metabolismo , Femenino , Humanos , Masculino , Ratones , Células Madre/metabolismo
12.
Proc Natl Acad Sci U S A ; 117(10): 5339-5350, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32094197

RESUMEN

Aging manifests with architectural alteration and functional decline of multiple organs throughout an organism. In mammals, aged skin is accompanied by a marked reduction in hair cycling and appearance of bald patches, leading researchers to propose that hair follicle stem cells (HFSCs) are either lost, differentiate, or change to an epidermal fate during aging. Here, we employed single-cell RNA-sequencing to interrogate aging-related changes in the HFSCs. Surprisingly, although numbers declined, aging HFSCs were present, maintained their identity, and showed no overt signs of shifting to an epidermal fate. However, they did exhibit prevalent transcriptional changes particularly in extracellular matrix genes, and this was accompanied by profound structural perturbations in the aging SC niche. Moreover, marked age-related changes occurred in many nonepithelial cell types, including resident immune cells, sensory neurons, and arrector pili muscles. Each of these SC niche components has been shown to influence HF regeneration. When we performed skin injuries that are known to mobilize young HFSCs to exit their niche and regenerate HFs, we discovered that aged skin is defective at doing so. Interestingly, however, in transplantation assays in vivo, aged HFSCs regenerated HFs when supported with young dermis, while young HFSCs failed to regenerate HFs when combined with aged dermis. Together, our findings highlight the importance of SC:niche interactions and favor a model where youthfulness of the niche microenvironment plays a dominant role in dictating the properties of its SCs and tissue health and fitness.


Asunto(s)
Folículo Piloso/fisiología , Regeneración/fisiología , Envejecimiento de la Piel/fisiología , Nicho de Células Madre/fisiología , Células Madre/fisiología , Animales , Dermis/fisiología , Células Epidérmicas/fisiología , Epidermis/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculos/fisiología , Repitelización , Regeneración/genética , Células Receptoras Sensoriales/fisiología , Envejecimiento de la Piel/genética , Nicho de Células Madre/genética , Trasplante de Células Madre , Transcriptoma , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología
13.
Pharmacol Ther ; 206: 107448, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31836455

RESUMEN

Cancer hijacks embryonic development and adult wound repair mechanisms to fuel malignancy. Cancer frequently originates from de-regulated adult stem cells or progenitors, which are otherwise essential units for postnatal tissue remodeling and repair. Cancer genomics studies have revealed convergence of multiple cancers across organ sites, including squamous cell carcinomas (SCCs), a common group of cancers arising from the head and neck, esophagus, lung, cervix and skin. In this review, we summarize our current knowledge on the molecular drivers of SCCs, including these five major organ sites. We especially focus our discussion on lineage dependent driver genes and pathways, in the context of squamous development and stratification. We then use skin as a model to discuss the notion of field cancerization during SCC carcinogenesis, and cancer as a wound that never heals. Finally, we turn to the idea of context dependency widely observed in cancer driver genes, and outline literature support and possible explanations for their lineage specific functions. Through these discussions, we aim to provide an up-to-date summary of molecular mechanisms driving tumor plasticity in squamous cancers. Such basic knowledge will be helpful to inform the clinics for better stratifying cancer patients, revealing novel drug targets and providing effective treatment options.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias Cutáneas/genética , Animales , Genómica , Humanos
14.
Nature ; 569(7757): 497-502, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31092920

RESUMEN

Cell competition-the sensing and elimination of less fit 'loser' cells by neighbouring 'winner' cells-was first described in Drosophila. Although cell competition has been proposed as a selection mechanism to optimize tissue and organ development, its evolutionary generality remains unclear. Here, by using live imaging, lineage tracing, single-cell transcriptomics and genetics, we identify two cell competition mechanisms that sequentially shape and maintain the architecture of stratified tissue during skin development in mice. In the single-layered epithelium of the early embryonic epidermis, winner progenitors kill and subsequently clear neighbouring loser cells by engulfment. Later, as the tissue begins to stratify, the basal layer instead expels losers through upward flux of differentiating progeny. This cell competition switch is physiologically relevant: when it is perturbed, so too is barrier formation. Our findings show that cell competition is a selective force that optimizes vertebrate tissue function, and illuminate how a tissue dynamically adjusts cell competition strategies to preserve fitness as its architectural complexity increases during morphogenesis.


Asunto(s)
Comunicación Celular , Células Epidérmicas/citología , Epidermis/embriología , Morfogénesis , Animales , Apoptosis , Células Clonales/citología , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Células Epidérmicas/metabolismo , Femenino , Masculino , Ratones , Fagocitosis , RNA-Seq , Análisis de la Célula Individual
15.
Nat Rev Genet ; 19(5): 311-325, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29479084

RESUMEN

Stem cells (SCs) govern tissue homeostasis and wound repair. They reside within niches, the special microenvironments within tissues that control SC lineage outputs. Upon injury or stress, new signals emanating from damaged tissue can divert nearby cells into adopting behaviours that are not part of their homeostatic repertoire. This behaviour, known as SC plasticity, typically resolves as wounds heal. However, in cancer, it can endure. Recent studies have yielded insights into the orchestrators of maintenance and lineage commitment for SCs belonging to three mammalian tissues: the haematopoietic system, the skin epithelium and the intestinal epithelium. We delineate the multifactorial determinants and general principles underlying the remarkable facets of SC plasticity, which lend promise for regenerative medicine and cancer therapeutics.


Asunto(s)
Plasticidad de la Célula , Neoplasias/metabolismo , Células Madre/metabolismo , Microambiente Tumoral , Cicatrización de Heridas , Animales , Humanos , Neoplasias/patología , Neoplasias/terapia , Células Madre/patología
16.
Cell Stem Cell ; 22(3): 398-413.e7, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29337183

RESUMEN

Tissue regeneration relies on resident stem cells (SCs), whose activity and lineage choices are influenced by the microenvironment. Exploiting the synchronized, cyclical bouts of tissue regeneration in hair follicles (HFs), we investigate how microenvironment dynamics shape the emergence of stem cell lineages. Employing epigenetic and ChIP-seq profiling, we uncover how signal-dependent transcription factors couple spatiotemporal cues to chromatin dynamics, thereby choreographing stem cell lineages. Using enhancer-driven reporters, mutagenesis, and genetics, we show that simultaneous BMP-inhibitory and WNT signals set the stage for lineage choices by establishing chromatin platforms permissive for diversification. Mechanistically, when binding of BMP effector pSMAD1 is relieved, enhancers driving HF-stem cell master regulators are silenced. Concomitantly, multipotent, lineage-fated enhancers silent in HF-stem cells become activated by exchanging WNT effectors TCF3/4 for LEF1. Throughout regeneration, lineage enhancers continue reliance upon LEF1 but then achieve specificity by accommodating additional incoming signaling effectors. Barriers to progenitor plasticity increase when diverse, signal-sensitive transcription factors shape LEF1-regulated enhancer dynamics.


Asunto(s)
Linaje de la Célula , Ensamble y Desensamble de Cromatina , Folículo Piloso/citología , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Acetilación , Animales , Secuencia de Bases , Proteínas Morfogenéticas Óseas/metabolismo , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Histonas/metabolismo , Lisina/metabolismo , Ratones Endogámicos C57BL , Fosforilación , Regeneración , Proteína Smad1/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Vía de Señalización Wnt
17.
Cell ; 169(4): 636-650.e14, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28434617

RESUMEN

Tissue stem cells contribute to tissue regeneration and wound repair through cellular programs that can be hijacked by cancer cells. Here, we investigate such a phenomenon in skin, where during homeostasis, stem cells of the epidermis and hair follicle fuel their respective tissues. We find that breakdown of stem cell lineage confinement-granting privileges associated with both fates-is not only hallmark but also functional in cancer development. We show that lineage plasticity is critical in wound repair, where it operates transiently to redirect fates. Investigating mechanism, we discover that irrespective of cellular origin, lineage infidelity occurs in wounding when stress-responsive enhancers become activated and override homeostatic enhancers that govern lineage specificity. In cancer, stress-responsive transcription factor levels rise, causing lineage commanders to reach excess. When lineage and stress factors collaborate, they activate oncogenic enhancers that distinguish cancers from wounds.


Asunto(s)
Carcinoma de Células Escamosas/patología , Linaje de la Célula , Células Epidérmicas , Folículo Piloso/citología , Neoplasias Cutáneas/patología , Piel/citología , Células Madre/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Epidermis/metabolismo , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Cutáneas/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Trasplante Heterólogo , Cicatrización de Heridas
18.
Cell ; 169(3): 483-496.e13, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28413068

RESUMEN

Adult tissue stem cells (SCs) reside in niches, which, through intercellular contacts and signaling, influence SC behavior. Once activated, SCs typically give rise to short-lived transit-amplifying cells (TACs), which then progress to differentiate into their lineages. Here, using single-cell RNA-seq, we unearth unexpected heterogeneity among SCs and TACs of hair follicles. We trace the roots of this heterogeneity to micro-niches along epithelial-mesenchymal interfaces, where progenitors display molecular signatures reflective of spatially distinct local signals and intercellular interactions. Using lineage tracing, temporal single-cell analyses, and chromatin landscaping, we show that SC plasticity becomes restricted in a sequentially and spatially choreographed program, culminating in seven spatially arranged unilineage progenitors within TACs of mature follicles. By compartmentalizing SCs into micro-niches, tissues gain precise control over morphogenesis and regeneration: some progenitors specify lineages immediately, whereas others retain potency, preserving self-renewing features established early while progressively restricting lineages as they experience dynamic changes in microenvironment.


Asunto(s)
Células Madre Adultas/citología , Linaje de la Célula , Folículo Piloso/citología , Nicho de Células Madre , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Vía de Señalización Wnt
19.
Nat Cell Biol ; 18(1): 111-21, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26619149

RESUMEN

MicroRNAs play diverse roles in both normal and malignant stem cells. Focusing on miRs and/or miR(∗)s abundant in squamous cell carcinoma (SCC) stem cells, we engineer an efficient, strand-specific expression library, and apply functional genomics screening in mice to identify which of 169 cancer-associated miRs are key drivers in malignant progression. Not previously linked functionally to cancer, miR-21(∗) was the second top hit, surfacing in >12% of tumours. miR-21(∗) also correlates with poor prognosis in human SCCs and enhances tumour progression in xenografts. On deleting the miR-21 gene and rescuing each strand separately, we document the dual, but independent, oncogenicity of miR-21 and miR-21(∗). A cohort of predicted miR-21(∗) targets inversely correlate with miR-21(∗) in SCCs. Of particular interest is Phactr4, which we show is a miR-21(∗) target in SCCs, acting through the Rb/E2F cell cycle axis. Through in vivo physiological miR screens, our findings add an interesting twist to an increasingly important oncomiR locus.


Asunto(s)
Carcinoma de Células Escamosas/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , Animales , Ciclo Celular/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Ratones
20.
Genes Dev ; 28(22): 2532-46, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25403182

RESUMEN

Previously, we identified miR-125b as a key regulator of the undifferentiated state of hair follicle stem cells. Here, we show that in both mice and humans, miR-125b is abundantly expressed, particularly at early stages of malignant progression to squamous cell carcinoma (SCC), the second most prevalent cancer worldwide. Moreover, when elevated in normal murine epidermis, miR-125b promotes tumor initiation and contributes to malignant progression. We further show that miR-125b can confer "oncomiR addiction" in early stage malignant progenitors by delaying their differentiation and favoring an SCC cancer stem cell (CSC)-like transcriptional program. To understand how, we systematically identified and validated miR125b targets that are specifically associated with tumors that are dependent on miR-125b. Through molecular and genetic analysis of these targets, we uncovered new insights underlying miR-125b's oncogenic function. Specifically, we show that, on the one hand, mir-125b directly represses stress-responsive MAP kinase genes and associated signaling. On the other hand, it indirectly prolongs activated (phosphorylated) EGFR signaling by repressing Vps4b (vacuolar protein-sorting 4 homolog B), encoding a protein implicated in negatively regulating the endosomal sorting complexes that are necessary for the recycling of active EGFR. Together, these findings illuminate miR-125b as an important microRNA regulator that is shared between normal skin progenitors and their early malignant counterparts.


Asunto(s)
Carcinoma de Células Escamosas/fisiopatología , Diferenciación Celular/genética , MicroARNs/metabolismo , Neoplasias Cutáneas/fisiopatología , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/metabolismo , Animales , Carcinoma de Células Escamosas/genética , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Progresión de la Enfermedad , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Queratinocitos/citología , Queratinocitos/patología , Ratones , MicroARNs/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal , Neoplasias Cutáneas/genética , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA