Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.544
Filtrar
3.
Front Immunol ; 15: 1424081, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39040108

RESUMO

Exosomes are found in various tissues of the body and carry abundant contents including nucleic acids, proteins, and metabolites, which continuously flow between cells of various tissues and mediate important intercellular communication. In addition, exosomes from different cellular sources possess different physiopathological immunomodulatory effects, which are closely related to the immune regeneration of normal or abnormal organs and tissues. Here, we focus on the mechanistic interactions between exosomes and the human immune system, introduce the immuno-regenerative therapeutic potential of exosomes in common clinical immune-related diseases, such as infectious diseases, autoimmune diseases, and tumors, and reveal the safety and efficacy of exosomes as a novel cell-free immune regenerative therapy.


Assuntos
Exossomos , Imunoterapia , Exossomos/imunologia , Exossomos/metabolismo , Humanos , Imunoterapia/métodos , Animais , Neoplasias/terapia , Neoplasias/imunologia , Comunicação Celular/imunologia , Imunomodulação , Doenças Autoimunes/terapia , Doenças Autoimunes/imunologia
4.
Front Immunol ; 15: 1362120, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38962016

RESUMO

Cancer stem cells (CSCs), accounting for only a minor cell proportion (< 1%) within tumors, have profound implications in tumor initiation, metastasis, recurrence, and treatment resistance due to their inherent ability of self-renewal, multi-lineage differentiation, and tumor-initiating potential. In recent years, accumulating studies indicate that CSCs and tumor immune microenvironment act reciprocally in driving tumor progression and diminishing the efficacy of cancer therapies. Extracellular vesicles (EVs), pivotal mediators of intercellular communications, build indispensable biological connections between CSCs and immune cells. By transferring bioactive molecules, including proteins, nucleic acids, and lipids, EVs can exert mutual influence on both CSCs and immune cells. This interaction plays a significant role in reshaping the tumor immune microenvironment, creating conditions favorable for the sustenance and propagation of CSCs. Deciphering the intricate interplay between CSCs and immune cells would provide valuable insights into the mechanisms of CSCs being more susceptible to immune escape. This review will highlight the EV-mediated communications between CSCs and each immune cell lineage in the tumor microenvironment and explore potential therapeutic opportunities.


Assuntos
Vesículas Extracelulares , Neoplasias , Células-Tronco Neoplásicas , Microambiente Tumoral , Microambiente Tumoral/imunologia , Humanos , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Animais , Comunicação Celular/imunologia , Evasão Tumoral , Imunomodulação
5.
Front Immunol ; 15: 1397967, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38947317

RESUMO

Introduction: CD39 plays an important role in the immunoregulation and inhibition of effector cells. It is expressed on immune cells, including Tregs, and on extracellular vesicles (EVs) budding from the plasma membrane. Platelet transfusion may induce alloimmunization against HLA-I antigens, leading to refractoriness to platelet transfusion with severe consequences for patients. Tregs may play a key role in determining whether alloimmunization occurs in patients with hematologic disorders. We hypothesized that CD39+ EVs might play an immunoregulatory role, particularly in the context of platelet transfusions in patients with hematologic disorders. Such alloimmunization leads to the production of alloantibodies and is sensitive to the regulatory action of CD39. Methods: We characterized CD39+ EVs in platelet concentrates by flow cytometry. The absolute numbers and cellular origins of CD39+ EVs were evaluated. We also performed functional tests to evaluate interactions with immune cells and their functions. Results: We found that CD39+ EVs from platelet concentrates had an inhibitory phenotype that could be transferred to the immune cells with which they interacted: CD4+ and CD8+ T lymphocytes (TLs), dendritic cells, monocytes, and B lymphocytes (BLs). Moreover, the concentration of CD39+ EVs in platelet concentrates varied and was very high in 10% of concentrates. The number of these EVs present was determinant for EV-cell interactions. Finally, functional interactions were observed with BLs, CD4+ TLs and CD39+ EVs for immunoglobulin production and lymphoproliferation, with potential implications for the immunological management of patients.


Assuntos
Plaquetas , Vesículas Extracelulares , Tetraspanina 29 , Humanos , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Plaquetas/imunologia , Plaquetas/metabolismo , Tetraspanina 29/metabolismo , Comunicação Celular/imunologia , Transfusão de Plaquetas , Feminino , Linfócitos B/imunologia , Linfócitos B/metabolismo , Masculino , Apirase/metabolismo , Apirase/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Antígenos CD
6.
Ups J Med Sci ; 1292024.
Artigo em Inglês | MEDLINE | ID: mdl-38863724

RESUMO

Cancer-associated fibroblasts (CAFs) are a heterogeneous cell population recognized as a key component of the tumour microenvironment (TME). Cancer-associated fibroblasts are known to play an important role in maintaining and remodelling the extracellular matrix (ECM) in the tumour stroma, supporting cancer progression and inhibiting the immune system's response against cancer cells. This review aims to summarize the immunomodulatory roles of CAFs, particularly focussing on their T-cell suppressive effects. Cancer-associated fibroblasts have several ways by which they can affect the tumour's immune microenvironment (TIME). For example, their interactions with macrophages and dendritic cells (DCs) create an immunosuppressive milieu that can indirectly affect T-cell anticancer immunity and enable immune evasion. In addition, a number of recent studies have confirmed CAF-mediated direct suppressive effects on T-cell anticancer capacity through ECM remodelling, promoting the expression of immune checkpoints, cytokine secretion and the release of extracellular vesicles. The consequential impact of CAFs on T-cell function is then reflected in affecting T-cell proliferation and apoptosis, migration and infiltration, differentiation and exhaustion. Emerging evidence highlights the existence of specific CAF subsets with distinct capabilities to modulate the immune landscape of TME in various cancers, suggesting the possibility of their exploitation as possible prognostic biomarkers and therapeutic targets.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias , Linfócitos T , Microambiente Tumoral , Humanos , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/imunologia , Microambiente Tumoral/imunologia , Linfócitos T/imunologia , Neoplasias/imunologia , Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Matriz Extracelular/metabolismo , Comunicação Celular/imunologia , Células Dendríticas/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo
7.
Front Immunol ; 15: 1388176, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38840908

RESUMO

The tumor microenvironment is closely linked to the initiation, promotion, and progression of solid tumors. Among its constitutions, immunologic cells emerge as critical players, facilitating immune evasion and tumor progression. Apart from their indirect impact on anti-tumor immunity, immunocytes directly influence neoplastic cells, either bolstering or impeding tumor advancement. However, current therapeutic modalities aimed at alleviating immunosuppression from regulatory cells on effector immune cell populations may not consistently yield satisfactory results in various solid tumors, such as breast carcinoma, colorectal cancer, etc. Therefore, this review outlines and summarizes the direct, dualistic effects of immunocytes such as T cells, innate lymphoid cells, B cells, eosinophils, and tumor-associated macrophages on tumor cells within the tumor microenvironment. The review also delves into the underlying mechanisms involved and presents the outcomes of clinical trials based on these direct effects, aiming to propose innovative and efficacious therapeutic strategies for addressing solid tumors.


Assuntos
Neoplasias , Microambiente Tumoral , Humanos , Microambiente Tumoral/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/patologia , Animais , Imunidade Inata , Comunicação Celular/imunologia , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo , Evasão Tumoral , Imunoterapia/métodos
8.
Front Immunol ; 15: 1403150, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38873597

RESUMO

The interplay between myeloid cells and T-lymphocytes is critical to the regulation of host defense and inflammation resolution. Dysregulation of this interaction can contribute to the development of chronic inflammatory diseases. Important among these diseases is atherosclerosis, which refers to focal lesions in the arterial intima driven by elevated apolipoprotein B-containing lipoproteins, notably low-density lipoprotein (LDL), and characterized by the formation of a plaque composed of inflammatory immune cells, a collection of dead cells and lipids called the necrotic core, and a fibrous cap. As the disease progresses, the necrotic core expands, and the fibrous cap becomes thin, which increases the risk of plaque rupture or erosion. Plaque rupture leads to a rapid thrombotic response that can give rise to heart attack, stroke, or sudden death. With marked lowering of circulating LDL, however, plaques become more stable and cardiac risk is lowered-a process known as atherosclerosis regression. A critical aspect of both atherosclerosis progression and regression is the crosstalk between innate (myeloid cells) and adaptive (T-lymphocytes) immune cells. Myeloid cells are specialized at clearing apoptotic cells by a process called efferocytosis, which is necessary for inflammation resolution. In advanced disease, efferocytosis is impaired, leading to secondary necrosis of apoptotic cells, inflammation, and, most importantly, defective tissue resolution. In regression, efferocytosis is reawakened aiding in inflammation resolution and plaque stabilization. Here, we will explore how efferocytosing myeloid cells could affect T-cell function and vice versa through antigen presentation, secreted factors, and cell-cell contacts and how this cellular crosstalk may contribute to the progression or regression of atherosclerosis.


Assuntos
Aterosclerose , Células Mieloides , Linfócitos T , Humanos , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , Animais , Comunicação Celular/imunologia , Fagocitose , Apoptose , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia
9.
Immun Inflamm Dis ; 12(6): e1325, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38934401

RESUMO

OBJECTIVE: Asthma, a chronic inflammatory disease with diverse pathomechanisms, presents challenges in developing personalized diagnostic and therapeutic approaches. This review aims to provide a comprehensive overview of the role of exosomes, small extracellular vesicles, in asthma pathophysiology and explores their potential as diagnostic biomarkers and therapeutic tools. METHODS: A literature search was conducted to identify recent studies investigating the involvement of exosomes in asthma. The retrieved articles were analyzed to extract relevant information on the role of exosomes in maintaining lung microenvironment homeostasis, regulating inflammatory responses, and their diagnostic and therapeutic potential for asthma. RESULTS: Exosomes secreted by various cell types, have emerged as crucial mediators of intercellular communication in healthy and diseased conditions. Evidence suggest that exosomes play a significant role in maintaining lung microenvironment homeostasis and contribute to asthma pathogenesis by regulating inflammatory responses. Differential exosomal content between healthy individuals and asthmatics holds promise for the development of novel asthma biomarkers. Furthermore, exosomes secreted by immune and nonimmune cells, as well as those detected in biofluids, demonstrate potential in promoting or regulating immune responses, making them attractive candidates for designing new treatment strategies for inflammatory conditions such as asthma. CONCLUSION: Exosomes, with their ability to modulate immune responses and deliver therapeutic cargo, offer potential as targeted therapeutic tools in asthma management. Further research and clinical trials are required to fully understand the mechanisms underlying exosome-mediated effects and translate these findings into effective diagnostic and therapeutic strategies for asthma patients.


Assuntos
Asma , Biomarcadores , Exossomos , Exossomos/metabolismo , Exossomos/imunologia , Humanos , Asma/imunologia , Asma/metabolismo , Asma/terapia , Asma/diagnóstico , Animais , Pulmão/imunologia , Pulmão/patologia , Pulmão/metabolismo , Comunicação Celular/imunologia
10.
Oncoimmunology ; 13(1): 2367843, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38887373

RESUMO

Conventional type 1 dendritic cells (cDC1) are critical regulators of anti-tumoral T-cell responses. The structure and abundance of intercellular contacts between cDC1 and CD8 T cells in cancer tissues is important to determine the outcome of the T-cell response. However, the molecular determinants controlling the stability of cDC1-CD8 interactions during cancer progression remain poorly investigated. Here, we generated a genetic model of non-small cell lung cancer crossed to a fluorescent cDC1 reporter (KP-XCR1venus) to allow the detection of cDC1-CD8T cell clusters in tumor tissues across tumor stages. We found that cDC1-CD8 clusters are abundant and productive at the early stages of tumor development but progressively diminish in advanced tumors. Transcriptional profiling and flow cytometry identified the adhesion molecule ALCAM/CD166 (Activated Leukocyte Cell Adhesion Molecule, ligand of CD6) as highly expressed by lung cDC1 and significantly downregulated in advanced tumors. Analysis of human datasets indicated that ALCAM is downregulated in non-small cell lung cancer and its expression correlates to better prognosis. Mechanistically, triggering ALCAM on lung cDC1 induces cytoskeletal remodeling and contact formation whereas its blockade prevents T-cell activation. Together, our results indicate that ALCAM is important to stabilize cDC1-CD8 interactions at early tumor stages, while its loss in advanced tumors contributes to immune evasion.


Assuntos
Antígenos CD , Linfócitos T CD8-Positivos , Carcinoma Pulmonar de Células não Pequenas , Células Dendríticas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Animais , Camundongos , Antígenos CD/metabolismo , Antígenos CD/genética , Antígenos CD/imunologia , Proteínas Fetais/metabolismo , Proteínas Fetais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Comunicação Celular/imunologia , Molécula de Adesão de Leucócito Ativado
11.
Adv Immunol ; 162: 23-58, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38866438

RESUMO

The intestine represents the most complex cellular network in the whole body. It is constantly faced with multiple types of immunostimulatory agents encompassing from food antigen, gut microbiome, metabolic waste products, and dead cell debris. Within the intestine, most T cells are found in three primary compartments: the organized gut-associated lymphoid tissue, the lamina propria, and the epithelium. The well-orchestrated epithelial-immune-microbial interaction is critically important for the precise immune response. The main role of intestinal mesenchymal stromal cells is to support a structural framework within the gut wall. However, recent evidence from stromal cell studies indicates that they also possess significant immunomodulatory functions, such as maintaining intestinal tolerance via the expression of PDL1/2 and MHC-II molecules, and promoting the development of CD103+ dendritic cells, and IgA+ plasma cells, thereby enhancing intestinal homeostasis. In this review, we will summarize the current understanding of CD8+ T cells and stromal cells alongside the intestinal tract and discuss the reciprocal interactions between T subsets and mesenchymal stromal cell populations. We will focus on how the tissue residency, migration, and function of CD8+ T cells could be potentially regulated by mesenchymal stromal cell populations and explore the molecular mediators, such as TGF-ß, IL-33, and MHC-II molecules that might influence these processes. Finally, we discuss the potential pathophysiological impact of such interaction in intestine hemostasis as well as diseases of inflammation, infection, and malignancies.


Assuntos
Linfócitos T CD8-Positivos , Homeostase , Células-Tronco Mesenquimais , Humanos , Células-Tronco Mesenquimais/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Mucosa Intestinal/imunologia , Comunicação Celular/imunologia , Intestinos/imunologia
12.
Int Immunopharmacol ; 137: 112442, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-38889508

RESUMO

Liver disease ranks as the eleventh leading cause of mortality, leading to approximately 2 million deaths annually worldwide. Neutrophils are a type of immune cell that are abundant in peripheral blood and play a vital role in innate immunity by quickly reaching the site of liver injury. They exert their influence on liver diseases through autocrine, paracrine, and immunomodulatory mechanisms. Extracellular vesicles, phospholipid bilayer vesicles, transport a variety of substances, such as proteins, nucleic acids, lipids, and pathogenic factors, for intercellular communication. They regulate cell communication and perform their functions by delivering biological information. Current research has revealed the involvement of the interaction between neutrophils and extracellular vesicles in the pathogenesis of liver disease. Moreover, more research has focused on targeting neutrophils as a therapeutic strategy to attenuate disease progression. Therefore, this article summarizes the roles of neutrophils, extracellular vesicles, and their interactions in noncancerous liver diseases.


Assuntos
Comunicação Celular , Vesículas Extracelulares , Hepatopatias , Neutrófilos , Humanos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Hepatopatias/imunologia , Hepatopatias/metabolismo , Hepatopatias/patologia , Animais , Comunicação Celular/imunologia , Imunidade Inata , Fígado/metabolismo , Fígado/patologia , Fígado/imunologia
13.
Front Immunol ; 15: 1394108, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799455

RESUMO

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by persistent synovial inflammation and progressive joint destruction. Macrophages are key effector cells that play a central role in RA pathogenesis through their ability to polarize into distinct functional phenotypes. An imbalance favoring pro-inflammatory M1 macrophages over anti-inflammatory M2 macrophages disrupts immune homeostasis and exacerbates joint inflammation. Multiple signaling pathways, including Notch, JAK/STAT, NF-κb, and MAPK, regulate macrophage polarization towards the M1 phenotype in RA. Metabolic reprogramming also contributes to this process, with M1 macrophages prioritizing glycolysis while M2 macrophages utilize oxidative phosphorylation. Redressing this imbalance by modulating macrophage polarization and metabolic state represents a promising therapeutic strategy. Furthermore, complex bidirectional interactions exist between synovial macrophages and fibroblast-like synoviocytes (FLS), forming a self-perpetuating inflammatory loop. Macrophage-derived factors promote aggressive phenotypes in FLS, while FLS-secreted mediators contribute to aberrant macrophage activation. Elucidating the signaling networks governing macrophage polarization, metabolic adaptations, and crosstalk with FLS is crucial to developing targeted therapies that can restore immune homeostasis and mitigate joint pathology in RA.


Assuntos
Artrite Reumatoide , Fibroblastos , Ativação de Macrófagos , Macrófagos , Transdução de Sinais , Membrana Sinovial , Humanos , Artrite Reumatoide/metabolismo , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/imunologia , Membrana Sinovial/patologia , Fibroblastos/metabolismo , Fibroblastos/imunologia , Animais , Ativação de Macrófagos/imunologia , Comunicação Celular/imunologia , Reprogramação Metabólica
14.
Trends Immunol ; 45(6): 428-441, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38763820

RESUMO

Chronic obstructive pulmonary disease (COPD) is characterized by infiltration of the airways and lung parenchyma by inflammatory cells. Lung pathology results from the cumulative effect of complex and aberrant interactions between multiple cell types. However, three cell types, natural killer cells (NK), dendritic cells (DCs), and regulatory T cells (Tregs), are understudied and underappreciated. We propose that their mutual interactions significantly contribute to the development of COPD. Here, we highlight recent advances in NK, DC, and Treg biology with relevance to COPD, discuss their pairwise bidirectional interactions, and identify knowledge gaps that must be bridged to develop novel therapies. Understanding their interactions will be crucial for therapeutic use of autologous Treg, an approach proving effective in other diseases with immune components.


Assuntos
Comunicação Celular , Células Dendríticas , Células Matadoras Naturais , Doença Pulmonar Obstrutiva Crônica , Linfócitos T Reguladores , Animais , Humanos , Comunicação Celular/imunologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Linfócitos T Reguladores/imunologia
15.
Eur J Haematol ; 113(1): 4-15, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38698678

RESUMO

Chronic lymphocytic leukemia (CLL) is a mature-type B cell malignancy correlated with significant changes and defects in both the innate and adaptive arms of the immune system, together with a high dependency on the tumor microenvironment. Overall, the tumor microenvironment (TME) in CLL provides a supportive niche for leukemic cells to grow and survive, and interactions between CLL cells and the TME can contribute to disease progression and treatment resistance. Therefore, the increasing knowledge of the complicated interaction between immune cells and tumor cells, which is responsible for immune evasion and cancer progression, has provided an opportunity for the development of new therapeutic approaches. In this review, we outline tumor microenvironment-driven contributions to the licensing of immune escape mechanisms in CLL patients.


Assuntos
Comunicação Celular , Leucemia Linfocítica Crônica de Células B , Evasão Tumoral , Microambiente Tumoral , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/etiologia , Leucemia Linfocítica Crônica de Células B/patologia , Microambiente Tumoral/imunologia , Humanos , Comunicação Celular/imunologia , Animais , Suscetibilidade a Doenças
16.
Proc Natl Acad Sci U S A ; 121(22): e2322479121, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38771871

RESUMO

The significance of biochemical cues in the tumor immune microenvironment in affecting cancer metastasis is well established, but the role of physical factors in the microenvironment remains largely unexplored. In this article, we investigated how the mechanical interaction between cancer cells and immune cells, mediated by extracellular matrix (ECM), influences immune escape of cancer cells. We focus on the mechanical regulation of macrophages' targeting ability on two distinct types of colorectal carcinoma (CRC) cells with different metastatic potentials. Our results show that macrophages can effectively target CRC cells with low metastatic potential, due to the strong contraction exhibited by the cancer cells on the ECM, and that cancer cells with high metastatic potential demonstrated weakened contractions on the ECM and can thus evade macrophage attack to achieve immune escape. Our findings regarding the intricate mechanical interactions between immune cells and cancer cells can serve as a crucial reference for further exploration of cancer immunotherapy strategies.


Assuntos
Neoplasias Colorretais , Matriz Extracelular , Macrófagos , Evasão Tumoral , Microambiente Tumoral , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Macrófagos/imunologia , Humanos , Microambiente Tumoral/imunologia , Matriz Extracelular/metabolismo , Matriz Extracelular/imunologia , Linhagem Celular Tumoral , Metástase Neoplásica , Animais , Camundongos , Comunicação Celular/imunologia
17.
J Immunol ; 212(12): 1958-1970, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38700420

RESUMO

Fibroblasts acquire a proinflammatory phenotype in inflammatory bowel disease, but the factors driving this process and how fibroblasts contribute to mucosal immune responses are incompletely understood. TNF superfamily member 12 (TNFSF12, or TNF-like weak inducer of apoptosis [TWEAK]) has gained interest as a mediator of chronic inflammation. In this study, we explore its role as a driver of inflammatory responses in fibroblasts and its contribution to fibroblast-monocyte interaction using human primary colonic fibroblasts, THP-1 and primary monocytes. Recombinant human TWEAK induced the expression of cytokines, chemokines, and immune receptors in primary colonic fibroblasts. The TWEAK upregulated transcriptome shared 29% homology with a previously published transcriptional profile of inflammatory fibroblasts from ulcerative colitis. TWEAK elevated surface expression of activated fibroblast markers and adhesion molecules (podoplanin [PDPN], ICAM-1, and VCAM-1) and secretion of IL-6, CCL2, and CXCL10. In coculture, fibroblasts induced monocyte adhesion and secretion of CXCL1 and IL-8, and they promoted a CD14high/ICAM-1high phenotype in THP-1 cells, which was enhanced when fibroblasts were prestimulated with TWEAK. Primary monocytes in coculture with TWEAK-treated fibroblasts had altered surface expression of CD16 and triggering receptor expressed on myeloid cells-1 (TREM-1) as well as increased CXCL1 and CXCL10 secretion. Conversely, inhibition of the noncanonical NF-κB pathway on colonic fibroblasts with a NF-κB-inducing kinase small molecule inhibitor impaired their ability to induce a CD14high phenotype on monocytes. Our results indicate that TWEAK promotes an inflammatory fibroblast-monocyte crosstalk that may be amenable for therapeutic intervention.


Assuntos
Diferenciação Celular , Colo , Citocina TWEAK , Fibroblastos , Monócitos , Humanos , Citocina TWEAK/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Fibroblastos/metabolismo , Fibroblastos/imunologia , Colo/imunologia , Colo/patologia , Colo/metabolismo , Diferenciação Celular/imunologia , Comunicação Celular/imunologia , Inflamação/imunologia , Células THP-1 , Técnicas de Cocultura , Citocinas/metabolismo , Adesão Celular
18.
Cancer Immunol Res ; 12(7): 822-839, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38739030

RESUMO

Cytotoxic T lymphocytes (CTL) are pivotal in combating cancer, yet their efficacy is often hindered by the immunosuppressive tumor microenvironment, resulting in CTL exhaustion. This study investigates the role of interleukin-3 (IL3) in orchestrating antitumor immunity through CTL modulation. We found that intratumoral CTLs exhibited a progressive decline in IL3 production, which was correlated with impaired cytotoxic function. Augmenting IL3 supplementation, through intraperitoneal administration of recombinant IL3, IL3-expressing tumor cells, or IL3-engineered CD8+ T cells, conferred protection against tumor progression, concomitant with increased CTL activity. CTLs were critical for this therapeutic efficacy as IL3 demonstrated no impact on tumor growth in Rag1 knockout mice or following CD8+ T-cell depletion. Rather than acting directly, CTL-derived IL3 exerted its influence on basophils, concomitantly amplifying antitumor immunity within CTLs. Introducing IL3-activated basophils retarded tumor progression, whereas basophil depletion diminished the effectiveness of IL3 supplementation. Furthermore, IL3 prompted basophils to produce IL4, which subsequently elevated CTL IFNγ production and viability. Further, the importance of basophil-derived IL4 was evident from the absence of benefits of IL3 supplementation in IL4 knockout tumor-bearing mice. Overall, this research has unveiled a role for IL3-mediated CTL-basophil cross-talk in regulating antitumor immunity and suggests harnessing IL3 sustenance as a promising approach for optimizing and enhancing cancer immunotherapy. See related Spotlight, p. 798.


Assuntos
Interleucina-3 , Camundongos Knockout , Linfócitos T Citotóxicos , Animais , Camundongos , Linfócitos T Citotóxicos/imunologia , Interleucina-3/metabolismo , Interleucina-3/imunologia , Camundongos Endogâmicos C57BL , Linhagem Celular Tumoral , Microambiente Tumoral/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Comunicação Celular/imunologia , Humanos
19.
Gene ; 925: 148601, 2024 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-38788817

RESUMO

Tumor-derived exosomes (TDEs), as topologies of tumor cells, not only carry biological information from the mother, but also act as messengers for cellular communication. It has been demonstrated that TDEs play a key role in inducing an immunosuppressive tumor microenvironment (TME). They can reprogram immune cells indirectly or directly by delivering inhibitory proteins, cytokines, RNA and other substances. They not only inhibit the maturation and function of dendritic cells (DCs) and natural killer (NK) cells, but also remodel M2 macrophages and inhibit T cell infiltration to promote immunosuppression and create a favorable ecological niche for tumor growth, invasion and metastasis. Based on the specificity of TDEs, targeting TDEs has become a new strategy to monitor tumor progression and enhance treatment efficacy. This paper reviews the intricate molecular mechanisms underlying the immunosuppressive effects induced by TDEs to establish a theoretical foundation for cancer therapy. Additionally, the challenges of TDEs as a novel approach to tumor treatment are discussed.


Assuntos
Exossomos , Neoplasias , Microambiente Tumoral , Exossomos/imunologia , Exossomos/metabolismo , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Microambiente Tumoral/imunologia , Animais , Células Dendríticas/imunologia , Reprogramação Celular/imunologia , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Comunicação Celular/imunologia
20.
J Autoimmun ; 146: 103240, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38754238

RESUMO

BACKGROUND: Giant cell arteritis (GCA) is an immune-mediated large-vessels vasculitis with complex etiology. Although the pathogenic mechanisms remain poorly understood, a central role for CD4+ T cells has been demonstrated. In this context, understanding the transcriptome dysregulation in GCA CD4+ T cells will yield new insights into its pathogenesis. METHODS: Transcriptome analysis was conducted on CD4+ T cells from 70 patients with GCA with different disease activity and treatment status (active patients before treatment and patients in remission with and without glucocorticoid treatment), and 28 healthy controls. The study also evaluated potential impacts of DNA methylation on gene expression alterations and assessed cross-talk with CD14+ monocytes. RESULTS: This study has uncovered a substantial number of genes and pathways potentially contributing to the pathogenicity of CD4+ T cells in GCA. Specifically, CD4+ T cells from GCA patients with active disease exhibited altered expression levels of genes involved in multiple immune-related processes, including various interleukins (IL) signaling pathways. Notably, IL-2, a decisive interleukin for regulatory T cells homeostasis, was among the most significant. Additionally, impaired apoptotic pathways appear crucial in GCA development. Our findings also suggest that histone-related epigenetic pathways may be implicated in promoting an inflammatory phenotype in GCA active patients. Finally, our study observed altered signaling communication, such as the Jagged-Notch signaling, between CD4+ T cells and monocytes that could have pathogenic relevance in GCA. CONCLUSIONS: Our study suggests the participation of novel cytokines and pathways and the occurrence of a disruption of monocyte-T cell crosstalk driving GCA pathogenesis.


Assuntos
Linfócitos T CD4-Positivos , Perfilação da Expressão Gênica , Arterite de Células Gigantes , Monócitos , Transdução de Sinais , Transcriptoma , Humanos , Arterite de Células Gigantes/imunologia , Arterite de Células Gigantes/genética , Monócitos/imunologia , Monócitos/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Feminino , Masculino , Idoso , Metilação de DNA , Pessoa de Meia-Idade , Idoso de 80 Anos ou mais , Epigênese Genética , Comunicação Celular/imunologia , Regulação da Expressão Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...