Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.126
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(13): 3445-3459.e15, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38838668

RESUMEN

Understanding cellular force transmission dynamics is crucial in mechanobiology. We developed the DNA-based ForceChrono probe to measure force magnitude, duration, and loading rates at the single-molecule level within living cells. The ForceChrono probe circumvents the limitations of in vitro single-molecule force spectroscopy by enabling direct measurements within the dynamic cellular environment. Our findings reveal integrin force loading rates of 0.5-2 pN/s and durations ranging from tens of seconds in nascent adhesions to approximately 100 s in mature focal adhesions. The probe's robust and reversible design allows for continuous monitoring of these dynamic changes as cells undergo morphological transformations. Additionally, by analyzing how mutations, deletions, or pharmacological interventions affect these parameters, we can deduce the functional roles of specific proteins or domains in cellular mechanotransduction. The ForceChrono probe provides detailed insights into the dynamics of mechanical forces, advancing our understanding of cellular mechanics and the molecular mechanisms of mechanotransduction.


Asunto(s)
Mecanotransducción Celular , Imagen Individual de Molécula , Animales , Humanos , Ratones , Fenómenos Biomecánicos , Adhesión Celular , ADN/química , ADN/metabolismo , Adhesiones Focales/metabolismo , Integrinas/metabolismo , Microscopía de Fuerza Atómica/métodos , Imagen Individual de Molécula/métodos , Línea Celular , Supervivencia Celular , Emparejamiento Base , Calibración
2.
Cell ; 187(12): 2990-3005.e17, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38772370

RESUMEN

Integrins link the extracellular environment to the actin cytoskeleton in cell migration and adhesiveness. Rapid coordination between events outside and inside the cell is essential. Single-molecule fluorescence dynamics show that ligand binding to the bent-closed integrin conformation, which predominates on cell surfaces, is followed within milliseconds by two concerted changes, leg extension and headpiece opening, to give the high-affinity integrin conformation. The extended-closed integrin conformation is not an intermediate but can be directly accessed from the extended-open conformation and provides a pathway for ligand dissociation. In contrast to ligand, talin, which links the integrin ß-subunit cytoplasmic domain to the actin cytoskeleton, modestly stabilizes but does not induce extension or opening. Integrin activation is thus initiated by outside-in signaling and followed by inside-out signaling. Our results further imply that talin binding is insufficient for inside-out integrin activation and that tensile force transmission through the ligand-integrin-talin-actin cytoskeleton complex is required.


Asunto(s)
Integrinas , Talina , Animales , Humanos , Ratones , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/química , Adhesión Celular , Células CHO , Cricetulus , Integrinas/metabolismo , Integrinas/química , Ligandos , Unión Proteica , Conformación Proteica , Transducción de Señal , Imagen Individual de Molécula , Talina/metabolismo , Talina/química
3.
Annu Rev Immunol ; 32: 51-82, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24313777

RESUMEN

The cytokine TGF-ß plays an integral role in regulating immune responses. TGF-ß has pleiotropic effects on adaptive immunity, especially in the regulation of effector and regulatory CD4(+) T cell responses. Many immune and nonimmune cells can produce TGF-ß, but it is always produced as an inactive complex that must be activated to exert functional effects. Thus, activation of latent TGF-ß provides a crucial layer of regulation that controls TGF-ß function. In this review, we highlight some of the important functional roles for TGF-ß in immunity, focusing on its context-specific roles in either dampening or promoting T cell responses. We also describe how activation of TGF-ß controls its function in the immune system, with a focus on the key roles for members of the integrin family in this process.


Asunto(s)
Inmunidad/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Inmunidad Adaptativa , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Sistema Inmunológico/fisiología , Integrinas/metabolismo , Unión Proteica , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Factor de Crecimiento Transformador beta/química
4.
Cell ; 184(14): 3702-3716.e30, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34133940

RESUMEN

Many embryonic organs undergo epithelial morphogenesis to form tree-like hierarchical structures. However, it remains unclear what drives the budding and branching of stratified epithelia, such as in the embryonic salivary gland and pancreas. Here, we performed live-organ imaging of mouse embryonic salivary glands at single-cell resolution to reveal that budding morphogenesis is driven by expansion and folding of a distinct epithelial surface cell sheet characterized by strong cell-matrix adhesions and weak cell-cell adhesions. Profiling of single-cell transcriptomes of this epithelium revealed spatial patterns of transcription underlying these cell adhesion differences. We then synthetically reconstituted budding morphogenesis by experimentally suppressing E-cadherin expression and inducing basement membrane formation in 3D spheroid cultures of engineered cells, which required ß1-integrin-mediated cell-matrix adhesion for successful budding. Thus, stratified epithelial budding, the key first step of branching morphogenesis, is driven by an overall combination of strong cell-matrix adhesion and weak cell-cell adhesion by peripheral epithelial cells.


Asunto(s)
Uniones Célula-Matriz/metabolismo , Morfogénesis , Animales , Membrana Basal/metabolismo , Adhesión Celular , División Celular , Movimiento Celular , Rastreo Celular , Embrión de Mamíferos/citología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Integrinas/metabolismo , Ratones , Modelos Biológicos , Glándulas Salivales/citología , Glándulas Salivales/embriología , Glándulas Salivales/metabolismo , Transcriptoma/genética
5.
Cell ; 184(19): 4919-4938.e22, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34506722

RESUMEN

Replacing or editing disease-causing mutations holds great promise for treating many human diseases. Yet, delivering therapeutic genetic modifiers to specific cells in vivo has been challenging, particularly in large, anatomically distributed tissues such as skeletal muscle. Here, we establish an in vivo strategy to evolve and stringently select capsid variants of adeno-associated viruses (AAVs) that enable potent delivery to desired tissues. Using this method, we identify a class of RGD motif-containing capsids that transduces muscle with superior efficiency and selectivity after intravenous injection in mice and non-human primates. We demonstrate substantially enhanced potency and therapeutic efficacy of these engineered vectors compared to naturally occurring AAV capsids in two mouse models of genetic muscle disease. The top capsid variants from our selection approach show conserved potency for delivery across a variety of inbred mouse strains, and in cynomolgus macaques and human primary myotubes, with transduction dependent on target cell expressed integrin heterodimers.


Asunto(s)
Cápside/metabolismo , Dependovirus/metabolismo , Evolución Molecular Dirigida , Técnicas de Transferencia de Gen , Músculo Esquelético/metabolismo , Secuencia de Aminoácidos , Animales , Cápside/química , Células Cultivadas , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Integrinas/metabolismo , Macaca fascicularis , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/terapia , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/terapia , Multimerización de Proteína , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/uso terapéutico , ARN Guía de Kinetoplastida/metabolismo , Recombinación Genética/genética , Especificidad de la Especie , Transgenes
6.
Nat Immunol ; 24(6): 915-924, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37081147

RESUMEN

Immune cell locomotion is associated with amoeboid migration, a flexible mode of movement, which depends on rapid cycles of actin polymerization and actomyosin contraction1. Many immune cells do not necessarily require integrins, the major family of adhesion receptors in mammals, to move productively through three-dimensional tissue spaces2,3. Instead, they can use alternative strategies to transmit their actin-driven forces to the substrate, explaining their migratory adaptation to changing external environments4-6. However, whether these generalized concepts apply to all immune cells is unclear. Here, we show that the movement of mast cells (immune cells with important roles during allergy and anaphylaxis) differs fundamentally from the widely applied paradigm of interstitial immune cell migration. We identify a crucial role for integrin-dependent adhesion in controlling mast cell movement and localization to anatomical niches rich in KIT ligand, the major mast cell growth and survival factor. Our findings show that substrate-dependent haptokinesis is an important mechanism for the tissue organization of resident immune cells.


Asunto(s)
Actinas , Integrinas , Animales , Integrinas/metabolismo , Actinas/metabolismo , Mastocitos/metabolismo , Movimiento Celular , Leucocitos/metabolismo , Adhesión Celular , Mamíferos/metabolismo
7.
Nat Rev Mol Cell Biol ; 24(7): 495-516, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36849594

RESUMEN

Mechanical properties of extracellular matrices (ECMs) regulate essential cell behaviours, including differentiation, migration and proliferation, through mechanotransduction. Studies of cell-ECM mechanotransduction have largely focused on cells cultured in 2D, on top of elastic substrates with a range of stiffnesses. However, cells often interact with ECMs in vivo in a 3D context, and cell-ECM interactions and mechanisms of mechanotransduction in 3D can differ from those in 2D. The ECM exhibits various structural features as well as complex mechanical properties. In 3D, mechanical confinement by the surrounding ECM restricts changes in cell volume and cell shape but allows cells to generate force on the matrix by extending protrusions and regulating cell volume as well as through actomyosin-based contractility. Furthermore, cell-matrix interactions are dynamic owing to matrix remodelling. Accordingly, ECM stiffness, viscoelasticity and degradability often play a critical role in regulating cell behaviours in 3D. Mechanisms of 3D mechanotransduction include traditional integrin-mediated pathways that sense mechanical properties and more recently described mechanosensitive ion channel-mediated pathways that sense 3D confinement, with both converging on the nucleus for downstream control of transcription and phenotype. Mechanotransduction is involved in tissues from development to cancer and is being increasingly harnessed towards mechanotherapy. Here we discuss recent progress in our understanding of cell-ECM mechanotransduction in 3D.


Asunto(s)
Matriz Extracelular , Mecanotransducción Celular , Matriz Extracelular/metabolismo , Citoesqueleto de Actina , Diferenciación Celular , Integrinas/metabolismo
8.
Nat Rev Mol Cell Biol ; 24(2): 142-161, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36168065

RESUMEN

The ability of animal cells to sense, adhere to and remodel their local extracellular matrix (ECM) is central to control of cell shape, mechanical responsiveness, motility and signalling, and hence to development, tissue formation, wound healing and the immune response. Cell-ECM interactions occur at various specialized, multi-protein adhesion complexes that serve to physically link the ECM to the cytoskeleton and the intracellular signalling apparatus. This occurs predominantly via clustered transmembrane receptors of the integrin family. Here we review how the interplay of mechanical forces, biochemical signalling and molecular self-organization determines the composition, organization, mechanosensitivity and dynamics of these adhesions. Progress in the identification of core multi-protein modules within the adhesions and characterization of rearrangements of their components in response to force, together with advanced imaging approaches, has improved understanding of adhesion maturation and turnover and the relationships between adhesion structures and functions. Perturbations of adhesion contribute to a broad range of diseases and to age-related dysfunction, thus an improved understanding of their molecular nature may facilitate therapeutic intervention in these conditions.


Asunto(s)
Adhesión Celular , Citoesqueleto , Matriz Extracelular , Integrinas , Animales , Adhesión Celular/fisiología , Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Integrinas/metabolismo , Transducción de Señal , Adherencias Tisulares/patología
9.
Cell ; 180(3): 490-501.e16, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31955848

RESUMEN

Integrin αvß8 binds with exquisite specificity to latent transforming growth factor-ß (L-TGF-ß). This binding is essential for activating L-TGF-ß presented by a variety of cell types. Inhibiting αvß8-mediated TGF-ß activation blocks immunosuppressive regulatory T cell differentiation, which is a potential therapeutic strategy in cancer. Using cryo-electron microscopy, structure-guided mutagenesis, and cell-based assays, we reveal the binding interactions between the entire αvß8 ectodomain and its intact natural ligand, L-TGF-ß, as well as two different inhibitory antibody fragments to understand the structural underpinnings of αvß8 binding specificity and TGF-ß activation. Our studies reveal a mechanism of TGF-ß activation where mature TGF-ß signals within the confines of L-TGF-ß and the release and diffusion of TGF-ß are not required. The structural details of this mechanism provide a rational basis for therapeutic strategies to inhibit αvß8-mediated L-TGF-ß activation.


Asunto(s)
Microscopía por Crioelectrón/métodos , Integrinas/química , Integrinas/metabolismo , Proteínas de Unión a TGF-beta Latente/química , Proteínas de Unión a TGF-beta Latente/metabolismo , Factor de Crecimiento Transformador beta1/química , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Anticuerpos/inmunología , Sitios de Unión , Bronquios/citología , Células CHO , Cricetulus , Femenino , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Integrinas/inmunología , Activación de Linfocitos , Masculino , Visón , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Linfocitos T Reguladores/inmunología
10.
Cell ; 182(3): 545-562.e23, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32621799

RESUMEN

Scar tissue size following myocardial infarction is an independent predictor of cardiovascular outcomes, yet little is known about factors regulating scar size. We demonstrate that collagen V, a minor constituent of heart scars, regulates the size of heart scars after ischemic injury. Depletion of collagen V led to a paradoxical increase in post-infarction scar size with worsening of heart function. A systems genetics approach across 100 in-bred strains of mice demonstrated that collagen V is a critical driver of postinjury heart function. We show that collagen V deficiency alters the mechanical properties of scar tissue, and altered reciprocal feedback between matrix and cells induces expression of mechanosensitive integrins that drive fibroblast activation and increase scar size. Cilengitide, an inhibitor of specific integrins, rescues the phenotype of increased post-injury scarring in collagen-V-deficient mice. These observations demonstrate that collagen V regulates scar size in an integrin-dependent manner.


Asunto(s)
Cicatriz/metabolismo , Colágeno Tipo V/deficiencia , Colágeno Tipo V/metabolismo , Lesiones Cardíacas/metabolismo , Contracción Miocárdica/genética , Miofibroblastos/metabolismo , Animales , Cicatriz/genética , Cicatriz/fisiopatología , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Colágeno Tipo V/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Femenino , Fibrosis/genética , Fibrosis/metabolismo , Regulación de la Expresión Génica/genética , Integrinas/antagonistas & inhibidores , Integrinas/genética , Integrinas/metabolismo , Isoproterenol/farmacología , Masculino , Mecanotransducción Celular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía de Fuerza Atómica/instrumentación , Microscopía Electrónica de Transmisión , Contracción Miocárdica/efectos de los fármacos , Miofibroblastos/citología , Miofibroblastos/patología , Miofibroblastos/ultraestructura , Análisis de Componente Principal , Proteómica , RNA-Seq , Análisis de la Célula Individual
11.
Cell ; 182(3): 563-577.e20, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32615086

RESUMEN

Adipose tissues dynamically remodel their cellular composition in response to external cues by stimulating beige adipocyte biogenesis; however, the developmental origin and pathways regulating this process remain insufficiently understood owing to adipose tissue heterogeneity. Here, we employed single-cell RNA-seq and identified a unique subset of adipocyte progenitor cells (APCs) that possessed the cell-intrinsic plasticity to give rise to beige fat. This beige APC population is proliferative and marked by cell-surface proteins, including PDGFRα, Sca1, and CD81. Notably, CD81 is not only a beige APC marker but also required for de novo beige fat biogenesis following cold exposure. CD81 forms a complex with αV/ß1 and αV/ß5 integrins and mediates the activation of integrin-FAK signaling in response to irisin. Importantly, CD81 loss causes diet-induced obesity, insulin resistance, and adipose tissue inflammation. These results suggest that CD81 functions as a key sensor of external inputs and controls beige APC proliferation and whole-body energy homeostasis.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo Beige/metabolismo , Metabolismo Energético/genética , Quinasa 1 de Adhesión Focal/metabolismo , Transducción de Señal/genética , Células Madre/metabolismo , Tetraspanina 28/metabolismo , Adipocitos/metabolismo , Tejido Adiposo Beige/citología , Tejido Adiposo Beige/crecimiento & desarrollo , Tejido Adiposo Blanco/metabolismo , Adulto , Animales , Ataxina-1/metabolismo , Femenino , Fibronectinas/farmacología , Quinasa 1 de Adhesión Focal/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Resistencia a la Insulina/genética , Integrinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/genética , Obesidad/metabolismo , RNA-Seq , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de la Célula Individual , Células Madre/citología , Tetraspanina 28/genética
12.
Cell ; 177(3): 499-501, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30952447

RESUMEN

One of the 2019 Canada Gairdner International Awards recognizes Timothy Springer's discovery of the first immune system adhesion molecules involved in lymphocyte homing and the translation of those discoveries into therapeutics for autoimmune disease and cancer.


Asunto(s)
Adhesión Celular/fisiología , Integrinas/metabolismo , Linfocitos T/metabolismo , Citoesqueleto de Actina , Anticuerpos Monoclonales/inmunología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Humanos , Glicoproteínas de Membrana/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Selectina-P/metabolismo , Linfocitos T/inmunología
13.
Cell ; 179(1): 120-131.e13, 2019 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-31539492

RESUMEN

Focal adhesions (FAs) are protein machineries essential for cell adhesion, migration, and differentiation. Talin is an integrin-activating and tension-sensing FA component directly connecting integrins in the plasma membrane with the actomyosin cytoskeleton. To understand how talin function is regulated, we determined a cryoelectron microscopy (cryo-EM) structure of full-length talin1 revealing a two-way mode of autoinhibition. The actin-binding rod domains fold into a 15-nm globular arrangement that is interlocked by the integrin-binding FERM head. In turn, the rod domains R9 and R12 shield access of the FERM domain to integrin and the phospholipid PIP2 at the membrane. This mechanism likely ensures synchronous inhibition of integrin, membrane, and cytoskeleton binding. We also demonstrate that compacted talin1 reversibly unfolds to an ∼60-nm string-like conformation, revealing interaction sites for vinculin and actin. Our data explain how fast switching between active and inactive conformations of talin could regulate FA turnover, a process critical for cell adhesion and signaling.


Asunto(s)
Adhesiones Focales/metabolismo , Dominios y Motivos de Interacción de Proteínas , Talina/química , Talina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Sitios de Unión , Adhesión Celular/fisiología , Microscopía por Crioelectrón , Citoesqueleto/metabolismo , Dimerización , Escherichia coli/metabolismo , Humanos , Integrinas/metabolismo , Modelos Moleculares , Unión Proteica , Transducción de Señal/fisiología , Vinculina/metabolismo
14.
Nat Immunol ; 22(3): 381-390, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33589816

RESUMEN

The integrin α4ß7 selectively regulates lymphocyte trafficking and adhesion in the gut and gut-associated lymphoid tissue (GALT). Here, we describe unexpected involvement of the tyrosine phosphatase Shp1 and the B cell lectin CD22 (Siglec-2) in the regulation of α4ß7 surface expression and gut immunity. Shp1 selectively inhibited ß7 endocytosis, enhancing surface α4ß7 display and lymphocyte homing to GALT. In B cells, CD22 associated in a sialic acid-dependent manner with integrin ß7 on the cell surface to target intracellular Shp1 to ß7. Shp1 restrained plasma membrane ß7 phosphorylation and inhibited ß7 endocytosis without affecting ß1 integrin. B cells with reduced Shp1 activity, lacking CD22 or expressing CD22 with mutated Shp1-binding or carbohydrate-binding domains displayed parallel reductions in surface α4ß7 and in homing to GALT. Consistent with the specialized role of α4ß7 in intestinal immunity, CD22 deficiency selectively inhibited intestinal antibody and pathogen responses.


Asunto(s)
Linfocitos B/enzimología , Inmunidad Mucosa , Cadenas beta de Integrinas/metabolismo , Integrinas/metabolismo , Mucosa Intestinal/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/virología , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Endocitosis , Femenino , Cadenas beta de Integrinas/inmunología , Integrinas/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/virología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Rotavirus/inmunología , Rotavirus/patogenicidad , Infecciones por Rotavirus/genética , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/deficiencia , Lectina 2 Similar a Ig de Unión al Ácido Siálico/genética , Transducción de Señal , Técnicas de Cultivo de Tejidos
15.
Cell ; 174(1): 131-142.e13, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29958103

RESUMEN

Macrophages protect the body from damage and disease by targeting antibody-opsonized cells for phagocytosis. Though antibodies can be raised against antigens with diverse structures, shapes, and sizes, it is unclear why some are more effective at triggering immune responses than others. Here, we define an antigen height threshold that regulates phagocytosis of both engineered and cancer-specific antigens by macrophages. Using a reconstituted model of antibody-opsonized target cells, we find that phagocytosis is dramatically impaired for antigens that position antibodies >10 nm from the target surface. Decreasing antigen height drives segregation of antibody-bound Fc receptors from the inhibitory phosphatase CD45 in an integrin-independent manner, triggering Fc receptor phosphorylation and promoting phagocytosis. Our work shows that close contact between macrophage and target is a requirement for efficient phagocytosis, suggesting that therapeutic antibodies should target short antigens in order to trigger Fc receptor activation through size-dependent physical segregation.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos/química , Macrófagos/inmunología , Proteínas Opsoninas/metabolismo , Fagocitosis , Animales , Anticuerpos Monoclonales/química , Antígenos/genética , Antígenos/inmunología , Antígeno Carcinoembrionario/química , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Edición Génica , Integrinas/metabolismo , Antígenos Comunes de Leucocito/química , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Macrófagos/citología , Ratones , Proteínas Opsoninas/química , Fosforilación , Células RAW 264.7 , Receptores Fc/inmunología , Receptores Fc/metabolismo , Liposomas Unilamelares/química
16.
Cell ; 174(1): 156-171.e16, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29909984

RESUMEN

Extracellular proTGF-ß is covalently linked to "milieu" molecules in the matrix or on cell surfaces and is latent until TGF-ß is released by integrins. Here, we show that LRRC33 on the surface of microglia functions as a milieu molecule and enables highly localized, integrin-αVß8-dependent TGF-ß activation. Lrrc33-/- mice lack CNS vascular abnormalities associated with deficiency in TGF-ß-activating integrins but have microglia with a reactive phenotype and after 2 months develop ascending paraparesis with loss of myelinated axons and death by 5 months. Whole bone marrow transplantation results in selective repopulation of Lrrc33-/- brains with WT microglia and halts disease progression. The phenotypes of WT and Lrrc33-/- microglia in the same brain suggest that there is little spreading of TGF-ß activated from one microglial cell to neighboring microglia. Our results suggest that interactions between integrin-bearing cells and cells bearing milieu molecule-associated TGF-ß provide localized and selective activation of TGF-ß.


Asunto(s)
Proteínas Portadoras/metabolismo , Microglía/metabolismo , Sistema Nervioso/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Axones/metabolismo , Trasplante de Médula Ósea , Encéfalo/metabolismo , Proteínas Portadoras/clasificación , Proteínas Portadoras/genética , Células Cultivadas , Integrinas/metabolismo , Estimación de Kaplan-Meier , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Mutagénesis Sitio-Dirigida , Enfermedades Neurodegenerativas/mortalidad , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Filogenia , Unión Proteica , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Factor de Crecimiento Transformador beta/genética
17.
Immunity ; 56(6): 1285-1302.e7, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37269830

RESUMEN

The integrin CD49a marks highly cytotoxic epidermal-tissue-resident memory (TRM) cells, but their differentiation from circulating populations remains poorly defined. We demonstrate enrichment of RUNT family transcription-factor-binding motifs in human epidermal CD8+CD103+CD49a+ TRM cells, paralleled by high RUNX2 and RUNX3 protein expression. Sequencing of paired skin and blood samples revealed clonal overlap between epidermal CD8+CD103+CD49a+ TRM cells and circulating memory CD8+CD45RA-CD62L+ T cells. In vitro stimulation of circulating CD8+CD45RA-CD62L+ T cells with IL-15 and TGF-ß induced CD49a expression and cytotoxic transcriptional profiles in a RUNX2- and RUNX3-dependent manner. We therefore identified a reservoir of circulating cells with cytotoxic TRM potential. In melanoma patients, high RUNX2, but not RUNX3, transcription correlated with a cytotoxic CD8+CD103+CD49a+ TRM cell signature and improved patient survival. Together, our results indicate that combined RUNX2 and RUNX3 activity promotes the differentiation of cytotoxic CD8+CD103+CD49a+ TRM cells, providing immunosurveillance of infected and malignant cells.


Asunto(s)
Linfocitos T CD8-positivos , Melanoma , Humanos , Linfocitos T CD8-positivos/metabolismo , Integrina alfa1/metabolismo , Integrinas/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Memoria Inmunológica , Antígenos Comunes de Leucocito/metabolismo , Melanoma/metabolismo
18.
Cell ; 171(6): 1368-1382.e23, 2017 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-29195076

RESUMEN

Blood platelets are critical for hemostasis and thrombosis and play diverse roles during immune responses. Despite these versatile tasks in mammalian biology, their skills on a cellular level are deemed limited, mainly consisting in rolling, adhesion, and aggregate formation. Here, we identify an unappreciated asset of platelets and show that adherent platelets use adhesion receptors to mechanically probe the adhesive substrate in their local microenvironment. When actomyosin-dependent traction forces overcome substrate resistance, platelets migrate and pile up the adhesive substrate together with any bound particulate material. They use this ability to act as cellular scavengers, scanning the vascular surface for potential invaders and collecting deposited bacteria. Microbe collection by migrating platelets boosts the activity of professional phagocytes, exacerbating inflammatory tissue injury in sepsis. This assigns platelets a central role in innate immune responses and identifies them as potential targets to dampen inflammatory tissue damage in clinical scenarios of severe systemic infection.


Asunto(s)
Infecciones Bacterianas/inmunología , Plaquetas/inmunología , Animales , Bacterias/clasificación , Plaquetas/citología , Vasos Sanguíneos/lesiones , Vasos Sanguíneos/patología , Calcio/metabolismo , Movimiento Celular , Polaridad Celular , Humanos , Inflamación/inmunología , Integrinas/metabolismo , Ratones , Miosinas/metabolismo , Neutrófilos/citología
19.
Immunity ; 55(2): 290-307.e5, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35090581

RESUMEN

Tbet+CD11c+ B cells arise during type 1 pathogen challenge, aging, and autoimmunity in mice and humans. Here, we examined the developmental requirements of this B cell subset. In acute infection, T follicular helper (Tfh) cells, but not Th1 cells, drove Tbet+CD11c+ B cell generation through proximal delivery of help. Tbet+CD11c+ B cells developed prior to germinal center (GC) formation, exhibiting phenotypic and transcriptional profiles distinct from GC B cells. Fate tracking revealed that most Tbet+CD11c+ B cells developed independently of GC entry and cell-intrinsic Bcl6 expression. Tbet+CD11c+ and GC B cells exhibited minimal repertoire overlap, indicating distinct developmental pathways. As the infection resolved, Tbet+CD11c+ B cells localized to the marginal zone where splenic retention depended on integrins LFA-1 and VLA-4, forming a competitive memory subset that contributed to antibody production and secondary GC seeding upon rechallenge. Therefore, Tbet+CD11c+ B cells comprise a GC-independent memory subset capable of rapid and robust recall responses.


Asunto(s)
Linfocitos B/inmunología , Antígenos CD11/metabolismo , Subgrupos Linfocitarios/inmunología , Células T Auxiliares Foliculares/inmunología , Proteínas de Dominio T Box/metabolismo , Virosis/inmunología , Animales , Anticuerpos Antivirales/metabolismo , Linfocitos B/metabolismo , Diferenciación Celular/inmunología , Centro Germinal/inmunología , Alphainfluenzavirus/inmunología , Integrinas/metabolismo , Subgrupos Linfocitarios/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Células B de Memoria/inmunología , Células B de Memoria/metabolismo , Ratones , Bazo/inmunología
20.
Nat Rev Mol Cell Biol ; 20(8): 457-473, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31182865

RESUMEN

Integrins, and integrin-mediated adhesions, have long been recognized to provide the main molecular link attaching cells to the extracellular matrix (ECM) and to serve as bidirectional hubs transmitting signals between cells and their environment. Recent evidence has shown that their combined biochemical and mechanical properties also allow integrins to sense, respond to and interact with ECM of differing properties with exquisite specificity. Here, we review this work first by providing an overview of how integrin function is regulated from both a biochemical and a mechanical perspective, affecting integrin cell-surface availability, binding properties, activation or clustering. Then, we address how this biomechanical regulation allows integrins to respond to different ECM physicochemical properties and signals, such as rigidity, composition and spatial distribution. Finally, we discuss the importance of this sensing for major cell functions by taking cell migration and cancer as examples.


Asunto(s)
Movimiento Celular , Matriz Extracelular/metabolismo , Integrinas/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral , Animales , Adhesión Celular , Membrana Celular/metabolismo , Membrana Celular/patología , Humanos , Neoplasias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA