Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Nat Immunol ; 20(10): 1269-1278, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31534240

RESUMO

The immune response is orchestrated by a variety of immune cells. The function of each cell is determined by the collective signals from various immunoreceptors, whose expression and activity depend on the developmental stages of the cell and its environmental context. Recent studies have highlighted the presence of mechanical force on several immunoreceptor-ligand pairs and the important role of force in regulating their interaction and function. In this Perspective, we use the T cell antigen receptor as an example with which to review the current understanding of the mechanosensing properties of immunoreceptors. We discuss the types of forces that immunoreceptors may encounter and the effects of force on ligand bonding, conformational change and the triggering of immunoreceptors, as well as the effects of force on the downstream signal transduction, cell-fate decisions and effector function of immune cells.


Assuntos
Regulação Alostérica/imunologia , Sinapses Imunológicas/metabolismo , Mecanotransdução Celular/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores Imunológicos/metabolismo , Animais , Velocidade do Fluxo Sanguíneo , Adesão Celular , Movimento Celular , Microambiente Celular , Humanos , Imunidade , Migração e Rolagem de Leucócitos , Receptor Cross-Talk , Transdução de Sinais
2.
Nat Immunol ; 20(11): 1506-1516, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31611698

RESUMO

Fibroblastic reticular cells (FRCs) and their specialized collagen fibers termed 'conduits' form fundamental structural units supporting lymphoid tissues. In lymph nodes, conduits are known to transport interstitial fluid and small molecules from afferent lymphatics into the nodal parenchyma. However, the immunological contributions of conduit function have remained elusive. Here, we report that intestinal Peyer's patches (PPs) contain a specialized conduit system that directs the flow of water absorbed across the intestinal epithelium. Notably, PP FRCs responded to conduit fluid flow via the mechanosensitive ion channel Piezo1. Disruption of fluid flow or genetic deficiency of Piezo1 on CCL19-expressing stroma led to profound structural alterations in perivascular FRCs and associated high endothelial venules. This in turn impaired lymphocyte entry into PPs and initiation of mucosal antibody responses. These results identify a critical role for conduit-mediated fluid flow in the maintenance of PP homeostasis and mucosal immunity.


Assuntos
Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Linfócitos/imunologia , Mecanotransdução Celular/imunologia , Nódulos Linfáticos Agregados/imunologia , Animais , Anticorpos/imunologia , Anticorpos/metabolismo , Movimento Celular/imunologia , Quimiocina CCL19/metabolismo , Feminino , Mucosa Intestinal/metabolismo , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Ativação Linfocitária , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Nódulos Linfáticos Agregados/metabolismo , Água/metabolismo
3.
Nat Immunol ; 19(12): 1379-1390, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30420628

RESUMO

The T cell antigen receptor (TCR) expressed on thymocytes interacts with self-peptide major histocompatibility complex (pMHC) ligands to signal apoptosis or survival. Here, we found that negative-selection ligands induced thymocytes to exert forces on the TCR and the co-receptor CD8 and formed cooperative TCR-pMHC-CD8 trimolecular 'catch bonds', whereas positive-selection ligands induced less sustained thymocyte forces on TCR and CD8 and formed shorter-lived, independent TCR-pMHC and pMHC-CD8 bimolecular 'slip bonds'. Catch bonds were not intrinsic to either the TCR-pMHC or the pMHC-CD8 arm of the trans (cross-junctional) heterodimer but resulted from coupling of the extracellular pMHC-CD8 interaction to the intracellular interaction of CD8 with TCR-CD3 via associated kinases to form a cis (lateral) heterodimer capable of inside-out signaling. We suggest that the coupled trans-cis heterodimeric interactions form a mechanotransduction loop that reinforces negative-selection signaling that is distinct from positive-selection signaling in the thymus.


Assuntos
Mecanotransdução Celular/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Timócitos/imunologia , Timo/imunologia , Animais , Deleção Clonal/imunologia , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/metabolismo , Timócitos/metabolismo
4.
Nature ; 573(7772): 69-74, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31435009

RESUMO

Direct recognition of invading pathogens by innate immune cells is a critical driver of the inflammatory response. However, cells of the innate immune system can also sense their local microenvironment and respond to physiological fluctuations in temperature, pH, oxygen and nutrient availability, which are altered during inflammation. Although cells of the immune system experience force and pressure throughout their life cycle, little is known about how these mechanical processes regulate the immune response. Here we show that cyclical hydrostatic pressure, similar to that experienced by immune cells in the lung, initiates an inflammatory response via the mechanically activated ion channel PIEZO1. Mice lacking PIEZO1 in innate immune cells showed ablated pulmonary inflammation in the context of bacterial infection or fibrotic autoinflammation. Our results reveal an environmental sensory axis that stimulates innate immune cells to mount an inflammatory response, and demonstrate a physiological role for PIEZO1 and mechanosensation in immunity.


Assuntos
Pressão Hidrostática , Imunidade Inata , Canais Iônicos/metabolismo , Mecanotransdução Celular/imunologia , Animais , Endotelina-1/metabolismo , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Transdução de Sinais
5.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33758095

RESUMO

Osteoarthritis (OA) is a painful and debilitating condition of synovial joints without any disease-modifying therapies [A. M. Valdes, T. D. Spector, Nat. Rev. Rheumatol. 7, 23-32 (2011)]. We previously identified mechanosensitive PIEZO channels, PIEZO1 and PIEZO2, both expressed in articular cartilage, to function in chondrocyte mechanotransduction in response to injury [W. Lee et al., Proc. Natl. Acad. Sci. U.S.A. 111, E5114-E5122 (2014); W. Lee, F. Guilak, W. Liedtke, Curr. Top. Membr. 79, 263-273 (2017)]. We therefore asked whether interleukin-1-mediated inflammatory signaling, as occurs in OA, influences Piezo gene expression and channel function, thus indicative of maladaptive reprogramming that can be rationally targeted. Primary porcine chondrocyte culture and human osteoarthritic cartilage tissue were studied. We found that interleukin-1α (IL-1α) up-regulated Piezo1 in porcine chondrocytes. Piezo1 expression was significantly increased in human osteoarthritic cartilage. Increased Piezo1 expression in chondrocytes resulted in a feed-forward pathomechanism whereby increased function of Piezo1 induced excess intracellular Ca2+ at baseline and in response to mechanical deformation. Elevated resting state Ca2+ in turn rarefied the F-actin cytoskeleton and amplified mechanically induced deformation microtrauma. As intracellular substrates of this OA-related inflammatory pathomechanism, in porcine articular chondrocytes exposed to IL-1α, we discovered that enhanced Piezo1 expression depended on p38 MAP-kinase and transcription factors HNF4 and ATF2/CREBP1. CREBP1 directly bound to the proximal PIEZO1 gene promoter. Taken together, these signaling and genetic reprogramming events represent a detrimental Ca2+-driven feed-forward mechanism that can be rationally targeted to stem the progression of OA.


Assuntos
Condrócitos/metabolismo , Interleucina-1alfa/metabolismo , Canais Iônicos/genética , Mecanotransdução Celular/imunologia , Osteoartrite/imunologia , Fator 2 Ativador da Transcrição/metabolismo , Animais , Cálcio/metabolismo , Cartilagem Articular/citologia , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/imunologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Canais Iônicos/metabolismo , Mecanotransdução Celular/genética , Osteoartrite/genética , Osteoartrite/patologia , Cultura Primária de Células , Regiões Promotoras Genéticas/genética , Sus scrofa , Regulação para Cima/imunologia
6.
FASEB J ; 36(2): e22007, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35051300

RESUMO

The aim of this study was to further elucidate the molecular mechanisms that mediate pathologic foreign body response (FBR) to biomedical implants. The longevity of biomedical implants is limited by the FBR, which leads to implant failure and patient morbidity. Since the specific molecular mechanisms underlying fibrotic responses to biomedical implants have yet to be fully described, there are currently no targeted approaches to reduce pathologic FBR. We utilized proteomics analysis of human FBR samples to identify potential molecular targets for therapeutic inhibition of FBR. We then employed a murine model of FBR to further evaluate the role of this potential target. We performed histological and immunohistochemical analysis on the murine FBR capsule tissue, as well as single-cell RNA sequencing (scRNA-seq) on cells isolated from the capsules. We identified IQ motif containing GTPase activating protein 1 (IQGAP1) as the most promising of several targets, serving as a central molecular mediator in human and murine FBR compared to control subcutaneous tissue. IQGAP1-deficient mice displayed a significantly reduced FBR compared to wild-type mice as evidenced by lower levels of collagen deposition and maturity. Our scRNA-seq analysis revealed that decreasing IQGAP1 resulted in diminished transcription of mechanotransduction, inflammation, and fibrosis-related genes, which was confirmed on the protein level with immunofluorescent staining. The deficiency of IQGAP1 significantly attenuates FBR by deactivating downstream mechanotransduction signaling, inflammation, and fibrotic pathways. IQGAP1 may be a promising target for rational therapeutic design to mitigate pathologic FBR around biomedical implants.


Assuntos
Materiais Biocompatíveis/efeitos adversos , Corpos Estranhos/imunologia , Próteses e Implantes/efeitos adversos , Transdução de Sinais/imunologia , Proteínas Ativadoras de ras GTPase/imunologia , Animais , Colágeno/imunologia , Fibrose/imunologia , Humanos , Inflamação/imunologia , Masculino , Mecanotransdução Celular/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transcrição Gênica/imunologia
7.
Proc Natl Acad Sci U S A ; 116(34): 16949-16954, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31391300

RESUMO

The advent of molecular tension probes for real-time mapping of piconewton forces in living systems has had a major impact on mechanobiology. For example, DNA-based tension probes have revealed roles for mechanics in platelet, B cell, T cell, and fibroblast function. Nonetheless, imaging short-lived forces transmitted by low-abundance receptors remains a challenge. This is a particular problem for mechanoimmunology where ligand-receptor bindings are short lived, and a few antigens are sufficient for cell triggering. Herein, we present a mechanoselection strategy that uses locking oligonucleotides to preferentially and irreversibly bind DNA probes that are mechanically strained over probes at rest. Thus, infrequent and short-lived mechanical events are tagged. This strategy allows for integration and storage of mechanical information into a map of molecular tension history. Upon addition of unlocking oligonucleotides that drive toehold-mediated strand displacement, the probes reset to the real-time state, thereby erasing stored mechanical information. As a proof of concept, we applied this strategy to study OT-1 T cells, revealing that the T cell receptor (TCR) mechanically samples antigens carrying single amino acid mutations. Such events are not detectable using conventional tension probes. Each mutant peptide ligand displayed a different level of mechanical sampling and spatial scanning by the TCR that strongly correlated with its functional potency. Finally, we show evidence that T cells transmit pN forces through the programmed cell death receptor-1 (PD1), a major target in cancer immunotherapy. We anticipate that mechanical information storage will be broadly useful in studying the mechanobiology of the immune system.


Assuntos
Antígenos , Sondas de DNA , Mecanotransdução Celular , Peptídeos , Receptores de Antígenos de Linfócitos T , Linfócitos T , Antígenos/química , Antígenos/genética , Antígenos/imunologia , Linhagem Celular , Sondas de DNA/química , Sondas de DNA/genética , Sondas de DNA/imunologia , Humanos , Mecanotransdução Celular/genética , Mecanotransdução Celular/imunologia , Mutação , Peptídeos/química , Peptídeos/genética , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/química , Linfócitos T/citologia , Linfócitos T/imunologia
8.
Proc Natl Acad Sci U S A ; 115(5): 992-997, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29343642

RESUMO

While cell-based immunotherapy, especially chimeric antigen receptor (CAR)-expressing T cells, is becoming a paradigm-shifting therapeutic approach for cancer treatment, there is a lack of general methods to remotely and noninvasively regulate genetics in live mammalian cells and animals for cancer immunotherapy within confined local tissue space. To address this limitation, we have identified a mechanically sensitive Piezo1 ion channel (mechanosensor) that is activatable by ultrasound stimulation and integrated it with engineered genetic circuits (genetic transducer) in live HEK293T cells to convert the ultrasound-activated Piezo1 into transcriptional activities. We have further engineered the Jurkat T-cell line and primary T cells (peripheral blood mononuclear cells) to remotely sense the ultrasound wave and transduce it into transcriptional activation for the CAR expression to recognize and eradicate target tumor cells. This approach is modular and can be extended for remote-controlled activation of different cell types with high spatiotemporal precision for therapeutic applications.


Assuntos
Imunoterapia/métodos , Neoplasias/terapia , Animais , Fenômenos Biomecânicos , Sinalização do Cálcio , Genes Sintéticos , Engenharia Genética , Técnicas Genéticas , Células HEK293 , Humanos , Canais Iônicos/genética , Canais Iônicos/imunologia , Células Jurkat , Mecanotransdução Celular/genética , Mecanotransdução Celular/imunologia , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Biologia Sintética , Linfócitos T/imunologia , Ultrassom
9.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34638778

RESUMO

Inflammatory Bowel Disease (IBD) comprises a series of chronic and relapsing intestinal diseases, with Crohn's disease and ulcerative colitis being the most common. The abundant and uncontrolled deposition of extracellular matrix, namely fibrosis, is one of the major hallmarks of IBD and is responsible for the progressive narrowing and closure of the intestine, defined as stenosis. Although fibrosis is usually considered the product of chronic inflammation, the substantial failure of anti-inflammatory therapies to target and reduce fibrosis in IBD suggests that fibrosis might be sustained in an inflammation-independent manner. Pharmacological therapies targeting integrins have recently shown great promise in the treatment of IBD. The efficacy of these therapies mainly relies on their capacity to target the integrin-mediated recruitment and functionality of the immune cells at the damage site. However, by nature, integrins also act as mechanosensitive molecules involved in the intracellular transduction of signals and modifications originating from the extracellular matrix. Therefore, understanding integrin signaling in the context of IBD may offer important insights into mechanisms of matrix remodeling, which are uncoupled from inflammation and could underlie the onset and persistency of intestinal fibrosis. In this review, we present the currently available knowledge on the role of integrins in the etiopathogenesis of IBD, highlighting their role in the context of immune-dependent and independent mechanisms.


Assuntos
Matriz Extracelular/imunologia , Doenças Inflamatórias Intestinais/imunologia , Integrinas/imunologia , Mecanotransdução Celular/imunologia , Animais , Matriz Extracelular/patologia , Fibrose , Humanos , Doenças Inflamatórias Intestinais/patologia
10.
Mol Med ; 26(1): 95, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-33054759

RESUMO

Pulmonary fibrosis arises from the repeated epithelial mild injuries and insufficient repair lead to over activation of fibroblasts and excessive deposition of extracellular matrix, which result in a mechanical stretched niche. However, increasing mechanical stress likely exists before the establishment of fibrosis since early micro injuries increase local vascular permeability and prompt cytoskeletal remodeling which alter cellular mechanical forces. It is noteworthy that COVID-19 patients with severe hypoxemia will receive mechanical ventilation as supportive treatment and subsequent pathology studies indicate lung fibrosis pattern. At advanced stages, mechanical stress originates mainly from the stiff matrix since boundaries between stiff and compliant parts of the tissue could generate mechanical stress. Therefore, mechanical stress has a significant role in the whole development process of pulmonary fibrosis. The alveoli are covered by abundant capillaries and function as the main gas exchange unit. Constantly subject to variety of damages, the alveolar epithelium injuries were recently recognized to play a vital role in the onset and development of idiopathic pulmonary fibrosis. In this review, we summarize the literature regarding the effects of mechanical stress on the fundamental cells constituting the alveoli in the process of pulmonary fibrosis, particularly on epithelial cells, capillary endothelial cells, fibroblasts, mast cells, macrophages and stem cells. Finally, we briefly review this issue from a more comprehensive perspective: the metabolic and epigenetic regulation.


Assuntos
Infecções por Coronavirus/imunologia , Epigênese Genética/imunologia , Fibrose Pulmonar Idiopática/imunologia , Mecanotransdução Celular/imunologia , Pneumonia Viral/imunologia , Embolia Pulmonar/imunologia , Insuficiência Respiratória/imunologia , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/patologia , Betacoronavirus/imunologia , Betacoronavirus/patogenicidade , Fenômenos Biomecânicos , COVID-19 , Infecções por Coronavirus/genética , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Citocinas/genética , Citocinas/imunologia , Células Endoteliais/imunologia , Células Endoteliais/patologia , Fibroblastos/imunologia , Fibroblastos/patologia , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/patologia , Fibrose Pulmonar Idiopática/virologia , Pulmão/irrigação sanguínea , Pulmão/imunologia , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/patologia , Mecanotransdução Celular/genética , Pandemias , Pneumonia Viral/genética , Pneumonia Viral/patologia , Pneumonia Viral/virologia , Embolia Pulmonar/genética , Embolia Pulmonar/patologia , Embolia Pulmonar/virologia , Insuficiência Respiratória/genética , Insuficiência Respiratória/patologia , Insuficiência Respiratória/virologia , SARS-CoV-2 , Estresse Mecânico
11.
Blood ; 131(7): 787-796, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29203584

RESUMO

Immune thrombocytopenia (ITP) is a prevalent autoimmune disease characterized by autoantibody-induced platelet clearance. Some ITP patients are refractory to standard immunosuppressive treatments such as intravenous immunoglobulin (IVIg). These patients often have autoantibodies that target the ligand-binding domain (LBD) of glycoprotein Ibα (GPIbα), a major subunit of the platelet mechanoreceptor complex GPIb-IX. However, the molecular mechanism of this Fc-independent platelet clearance is not clear. Here, we report that many anti-LBD monoclonal antibodies such as 6B4, but not AK2, activated GPIb-IX in a shear-dependent manner and induced IVIg-resistant platelet clearance in mice. Single-molecule optical tweezer measurements of antibodies pulling on full-length GPIb-IX demonstrated that the unbinding force needed to dissociate 6B4 from the LBD far exceeds the force required to unfold the juxtamembrane mechanosensory domain (MSD) in GPIbα, unlike the AK2-LBD unbinding force. Binding of 6B4, not AK2, induced shear-dependent unfolding of the MSD on the platelet, as evidenced by increased exposure of a linear sequence therein. Imaging flow cytometry and aggregometry measurements of platelets and LBD-coated platelet-mimetic beads revealed that 6B4 can sustain crosslinking of platelets under shear, whereas 6B4 Fab and AK2 cannot. These results suggest a novel mechanism by which anti-LBD antibodies can exert a pulling force on GPIb-IX via platelet crosslinking, activating GPIb-IX by unfolding its MSD and inducing Fc-independent platelet clearance.


Assuntos
Plaquetas/efeitos dos fármacos , Fragmentos Fc das Imunoglobulinas/farmacologia , Imunoglobulinas Intravenosas/farmacologia , Mecanotransdução Celular/efeitos dos fármacos , Púrpura Trombocitopênica Idiopática/sangue , Púrpura Trombocitopênica Idiopática/etiologia , Animais , Anticorpos Monoclonais/farmacologia , Plaquetas/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/fisiologia , Mecanotransdução Celular/imunologia , Camundongos , Camundongos Transgênicos , Complexo Glicoproteico GPIb-IX de Plaquetas/imunologia , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Púrpura Trombocitopênica Idiopática/imunologia , Resistência ao Cisalhamento/efeitos dos fármacos , Resistência ao Cisalhamento/fisiologia , Transdução de Sinais/efeitos dos fármacos
12.
Stem Cells ; 37(7): 844-854, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30913336

RESUMO

Pluripotent stem cells (PSCs) are an attractive, reliable source for generating functional cardiomyocytes for regeneration of infarcted heart. However, inefficient cell engraftment into host tissue remains a notable challenge to therapeutic success due to mechanical damage or relatively inhospitable microenvironment. Evidence has shown that excessively formed scar tissues around cell delivery sites present as mechanical and biological barriers that inhibit migration and engraftment of implanted cells. In this review, we focus on the functional responses of stem cells and cardiomyocytes during the process of cardiac fibrosis and scar formation. Survival, migration, contraction, and coupling function of implanted cells may be affected by matrix remodeling, inflammatory factors, altered tissue stiffness, and presence of electroactive myofibroblasts in the fibrotic microenvironment. Although paracrine factors from implanted cells can improve cardiac fibrosis, the transient effect is insufficient for complete repair of an infarcted heart. Furthermore, investigation of interactions between implanted cells and fibroblasts including myofibroblasts helps the identification of new targets to optimize the host substrate environment for facilitating cell engraftment and functional integration. Several antifibrotic approaches, including the use of pharmacological agents, gene therapies, microRNAs, and modified biomaterials, can prevent progression of heart failure and have been developed as adjunct therapies for stem cell-based regeneration. Investigation and optimization of new biomaterials is also required to enhance cell engraftment of engineered cardiac tissue and move PSCs from a laboratory setting into translational medicine.


Assuntos
Cicatriz/prevenção & controle , Terapia Genética/métodos , Infarto do Miocárdio/terapia , Células-Tronco Pluripotentes/citologia , Transplante de Células-Tronco/métodos , Engenharia Tecidual/métodos , Animais , Cicatriz/genética , Cicatriz/imunologia , Cicatriz/patologia , Modelos Animais de Doenças , Fibrose , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Mecanotransdução Celular/genética , Mecanotransdução Celular/imunologia , MicroRNAs/genética , MicroRNAs/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Comunicação Parácrina/genética , Comunicação Parácrina/imunologia , Células-Tronco Pluripotentes/metabolismo , Regeneração/genética , Regeneração/imunologia
13.
Crit Rev Immunol ; 39(3): 165-173, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32421961

RESUMO

A growing number of studies and case series point to a dark side of immune checkpoint inhibitors, as they may cause rapid tumor growth with potentially deleterious effects. The pathophysiological mechanism of hyper-progressive disease (HPD) is still unknown; in addition, there are no reliable predictive biomarkers that facilitate the process of patient selection for immunomodulatory antibody-based therapy. The proposed model attempts to reveal the mechanism of such paradoxical response, in a subset of patients receiving anti-PD1/PD-L1 immunotherapy, depending on the biomechanistic properties of the crystal structure of PD-1 protein. PD-1 can exhibit a signaling pattern depending on mechanotransduction upon the formation of PD-1/monoclonal antibody (mAb)/Fc-gamma receptor (Fc-γR) axes resulting from the interaction between PD-1/mAb complex, on the surface of tumor-specific T-cells, and Fc-γR-bearing tumor-associated macrophages (TAMs) within the tumor microenvironment. The generated mechanical force activates ITIM and ITSM on the cytoplasmic endodomain of the PD-1 receptor, leading to suppression of the effector function of tumor-specific T-cells which effectively unleashes cancer cells from the cytotoxic barrier and causes HPD in affected patients. This model provides clues about why patients receiving anti-PD1/PD-L1 mAbs are more prone to develop HPD as well as the variability of the ICIs response among treated patients. Additionally, it features the effect of specific immunophenotypic dynamics, such as TAM infiltration, on the final outcome of antibody-based immunotherapy and gives new insights for designing next-generation immunomodulatory interventions.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunoterapia/métodos , Mecanotransdução Celular/imunologia , Neoplasias/terapia , Animais , Antígeno B7-H1/metabolismo , Humanos , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Conformação Proteica , Transdução de Sinais , Relação Estrutura-Atividade , Resultado do Tratamento
14.
J Immunol ; 200(4): 1255-1260, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29330322

RESUMO

TCRs recognize peptides on MHC molecules and induce downstream signaling, leading to activation and clonal expansion. In addition to the strength of the interaction of TCRs with peptides on MHC molecules, mechanical forces contribute to optimal T cell activation, as reflected by the superior efficiency of immobilized TCR-cross-linking Abs compared with soluble Abs in TCR triggering, although a dedicated mechanotransduction module is not identified. We found that the professional mechanosensor protein Piezo1 is critically involved in human T cell activation. Although a deficiency in Piezo1 attenuates downstream events on ex vivo TCR triggering, a Piezo1 agonist can obviate the need to immobilize TCR-cross-linking Abs. Piezo1-driven Ca2+ influx, leading to calpain activation and organization of cortical actin scaffold, links this mechanosensor to optimal TCR signaling. Thus, we discovered a hitherto unknown regulatory mechanism for human T cell activation and provide the first evidence, to our knowledge, for the involvement of Piezo1 mechanosensors in immune regulation.


Assuntos
Canais Iônicos/imunologia , Ativação Linfocitária/imunologia , Mecanotransdução Celular/imunologia , Linfócitos T/imunologia , Humanos , Mecanorreceptores/imunologia
15.
Proc Natl Acad Sci U S A ; 114(39): E8204-E8213, 2017 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-28811364

RESUMO

T lymphocytes use surface [Formula: see text] T-cell receptors (TCRs) to recognize peptides bound to MHC molecules (pMHCs) on antigen-presenting cells (APCs). How the exquisite specificity of high-avidity T cells is achieved is unknown but essential, given the paucity of foreign pMHC ligands relative to the ubiquitous self-pMHC array on an APC. Using optical traps, we determine physicochemical triggering thresholds based on load and force direction. Strikingly, chemical thresholds in the absence of external load require orders of magnitude higher pMHC numbers than observed physiologically. In contrast, force applied in the shear direction ([Formula: see text]10 pN per TCR molecule) triggers T-cell Ca2+ flux with as few as two pMHC molecules at the interacting surface interface with rapid positional relaxation associated with similarly directed motor-dependent transport via [Formula: see text]8-nm steps, behaviors inconsistent with serial engagement during initial TCR triggering. These synergistic directional forces generated during cell motility are essential for adaptive T-cell immunity against infectious pathogens and cancers.


Assuntos
Apresentação de Antígeno/imunologia , Ativação Linfocitária/imunologia , Mecanotransdução Celular/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos/imunologia , Linhagem Celular , Camundongos , Camundongos Knockout , Pinças Ópticas
16.
Proc Natl Acad Sci U S A ; 114(21): E4175-E4183, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28490501

RESUMO

T-cell receptor (TCR) triggering and subsequent T-cell activation are essential for the adaptive immune response. Recently, multiple lines of evidence have shown that force transduction across the TCR complex is involved during TCR triggering, and that the T cell might use its force-generation machinery to probe the mechanical properties of the opposing antigen-presenting cell, giving rise to different signaling and physiological responses. Mechanistically, actin polymerization and turnover have been shown to be essential for force generation by T cells, but how these actin dynamics are regulated spatiotemporally remains poorly understood. Here, we report that traction forces generated by T cells are regulated by dynamic microtubules (MTs) at the interface. These MTs suppress Rho activation, nonmuscle myosin II bipolar filament assembly, and actin retrograde flow at the T-cell-substrate interface. Our results suggest a novel role of the MT cytoskeleton in regulating force generation during T-cell activation.


Assuntos
Actomiosina/metabolismo , Ativação Linfocitária/imunologia , Mecanotransdução Celular/imunologia , Microtúbulos/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Citoesqueleto de Actina/metabolismo , Imunidade Adaptativa/imunologia , Células Apresentadoras de Antígenos/imunologia , Linhagem Celular , Humanos , Células Jurkat , Proteínas rho de Ligação ao GTP/metabolismo
17.
Ann Rheum Dis ; 78(6): 787-795, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30928902

RESUMO

OBJECTIVES: The mechanisms driving onset of joint inflammation in arthritides such as rheumatoid arthritis and spondyloarthritis and the conversion to disease chronicity are poorly understood. We hypothesised mechanostrain could play an instrumental role herein by engaging local and/or systemic pathways, thereby attenuating disease course and outcome. METHODS: The development of collagen antibody-induced arthritis (CAIA) in C57BL/6 mice was evaluated both clinically and histologically under different loading regimens: control, voluntary running or hindpaw unloading. Bone surface porosity was quantified by high-resolution µ-CT. Gene expression analyses were conducted by microarrays and qPCR on microdissected entheses, murine and human synovial tissues (both normal and inflamed). Serum cytokines and chemokines were measured by ELISA. The influence of complement activation and T regulatory (Treg) cell function on the induction and resolution phase of disease was studied by respectively pharmacological modulation and conditional Treg depletion. RESULTS: Voluntary running strongly impacts the course of arthritis by impairing the resolution phase of CAIA, leading to more persistent inflammation and bone surface porosity. Mechanical strain induced local complement activation, increased danger-associated molecular pattern expression, activating Fcγ receptors as well as changes in fibroblast phenotype. Interestingly, complement C5a receptor blockade inhibited the enhanced joint pathology caused by voluntary running. Moreover, Treg depletion led to a loss of disease resolution in CAIA mice, which was not observed under voluntary running conditions. CONCLUSIONS: Running promotes onset and chronicity of arthritis by local upregulation of complement activators and hampering regulatory T cell feedback loops.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Ativação do Complemento/fisiologia , Corrida/fisiologia , Linfócitos T Reguladores/imunologia , Adulto , Idoso , Animais , Artrite Experimental/fisiopatologia , Artrite Reumatoide/fisiopatologia , Doença Crônica , Progressão da Doença , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Mecanotransdução Celular/imunologia , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Properdina/biossíntese , Estresse Mecânico , Membrana Sinovial/metabolismo
18.
Int Immunol ; 30(6): 267-278, 2018 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-29800294

RESUMO

Macrophages participate in immunity, tissue repair and tissue homeostasis. Activation of Toll-like receptors (TLRs) by conserved exogenous or endogenous structures initiates signaling cascades that result in the release of cytokines such as tumor necrosis factor α (TNFα). Extracellular substrate stiffness is known to regulate functions of non-immune cells through a process called mechanotransduction, yet less is known about how physical cues affect macrophage function or TLR signaling. To investigate this question, we cultured murine primary bone marrow-derived macrophages (BMMs) and RAW264.7 cells on fibronectin-coated polyacrylamide (PA) gels of defined stiffnesses (1, 20 and 150 kPa) that approximate the physical properties of physiologic tissues. BMMs on all gels were smaller and more circular than those on rigid glass. Macrophages on intermediate stiffness 20 kPa PA gels were slightly larger and less circular than those on either 1 or 150 kPa. Secretion of the pro-inflammatory cytokine, TNFα, in response to stimulation of TLR4 and TLR9 was increased in macrophages grown on soft gels versus more rigid gels, particularly for BMMs. Inhibition of the rho-associated coiled-coil kinase 1/2 (ROCK1/2), key mediators in cell contractility and mechanotransduction, enhanced release of TNFα in response to stimulation of TLR4. ROCK1/2 inhibition enhanced phosphorylation of the TLR downstream signaling molecules, p38, ERK1/2 and NFκB. Our data indicate that physical cues from the extracellular environment regulate macrophage morphology and TLR signaling. These findings have important implications in the regulation of macrophage function in diseased tissues and offer a novel pharmacological target for the manipulation of macrophage function in vivo.


Assuntos
Macrófagos/enzimologia , Macrófagos/imunologia , Mecanotransdução Celular/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Quinases Associadas a rho/metabolismo , Resinas Acrílicas/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Quinases Associadas a rho/antagonistas & inibidores
19.
Biochem Biophys Res Commun ; 491(3): 629-635, 2017 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-28760342

RESUMO

Acupuncture therapy is performed by applying the needle insertion at discrete cutaneous locations and used for the treatments of diverse symptoms and disorders. In order to elucidate mechanistic basis on how acupuncture stimulation (AS) produces therapeutic effects, it is primarily important to understand tissue responses locally at the acupuncture site (acupoint). Here, we investigated integrin protein as molecular target responding to and integrating AS. Signals of α6 and ß1 integrins were clearly induced at zusanli acupoint 24 h after AS in areas of nuclear clusters around the needle track. Induction levels of integrin were largely reduced by needle insertion at non-acupuncture point or without needle rotation. Phospho-Erk1/2 was initially decreased below the basal level after AS but increased 24 h later. Induction pattern of phospho-Erk1/2 was as similar as that of α6 integrin in its selectivity to needling procedure and tissue distribution. We further found that mRNA expression of P2X3 purinergic receptor was upregulated in the dorsal root ganglion (DRG) after AS, but decreased by the inhibition of Erk1/2 activity at the acupuncture area. Moreover, AS-mediated integrin activation was required for Erk1/2 activation at the acupuncture site and regulation of pain sensitivity in the hind paw. The present results provide a new evidence on acupuncture-specific tissue response in terms of integrin induction, and further suggest that integrin activation may be involved in transmitting mechanosensory signals from the acupoint to afferent nerve fiber.


Assuntos
Terapia por Acupuntura/métodos , Integrina alfa6/imunologia , Integrina beta1/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Neuralgia/imunologia , Neuralgia/terapia , Pontos de Acupuntura , Animais , Masculino , Mecanotransdução Celular/imunologia , Neuralgia/diagnóstico , Estimulação Física/métodos , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
20.
Immunol Invest ; 46(1): 22-37, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27617892

RESUMO

The role of cytokines as regulators of stretch-related mechanisms is of special importance since mechano-sensitivity plays an important role in a wide variety of biological processes. Here, we elucidate the influence of cytokine application on mechano-sensitivity and mechano-transduction. The atrial myocardial stretch induces production of interleukin (IL)-2, IL-6, IL-13, IL-17A, and IL-18 with exception of tumor necrosis factor α (TNF-α), IL-1ß, and vascular endothelial growth factor B (VEGF-B). Positive ionotropic effect was specific for VEGF-B, negative ionotropic effects were specific for TNF-α, IL-1ß, IL-2, IL-6, IL-13, IL-17A and IL-18, while IL-1α doesn't show direct ionotropic effect. The IL-2, IL-6, IL-17A, IL-18, and VEGF-B cause elongation of the APD, in comparison with the reduced APD caused by the IL-13. The TNF-α, IL-1ß, and IL-18 influences L-type Ca2+ channels, IL-2 has an inhibitory effect on the fast Na+ channels while IL-17A and VEGF-B were specific for Kir channels. With exception of the IL-1α, IL-2, and VEGF-B, all analyzed cytokines include nitric oxide dependent signaling with resultant combined effects on mechano-gated and Ca2+ channels. The relationships between these pathways and the time-dependence of their activation are of important considerations in the evaluation of cytokine-induced electrical abnormality, specific for cardiac dysfunctions. In general, the discussion presented in this review covers research devoted to counterbalance between different cytokines in the regulation of stretch-induced effects in rat atrial myocardium. ABBREVIATIONS: APs: action potentials; APD25: action potential durations to 25% of re-polarization; APD50: action potential durations to 50% of repolarization; APD90: action potential durations to 90% of repolarization; MGCs: mechanically gated channels.


Assuntos
Citocinas/imunologia , Citocinas/metabolismo , Átrios do Coração/patologia , Mecanotransdução Celular/imunologia , Contração Miocárdica , Miocárdio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Potenciais de Ação/imunologia , Animais , Função Atrial/imunologia , Condutividade Elétrica , Eletricidade , Humanos , Miocárdio/imunologia , Ratos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa