Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38858071

RESUMEN

How tissue architecture and function emerge during development and what facilitates their resilience and homeostatic dynamics during adulthood is a fundamental question in biology. Biological tissue barriers such as the skin epidermis have evolved strategies that integrate dynamic cellular turnover with high resilience against mechanical and chemical stresses. Interestingly, both dynamic and resilient functions are generated by a defined set of molecular and cell-scale processes, including adhesion and cytoskeletal remodeling, cell shape changes, cell division, and cell movement. These traits are coordinated in space and time with dynamic changes in cell fates and cell mechanics that are generated by contractile and adhesive forces. In this review, we discuss how studies on epidermal morphogenesis and homeostasis have contributed to our understanding of the dynamic interplay between biochemical and mechanical signals during tissue morphogenesis and homeostasis, and how the material properties of tissues dictate how cells respond to these active stresses, thereby linking cell-scale behaviors to tissue- and organismal-scale changes.

2.
Cell ; 187(12): 2898-2900, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38848672

RESUMEN

Epithelial folding is a fundamental biological process that requires epithelial interactions with the underlying mesenchyme. In this issue of Cell, Huycke et al. investigate intestinal villus formation. They discover that water-droplet-like behavior of mesenchymal cells drives their coalescence into uniformly patterned aggregates, which generate forces on the epithelium to initiate folding.


Asunto(s)
Epitelio , Mesodermo , Animales , Humanos , Células Epiteliales/metabolismo , Células Epiteliales/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/citología , Mesodermo/metabolismo , Mesodermo/citología , Epitelio/metabolismo
3.
Cell ; 187(11): 2652-2656, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38788688

RESUMEN

Mechanobiology-the field studying how cells produce, sense, and respond to mechanical forces-is pivotal in the analysis of how cells and tissues take shape in development and disease. As we venture into the future of this field, pioneers share their insights, shaping the trajectory of future research and applications.


Asunto(s)
Biofisica , Animales , Humanos , Fenómenos Biomecánicos , Forma de la Célula , Mecanotransducción Celular
4.
Artículo en Inglés | MEDLINE | ID: mdl-38600372

RESUMEN

From embryonic development, postnatal growth and adult homeostasis to reparative and disease states, cells and tissues undergo constant changes in genome activity, cell fate, proliferation, movement, metabolism and growth. Importantly, these biological state transitions are coupled to changes in the mechanical and material properties of cells and tissues, termed mechanical state transitions. These mechanical states share features with physical states of matter, liquids and solids. Tissues can switch between mechanical states by changing behavioural dynamics or connectivity between cells. Conversely, these changes in tissue mechanical properties are known to control cell and tissue function, most importantly the ability of cells to move or tissues to deform. Thus, tissue mechanical state transitions are implicated in transmitting information across biological length and time scales, especially during processes of early development, wound healing and diseases such as cancer. This Review will focus on the biological basis of tissue-scale mechanical state transitions, how they emerge from molecular and cellular interactions, and their roles in organismal development, homeostasis, regeneration and disease.

5.
Nat Cell Biol ; 26(2): 207-218, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38302719

RESUMEN

Morphogenesis and cell state transitions must be coordinated in time and space to produce a functional tissue. An excellent paradigm to understand the coupling of these processes is mammalian hair follicle development, which is initiated by the formation of an epithelial invagination-termed placode-that coincides with the emergence of a designated hair follicle stem cell population. The mechanisms directing the deformation of the epithelium, cell state transitions and physical compartmentalization of the placode are unknown. Here we identify a key role for coordinated mechanical forces stemming from contractile, proliferative and proteolytic activities across the epithelial and mesenchymal compartments in generating the placode structure. A ring of fibroblast cells gradually wraps around the placode cells to generate centripetal contractile forces, which, in collaboration with polarized epithelial myosin activity, promote elongation and local tissue thickening. These mechanical stresses further enhance compartmentalization of Sox9 expression to promote stem cell positioning. Subsequently, proteolytic remodelling locally softens the basement membrane to facilitate a release of pressure on the placode, enabling localized cell divisions, tissue fluidification and epithelial invagination into the underlying mesenchyme. Together, our experiments and modelling identify dynamic cell shape transformations and tissue-scale mechanical cooperation as key factors for orchestrating organ formation.


Asunto(s)
Folículo Piloso , Mamíferos , Animales , Forma de la Célula , Epitelio , Morfogénesis , División Celular , Folículo Piloso/metabolismo
6.
Nature ; 623(7988): 828-835, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37968399

RESUMEN

The skin epidermis is constantly renewed throughout life1,2. Disruption of the balance between renewal and differentiation can lead to uncontrolled growth and tumour initiation3. However, the ways in which oncogenic mutations affect the balance between renewal and differentiation and lead to clonal expansion, cell competition, tissue colonization and tumour development are unknown. Here, through multidisciplinary approaches that combine in vivo clonal analysis using intravital microscopy, single-cell analysis and functional analysis, we show how SmoM2-a constitutively active oncogenic mutant version of Smoothened (SMO) that induces the development of basal cell carcinoma-affects clonal competition and tumour initiation in real time. We found that expressing SmoM2 in the ear epidermis of mice induced clonal expansion together with tumour initiation and invasion. By contrast, expressing SmoM2 in the back-skin epidermis led to a clonal expansion that induced lateral cell competition without dermal invasion and tumour formation. Single-cell analysis showed that oncogene expression was associated with a cellular reprogramming of adult interfollicular cells into an embryonic hair follicle progenitor (EHFP) state in the ear but not in the back skin. Comparisons between the ear and the back skin revealed that the dermis has a very different composition in these two skin types, with increased stiffness and a denser collagen I network in the back skin. Decreasing the expression of collagen I in the back skin through treatment with collagenase, chronic UV exposure or natural ageing overcame the natural resistance of back-skin basal cells to undergoing EHFP reprogramming and tumour initiation after SmoM2 expression. Altogether, our study shows that the composition of the extracellular matrix regulates how susceptible different regions of the body are to tumour initiation and invasion.


Asunto(s)
Transformación Celular Neoplásica , Matriz Extracelular , Neoplasias Cutáneas , Microambiente Tumoral , Animales , Ratones , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Colágeno/metabolismo , Epidermis/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Neoplasias Cutáneas/patología , Carcinoma Basocelular/patología , Oído/patología , Colagenasas/metabolismo , Envejecimiento , Rayos Ultravioleta , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo
7.
Sci Rep ; 13(1): 7743, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37173371

RESUMEN

Epithelia maintain a functional barrier during tissue turnover while facing varying mechanical stress. This maintenance requires both dynamic cell rearrangements driven by actomyosin-linked intercellular adherens junctions and ability to adapt to and resist extrinsic mechanical forces enabled by keratin filament-linked desmosomes. How these two systems crosstalk to coordinate cellular movement and mechanical resilience is not known. Here we show that in stratifying epithelia the polarity protein aPKCλ controls the reorganization from stress fibers to cortical actomyosin during differentiation and upward movement of cells. Without aPKC, stress fibers are retained resulting in increased contractile prestress. This aberrant stress is counterbalanced by reorganization and bundling of keratins, thereby increasing mechanical resilience. Inhibiting contractility in aPKCλ-/- cells restores normal cortical keratin networks but also normalizes resilience. Consistently, increasing contractile stress is sufficient to induce keratin bundling and enhance resilience, mimicking aPKC loss. In conclusion, our data indicate that keratins sense the contractile stress state of stratified epithelia and balance increased contractility by mounting a protective response to maintain tissue integrity.


Asunto(s)
Actomiosina , Transducción de Señal , Actomiosina/metabolismo , Epitelio/metabolismo , Citoesqueleto/metabolismo , Queratinas/metabolismo , Células Epiteliales/metabolismo
8.
Trends Cell Biol ; 33(6): 477-494, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36270939

RESUMEN

The existence of stem cells (SCs) at the tip of the cellular differentiation hierarchy has fascinated the scientific community ever since their discovery in the early 1950s to 1960s. Despite the remarkable success of the SC theory and the development of SC-based treatments, fundamental features of SCs remain enigmatic. Recent advances in single-cell lineage tracing, live imaging, and genomic technologies have allowed capture of life histories and transcriptional signatures of individual cells, leaving SCs much less space to 'hide'. Focusing on epithelial SCs and comparing them to other SCs, we discuss new paradigms of the SC niche, dynamics, and pathology, highlighting key open questions in SC biology that need to be resolved for harnessing SC potential in regenerative medicine.


Asunto(s)
Células Madre Adultas , Humanos , Células Madre , Linaje de la Célula , Diferenciación Celular
9.
Cell ; 185(19): 3638-3638.e1, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36113430

RESUMEN

Cells are continuously exposed to tissue-specific extrinsic forces that are counteracted by cell-intrinsic force generation through the actomyosin cytoskeleton and alterations in the material properties of various cellular components, including the nucleus. Forces impact nuclei both directly through inducing deformation, which is sensed by various mechanosensitive components of the nucleus, as well as indirectly through the actomyosin cytoskeleton and mechanosensitive pathways activated in the cytoplasm. To view this SnapShot, open or download the PDF.


Asunto(s)
Actomiosina , Mecanotransducción Celular , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Mecanotransducción Celular/fisiología
10.
EMBO J ; 41(17): e111650, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35899396

RESUMEN

Mechanical inputs give rise to p38 and JNK activation, which mediate adaptive physiological responses in various tissues. In skeletal muscle, contraction-induced p38 and JNK signaling ensure adaptation to exercise, muscle repair, and hypertrophy. However, the mechanisms by which muscle fibers sense mechanical load to activate this signaling have remained elusive. Here, we show that the upstream MAP3K ZAKß is activated by cellular compression induced by osmotic shock and cyclic compression in vitro, and muscle contraction in vivo. This function relies on ZAKß's ability to recognize stress fibers in cells and Z-discs in muscle fibers when mechanically perturbed. Consequently, ZAK-deficient mice present with skeletal muscle defects characterized by fibers with centralized nuclei and progressive adaptation towards a slower myosin profile. Our results highlight how cells in general respond to mechanical compressive load and how mechanical forces generated during muscle contraction are translated into MAP kinase signaling.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos , Músculo Esquelético , Animales , Quinasas Quinasa Quinasa PAM , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Fosforilación , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/genética
12.
Nat Rev Genet ; 23(10): 624-643, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35606569

RESUMEN

Cells and tissues generate and are exposed to various mechanical forces that act across a range of scales, from tissues to cells to organelles. Forces provide crucial signals to inform cell behaviour during development and adult tissue homeostasis, and alterations in forces and in their downstream mechanotransduction pathways can influence disease progression. Recent advances have been made in our understanding of the mechanisms by which forces regulate chromatin organization and state, and of the mechanosensitive transcription factors that respond to the physical properties of the cell microenvironment to coordinate gene expression, cell states and behaviours. These insights highlight the relevance of mechanosensitive transcriptional regulation to physiology, disease and emerging therapies.


Asunto(s)
Cromatina , Mecanotransducción Celular , Cromatina/genética , Regulación de la Expresión Génica , Homeostasis , Mecanotransducción Celular/fisiología , Factores de Transcripción
13.
14.
Proc Natl Acad Sci U S A ; 119(11): e2106098119, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35259013

RESUMEN

SignificanceThe pseudokinase integrin-linked kinase (ILK) is a central component of focal adhesions, cytoplasmic multiprotein complexes that integrate and transduce biochemical and mechanical signals from the extracellular environment into the cell and vice versa. However, the precise molecular functions, particularly the mechanosensory properties of ILK and the significance of retained adenosine triphosphate (ATP) binding, are still unclear. Combining molecular-dynamics simulations with cell biology, we establish a role for ATP binding to pseudokinases. We find that ATP promotes the structural stability of ILK, allosterically influences the interaction between ILK and its binding partner parvin at adhesions, and enhances the mechanoresistance of this complex. On the cellular level, ATP binding facilitates efficient traction force buildup, focal adhesion stabilization, and efficient cell migration.


Asunto(s)
Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Actomiosina/química , Actomiosina/metabolismo , Regulación Alostérica , Sitios de Unión , Adhesión Celular , Movimiento Celular , Estabilidad de Enzimas , Adhesiones Focales , Mecanotransducción Celular , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Modelos Moleculares , Conformación Molecular , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Relación Estructura-Actividad , Especificidad por Sustrato
15.
Artículo en Inglés | MEDLINE | ID: mdl-34187806

RESUMEN

Cells generate and sense mechanical forces that trigger biochemical signals to elicit cellular responses that control cell fate changes. Mechanical forces also physically distort neighboring cells and the surrounding connective tissue, which propagate mechanochemical signals over long distances to guide tissue patterning, organogenesis, and adult tissue homeostasis. As the largest and stiffest organelle, the nucleus is particularly sensitive to mechanical force and deformation. Nuclear responses to mechanical force include adaptations in chromatin architecture and transcriptional activity that trigger changes in cell state. These force-driven changes also influence the mechanical properties of chromatin and nuclei themselves to prevent aberrant alterations in nuclear shape and help maintain genome integrity. This review will discuss principles of nuclear mechanotransduction and chromatin mechanics and their role in DNA damage and cell fate regulation.


Asunto(s)
Núcleo Celular , Mecanotransducción Celular , Diferenciación Celular , Cromatina , Genoma , Mecanotransducción Celular/fisiología
16.
Dev Cell ; 56(24): 3303-3304, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34932945

RESUMEN

Tissues need strategies to cope with genomic insults to maintain their integrity. In this issue of Developmental Cell, Kato et al. use in vivo fate tracing to observe selective elimination of epidermal stem cells (EpiSCs) harboring severe genomic lesions through their differentiation and compensatory expansion of surrounding intact cells.

17.
Nat Cell Biol ; 23(7): 771-781, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34239060

RESUMEN

Tissue turnover requires activation and lineage commitment of tissue-resident stem cells (SCs). These processes are impacted by ageing, but the mechanisms remain unclear. Here, we addressed the mechanisms of ageing in murine hair follicle SCs (HFSCs) and observed a widespread reduction in chromatin accessibility in aged HFSCs, particularly at key self-renewal and differentiation genes, characterized by bivalent promoters occupied by active and repressive chromatin marks. Consistent with this, aged HFSCs showed reduced ability to activate bivalent genes for efficient self-renewal and differentiation. These defects were niche dependent as the transplantation of aged HFSCs into young recipients or synthetic niches restored SC functions. Mechanistically, the aged HFSC niche displayed widespread alterations in extracellular matrix composition and mechanics, resulting in mechanical stress and concomitant transcriptional repression to silence promoters. As a consequence, increasing basement membrane stiffness recapitulated age-related SC changes. These data identify niche mechanics as a central regulator of chromatin state, which, when altered, leads to age-dependent SC exhaustion.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Senescencia Celular , Ensamble y Desensamble de Cromatina , Folículo Piloso/fisiología , Regiones Promotoras Genéticas , Nicho de Células Madre , Células Madre/fisiología , Animales , Diferenciación Celular/genética , Linaje de la Célula , Autorrenovación de las Células/genética , Células Cultivadas , Senescencia Celular/genética , Matriz Extracelular/fisiología , Silenciador del Gen , Folículo Piloso/citología , Folículo Piloso/metabolismo , Masculino , Mecanotransducción Celular , Ratones Endogámicos C57BL , Ratones Noqueados , Envejecimiento de la Piel , Células Madre/metabolismo , Estrés Mecánico , Transcripción Genética
18.
Mol Biol Cell ; 32(18): 1724-1736, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34081532

RESUMEN

The vascular system is precisely regulated to adjust blood flow to organismal demand, thereby guaranteeing adequate perfusion under varying physiological conditions. Mechanical forces, such as cyclic circumferential stretch, are among the critical stimuli that dynamically adjust vessel distribution and diameter, but the precise mechanisms of adaptation to changing forces are unclear. We find that endothelial monolayers respond to cyclic stretch by transient remodeling of the vascular endothelial cadherin-based adherens junctions and the associated actomyosin cytoskeleton. Time-resolved proteomic profiling reveals that this remodeling is driven by calcium influx through the mechanosensitive Piezo1 channel, triggering Rho activation to increase actomyosin contraction. As the mechanical stimulus persists, calcium signaling is attenuated through transient down-regulation of Piezo1 protein. At the same time, filamins are phosphorylated to increase monolayer stiffness, allowing mechanoadaptation to restore junctional integrity despite continuing exposure to stretch. Collectively, this study identifies a biphasic response to cyclic stretch, consisting of an initial calcium-driven junctional mechanoresponse, followed by mechanoadaptation facilitated by monolayer stiffening.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actomiosina , Antígenos CD/metabolismo , Cadherinas/metabolismo , Señalización del Calcio , Mecanotransducción Celular , Actomiosina/metabolismo , Uniones Adherentes/fisiología , Antígenos CD/genética , Fenómenos Biomecánicos , Cadherinas/genética , Calcimicina/farmacología , Ionóforos de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Citocalasina D/farmacología , Filaminas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Fosfoproteínas/análisis , Fosfoproteínas/metabolismo , Mapas de Interacción de Proteínas , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
19.
J Cell Biol ; 220(8)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34042944

RESUMEN

The mammary gland develops from the surface ectoderm during embryogenesis and proceeds through morphological phases defined as placode, hillock, bud, and bulb stages followed by branching morphogenesis. During this early morphogenesis, the mammary bud undergoes an invagination process where the thickened bud initially protrudes above the surface epithelium and then transforms to a bulb and sinks into the underlying mesenchyme. The signaling pathways regulating the early morphogenetic steps have been identified to some extent, but the underlying cellular mechanisms remain ill defined. Here, we use 3D and 4D confocal microscopy to show that the early growth of the mammary rudiment is accomplished by migration-driven cell influx, with minor contributions of cell hypertrophy and proliferation. We delineate a hitherto undescribed invagination mechanism driven by thin, elongated keratinocytes-ring cells-that form a contractile rim around the mammary bud and likely exert force via the actomyosin network. Furthermore, we show that conditional deletion of nonmuscle myosin IIA (NMIIA) impairs invagination, resulting in abnormal mammary bud shape.


Asunto(s)
Actomiosina/metabolismo , Movimiento Celular , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/metabolismo , Mecanotransducción Celular , Animales , Proliferación Celular , Células Epiteliales/ultraestructura , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Hipertrofia , Queratinocitos/metabolismo , Queratinocitos/ultraestructura , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/ultraestructura , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Morfogénesis
20.
J Invest Dermatol ; 141(11): 2602-2610.e3, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33965403

RESUMEN

The skin epidermis is attached to the underlying dermis by a laminin 332 (Lm332)-rich basement membrane. Consequently, loss of Lm332 leads to the severe blistering disorder epidermolysis bullosa junctionalis in humans and animals. Owing to the indispensable role of Lm332 in keratinocyte adhesion in vivo, the severity of the disease has limited research into other functions of the protein. We have conditionally disrupted Lm332 expression in basal keratinocytes of adult mice. Although blisters develop along the interfollicular epidermis, hair follicle basal cells provide sufficient anchorage of the epidermis to the dermis, making inducible deletion of the Lama3 gene compatible with life. Loss of Lm332 promoted the thickening of the epidermis and exaggerated desquamation. Global RNA expression analysis revealed major changes in the expression of keratins, cornified envelope proteins, and cellular stress markers. These modifications of the keratinocyte genetic program are accompanied by changes in cell shape and disorganization of the actin cytoskeleton. These data indicate that loss of Lm332-mediated progenitor cell adhesion alters cell fate and disturbs epidermal homeostasis.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Homeostasis/fisiología , Queratinocitos/citología , Citoesqueleto de Actina/fisiología , Alarminas/fisiología , Animales , Vesícula/etiología , Diferenciación Celular , Epidermis/patología , Queratinas/análisis , Ratones , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/análisis , Kalinina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...