Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 409
Filtrar
1.
Am J Reprod Immunol ; 92(3): e13921, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39225584

RESUMEN

PROBLEM: Endometrial immune cells are essential for maintaining homeostasis and the endometrial receptivity to embryo implantation. Understanding regional variations in endometrial immune cell populations is crucial for comprehending normal endometrial function and the pathophysiology of endometrial disorders. Despite previous studies focusing on the overall immune cell composition and function in the endometrium, regional variations in premenopausal women remain unclear. METHOD OF STUDY: Endometrial biopsies were obtained from four regions (anterior, posterior, left lateral, and right lateral) of premenopausal women undergoing hysteroscopy with no abnormalities. A 15-color human endometrial immune cell-focused flow cytometry panel was used for analysis. High-dimensional flow cytometry combined with a clustering algorithm was employed to unravel the complexity of endometrial immune cells. Additionally, multiplex immunofluorescent was performed for further validation. RESULTS: Our findings revealed no significant variation in the distribution and abundance of immune cells across different regions under normal conditions during the proliferative phase. Each region harbored similar immune cell subtypes, indicating a consistent immune microenvironment. However, when comparing normal regions to areas with focal hemorrhage, significant differences were observed. An increase in CD8+ T cells highlights the impact of localized abnormalities on the immune microenvironment. CONCLUSIONS: Our study demonstrates that the endometrial immune cell landscape is consistent across different anatomical regions during the proliferative phase in premenopausal women. This finding has important implications for understanding normal endometrial function and the pathophysiology of endometrial disorders.


Asunto(s)
Microambiente Celular , Endometrio , Humanos , Femenino , Endometrio/inmunología , Endometrio/patología , Adulto , Microambiente Celular/inmunología , Citometría de Flujo , Premenopausia/inmunología , Linfocitos T CD8-positivos/inmunología , Biopsia
2.
Front Immunol ; 15: 1432841, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39267742

RESUMEN

Traumatic spinal cord injury (tSCI) is a severe injury to the central nervous system that is categorized into primary and secondary injuries. Among them, the local microenvironmental imbalance in the spinal cord caused by secondary spinal cord injury includes accumulation of cytokines and chemokines, reduced angiogenesis, dysregulation of cellular energy metabolism, and dysfunction of immune cells at the site of injury, which severely impedes neurological recovery from spinal cord injury (SCI). In recent years, single-cell techniques have revealed the heterogeneity of multiple immune cells at the genomic, transcriptomic, proteomic, and metabolomic levels after tSCI, further deepening our understanding of the mechanisms underlying tSCI. However, spatial information about the tSCI microenvironment, such as cell location and cell-cell interactions, is lost in these approaches. The application of spatial multi-omics technology can solve this problem by combining the data obtained from immunohistochemistry and multiparametric analysis to reveal the changes in the microenvironment at different times of secondary injury after SCI. In this review, we systematically review the progress of spatial multi-omics techniques in the study of the microenvironment after SCI, including changes in the immune microenvironment and discuss potential future therapeutic strategies.


Asunto(s)
Microambiente Celular , Proteómica , Traumatismos de la Médula Espinal , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/metabolismo , Humanos , Microambiente Celular/inmunología , Proteómica/métodos , Animales , Metabolómica/métodos , Genómica/métodos , Transcriptoma , Análisis de la Célula Individual , Multiómica
3.
Nat Commun ; 15(1): 7809, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242588

RESUMEN

Innate lymphoid cells (ILCs) are critical in maintaining tissue homeostasis, and during infection and inflammation. Here we identify, by using combinatorial reporter mice, a rare ILC progenitor (ILCP) population, resident to the small intestinal lamina propria (siLP) in adult mice. Transfer of siLP-ILCP into recipients generates group 1 ILCs (including ILC1 and NK cells), ILC2s and ILC3s within the intestinal microenvironment, but almost exclusively group 1 ILCs in the liver, lung and spleen. Single cell gene expression analysis and high dimensional spectral cytometry analysis of the siLP-ILCPs and ILC progeny indicate that the phenotype of the group 1 ILC progeny is also influenced by the tissue microenvironment. Thus, a local pool of siLP-ILCP can contribute to pan-ILC generation in the intestinal microenvironment but has more restricted potential in other tissues, with a greater propensity than bone marrow-derived ILCPs to favour ILC1 and ILC3 production. Therefore, ILCP potential is influenced by both tissue of origin and the microenvironment during development. This may provide additional flexibility during the tuning of immune reactions.


Asunto(s)
Inmunidad Innata , Mucosa Intestinal , Células Progenitoras Linfoides , Ratones Endogámicos C57BL , Animales , Ratones , Mucosa Intestinal/inmunología , Mucosa Intestinal/citología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/inmunología , Microambiente Celular/inmunología , Linfocitos/inmunología , Intestino Delgado/inmunología , Intestino Delgado/citología , Femenino , Masculino
4.
Kidney360 ; 5(9): 1394-1401, 2024 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-39120952

RESUMEN

Lupus nephritis (LN) is the most common major organ manifestation of the autoimmune disease SLE (lupus), with 10% of those afflicted progressing to ESKD. The kidney in LN is characterized by a significant immune infiltrate and proinflammatory cytokine milieu that affects intrinsic renal cells and is, in part, responsible for the tissue damage observed in LN. It is now increasingly appreciated that LN is not due to unidirectional immune cell activation with subsequent kidney damage. Rather, the kidney microenvironment influences the recruitment, survival, differentiation, and activation of immune cells, which, in turn, modify kidney cell function. This review covers how the biochemical environment of the kidney ( i.e ., low oxygen tension and hypertonicity) and unique kidney cell types affect the intrarenal immune cells in LN. The pathways used by intrinsic renal cells to interact with immune cells, such as antigen presentation and cytokine production, are discussed in detail. An understanding of these mechanisms can lead to the design of more kidney-targeted treatments and the avoidance of systemic immunosuppressive effects and may represent the next frontier of LN therapies.


Asunto(s)
Riñón , Nefritis Lúpica , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Humanos , Riñón/inmunología , Riñón/patología , Citocinas/metabolismo , Citocinas/inmunología , Microambiente Celular/inmunología , Animales
5.
Curr Opin Immunol ; 89: 102453, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39173413

RESUMEN

The delicate balance between protective immunity against pathogens and the prevention of autoimmunity requires finely tuned generation and function of regulatory CD4+ T (Treg) cells. Here, we review recent progress in the understanding of a complex set of cues, which converge on Treg cells in lymphoid and nonlymphoid organs and in tumors and how these cues modulate Treg functions. We highlight the versatility of Treg cells underlying their ability to dynamically adapt to local microenvironments and perform a wide range of functions that extend beyond the archetypal role of Treg cells in moderating adverse effects of immune response-associated inflammation and in suppressing autoimmunity.


Asunto(s)
Linfocitos T Reguladores , Humanos , Linfocitos T Reguladores/inmunología , Animales , Microambiente Celular/inmunología , Autoinmunidad , Microambiente Tumoral/inmunología , Neoplasias/inmunología , Inflamación/inmunología
6.
Int J Mol Sci ; 25(16)2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39201715

RESUMEN

Androgenetic alopecia (AGA) is characterized by microinflammation and abnormal immune responses, particularly in the upper segment of hair follicles (HFs). However, the precise patterns of immune dysregulation remain unclear, partly due to limitations in current analysis techniques to preserve tissue architecture. The infundibulum, a major part of the upper segment of HFs, is associated with significant clusters of immune cells. In this study, we investigated immune cells around the infundibulum, referred to as peri-infundibular immune infiltration (PII). We employed spatial transcriptome profiling, a high-throughput analysis technology, to investigate the immunological disruptions within the PII region. Our comprehensive analysis included an evaluation of overall immune infiltrates, gene set enrichment analysis (GSEA), cellular deconvolution, differential expression analysis, over-representation analysis, protein-protein interaction (PPI) networks, and upstream regulator analysis to identify cell types and molecular dysregulation in immune cells. Our results demonstrated significant differences in immune signatures between the PII of AGA patients (PII-A) and the PII of control donors (PII-C). Specifically, PII-A exhibited an enrichment of CD4+ helper T cells, distinct immune response patterns, and a bias toward a T helper (Th) 2 response. Immunohistochemistry revealed disruptions in T cell subpopulations, with more CD4+ T cells displaying an elevated Th2 response and a reduced Th1-cytotoxic response compared to PII-C. These findings reveal the unique immune landscapes of PII-A and PII-C, suggesting potential for the development of innovative treatment approaches.


Asunto(s)
Alopecia , Perfilación de la Expresión Génica , Folículo Piloso , Transcriptoma , Humanos , Alopecia/genética , Alopecia/inmunología , Alopecia/metabolismo , Alopecia/patología , Folículo Piloso/inmunología , Folículo Piloso/metabolismo , Folículo Piloso/patología , Masculino , Adulto , Mapas de Interacción de Proteínas , Persona de Mediana Edad , Femenino , Microambiente Celular/inmunología , Microambiente Celular/genética , Células Th2/inmunología , Células Th2/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo
7.
Front Immunol ; 15: 1414387, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39026669

RESUMEN

Background: Sarcopenia is a condition characterized by the age-related loss of skeletal muscle mass and function. The pathogenesis of the disease is influenced by chronic low-grade inflammation. However, the specific changes in the immune landscape changes of sarcopenic muscle are not yet fully understood. Methods: To gain insights into the immune cell composition and interactions, we combined single-nucleus RNA sequencing data, bulk RNA sequencing dataset, and comprehensive bioinformatic analyses on the skeletal muscle samples from young, aged, and sarcopenic individuals. Histological staining was then performed on skeletal muscles to validate the distribution of immune cells in clinical samples. Results: We analyzed the transcriptomes of 101,862 single nuclei, revealing a total of 10 major cell types and 6 subclusters of immune cell types within the human skeletal muscle tissues. Notable variations were identified in the immune microenvironment between young and aged skeletal muscle. Among the immune cells from skeletal muscle microenvironment, macrophages constituted the largest fraction. A specific marker gene LYVE1 for skeletal muscle resident macrophages was further identified. Cellular subclasses included four distinct groups of resident macrophages, which play different roles in physiological or non-physiological conditions. Utilizing bulk RNA sequencing data, we observed a significant enrichment of macrophage-rich inflammation in sarcopenia. Conclusions: Our findings demonstrate age-related changes in the composition and cross-talk of immune cells in human skeletal muscle microenvironment, which contribute to chronic inflammation in aged or sarcopenia muscle. Furthermore, macrophages emerge as a potential therapeutic target, thus advancing our understanding of the pathogenesis of sarcopenia.


Asunto(s)
Perfilación de la Expresión Génica , Músculo Esquelético , Sarcopenia , Transcriptoma , Sarcopenia/inmunología , Sarcopenia/genética , Sarcopenia/patología , Humanos , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Músculo Esquelético/metabolismo , Anciano , Masculino , Adulto , Macrófagos/inmunología , Macrófagos/metabolismo , Femenino , Persona de Mediana Edad , Microambiente Celular/inmunología , Microambiente Celular/genética , Envejecimiento/inmunología , Envejecimiento/genética
8.
Front Immunol ; 15: 1397475, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38979407

RESUMEN

Monocytes are pivotal immune cells in eliciting specific immune responses and can exert a significant impact on the progression, prognosis, and immunotherapy of intracranial aneurysms (IAs). The objective of this study was to identify monocyte/macrophage (Mo/MΦ)-associated gene signatures to elucidate their correlation with the pathogenesis and immune microenvironment of IAs, thereby offering potential avenues for targeted therapy against IAs. Single-cell RNA-sequencing (scRNA-seq) data of IAs were acquired from the Gene Expression Synthesis (GEO) database. The significant infiltration of monocyte subsets in the parietal tissue of IAs was identified using single-cell RNA sequencing and high-dimensional weighted gene co-expression network analysis (hdWGCNA). The integration of six machine learning algorithms identified four crucial genes linked to these Mo/MΦ. Subsequently, we developed a multilayer perceptron (MLP) neural model for the diagnosis of IAs (independent external test AUC=1.0, sensitivity =100%, specificity =100%). Furthermore, we employed the CIBERSORT method and MCP counter to establish the correlation between monocyte characteristics and immune cell infiltration as well as patient heterogeneity. Our findings offer valuable insights into the molecular characterization of monocyte infiltration in IAs, which plays a pivotal role in shaping the immune microenvironment of IAs. Recognizing this characterization is crucial for comprehending the limitations associated with targeted therapies for IAs. Ultimately, the results were verified by real-time fluorescence quantitative PCR and Immunohistochemistry.


Asunto(s)
Aneurisma Intracraneal , Aprendizaje Automático , Macrófagos , Monocitos , Análisis de la Célula Individual , Humanos , Aneurisma Intracraneal/genética , Aneurisma Intracraneal/inmunología , Análisis de la Célula Individual/métodos , Monocitos/inmunología , Monocitos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Perfilación de la Expresión Génica , Transcriptoma , Microambiente Celular/inmunología , Microambiente Celular/genética , Masculino , Femenino , Redes Reguladoras de Genes , Biología Computacional/métodos
10.
Front Immunol ; 15: 1336839, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38947313

RESUMEN

Background: In spite of its high mortality rate and poor prognosis, the pathogenesis of sepsis is still incompletely understood. This study established a cuproptosis-based risk model to diagnose and predict the risk of sepsis. In addition, the cuproptosis-related genes were identified for targeted therapy. Methods: Single-cell sequencing analyses were used to characterize the cuproptosis activity score (CuAS) and intercellular communications in sepsis. Differential cuproptosis-related genes (CRGs) were identified in conjunction with single-cell and bulk RNA sequencing. LASSO and Cox regression analyses were employed to develop a risk model. Three external cohorts were conducted to assess the model's accuracy. Differences in immune infiltration, immune cell subtypes, pathway enrichment, and the expression of immunomodulators were further evaluated in distinct groups. Finally, various in-vitro experiments, such as flow cytometry, Western blot, and ELISA, were used to explore the role of LST1 in sepsis. Results: ScRNA-seq analysis demonstrated that CuAS was highly enriched in monocytes and was closely related to the poor prognosis of sepsis patients. Patients with higher CuAS exhibited prominent strength and numbers of cell-cell interactions. A total of five CRGs were identified based on the LASSO and Cox regression analyses, and a CRG-based risk model was established. The lower riskScore cohort exhibited enhanced immune cell infiltration, elevated immune scores, and increased expression of immune modulators, indicating the activation of an antibacterial response. Ultimately, in-vitro experiments demonstrated that LST1, a key gene in the risk model, was enhanced in the macrophage in response to LPS, which was closely related to the decrease of macrophage survival rate, the enhancement of apoptosis and oxidative stress injury, and the imbalance of the M1/M2 phenotype. Conclusions: This study constructed a cuproptosis-related risk model to accurately predict the prognosis of sepsis. We further characterized the cuproptosis-related gene LST1 to provide a theoretical framework for sepsis therapy.


Asunto(s)
Sepsis , Análisis de la Célula Individual , Sepsis/inmunología , Sepsis/genética , Humanos , Masculino , Femenino , Persona de Mediana Edad , Pronóstico , Análisis de Secuencia de ARN , Microambiente Celular/inmunología , Anciano
11.
J Cereb Blood Flow Metab ; 44(10): 1733-1744, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38833561

RESUMEN

Carotid atherosclerosis is a major cause of stroke. Hemodynamic forces, such as shear stress and oscillatory shear, play an important role in the initiation and progression of atherosclerosis. The alteration of the immune microenvironment is the fundamental pathological mechanism by which diverse external environmental factors impact the formation and progression of plaques. However, Current research on the relationship between hemodynamics and immunity in atherosclerosis still lack of comprehensive understanding. In this study, we combined computational fluid dynamics (CFD) and Mass cytometry (CyTOF) technologies to explore the changes in the immune microenvironment within plaques under different hemodynamic conditions. Our results indicated that neutrophils were enriched in adverse flow environments. M2-like CD163+CD86+ macrophages were predominantly enriched in high WSS and low OSI environments, while CD163-CD14+ macrophages were enriched in low WSS and high OSI environments. Functional analysis further revealed T cell pro-inflammatory activation and dysregulation in modulation, along with an imbalance in M1-like/M2-like macrophages, suggesting their potential involvement in the progression of atherosclerotic lesions mediated by adverse flow patterns. Our study elucidated the potential mechanisms by which hemodynamics regulated the immune microenvironment within plaques, providing intervention targets for future precision therapies.


Asunto(s)
Enfermedades de las Arterias Carótidas , Hemodinámica , Macrófagos , Enfermedades de las Arterias Carótidas/inmunología , Enfermedades de las Arterias Carótidas/fisiopatología , Humanos , Macrófagos/inmunología , Masculino , Hidrodinámica , Placa Aterosclerótica/inmunología , Animales , Neutrófilos/inmunología , Microambiente Celular/inmunología
12.
Cytokine ; 181: 156685, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38945040

RESUMEN

The close link between immune and pathogenesis of venous thromboembolism (VTE) has been recognized, but not fully elucidated. The current study was designed to identify immune microenvironment related signature and subtypes using explainable machine learning in VTE. We first observed an alteration of immune microenvironment in VTE patients and identified eight key immune cells involved in VTE. Then PTPN6, ITGB2, CR2, FPR2, MMP9 and ISG15 were determined as key immune microenvironment-related genes, which could divide VTE patients into two subtypes with different immune and metabolic characteristics. Also, we found that prunetin and torin-2 may be most promising to treat VTE patients in Cluster 1 and 2, respectively. By comparing six machine learning models in both training and external validation sets, XGboost was identified as the best one to predict the risk of VTE, followed by the interpretation of each immune microenvironment-related gene contributing to the model. Moreover, CR2 and FPR2 had high accuracy in distinguishing VTE and control, which may act as diagnostic biomarkers of VTE, and their expressions were validated by qPCR. Collectively, immune microenvironment related PTPN6, ITGB2, CR2, FPR2, MMP9 and ISG15 are key genes involved in the pathogenesis of VTE. The VTE risk prediction model and immune microenvironment subtypes based on those genes might benefit prevention, diagnosis, and the individualized treatment strategy in clinical practice of VTE.


Asunto(s)
Tromboembolia Venosa , Humanos , Tromboembolia Venosa/inmunología , Masculino , Femenino , Persona de Mediana Edad , Biomarcadores/metabolismo , Aprendizaje Automático , Microambiente Celular/inmunología
13.
Int J Mol Sci ; 25(11)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38891952

RESUMEN

The pancreas is an organ with both exocrine and endocrine functions, comprising a highly organized and complex tissue microenvironment composed of diverse cellular and non-cellular components. The impairment of microenvironmental homeostasis, mediated by the dysregulation of cell-to-cell crosstalk, can lead to pancreatic diseases such as pancreatitis, diabetes, and pancreatic cancer. Macrophages, key immune effector cells, can dynamically modulate their polarization status between pro-inflammatory (M1) and anti-inflammatory (M2) modes, critically influencing the homeostasis of the pancreatic microenvironment and thus playing a pivotal role in the pathogenesis of the pancreatic disease. This review aims to summarize current findings and provide detailed mechanistic insights into how alterations mediated by macrophage polarization contribute to the pathogenesis of pancreatic disorders. By analyzing current research comprehensively, this article endeavors to deepen our mechanistic understanding of regulatory molecules that affect macrophage polarity and the intricate crosstalk that regulates pancreatic function within the microenvironment, thereby facilitating the development of innovative therapeutic strategies that target perturbations in the pancreatic microenvironment.


Asunto(s)
Macrófagos , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , Enfermedades Pancreáticas/patología , Enfermedades Pancreáticas/inmunología , Enfermedades Pancreáticas/metabolismo , Microambiente Celular/inmunología , Páncreas/inmunología , Páncreas/patología , Páncreas/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Activación de Macrófagos/inmunología
14.
Front Immunol ; 15: 1360618, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38827737

RESUMEN

Psoriasis is a chronic inflammatory disease affecting skin and joints characterized by a chronically altered immune and inflammatory response. Several factors occur from the onset to the development of this disease due to different types of cells spatially and temporally localized in the affected area, such as, keratinocytes, macrophages, neutrophils and T helper lymphocytes. This scenario leads to the chronic release of high levels of inflammatory mediators (i.e., IL-17, IL-23, IL-22, TNF-α, S100 proteins, Defensins) and lastly parakeratosis and thickening of the stratum spinosum. Extracellular vesicles (EVs) are small double membraned biological nanoparticles that are secreted by all cell types and classified, based on dimension and biogenesis, into exosomes, microvesicles and apoptotic bodies. Their role as vessels for long range molecular signals renders them key elements in the pathogenesis of psoriasis, as well as innovative platforms for potential biomarker discovery and delivery of fine-tuned anti-inflammatory therapies. In this review, the role of EVs in the pathogenesis of psoriasis and the modulation of cellular microenvironment has been summarized. The biotechnological implementation of EVs for therapy and research for new biomarkers has been also discussed.


Asunto(s)
Biomarcadores , Vesículas Extracelulares , Psoriasis , Humanos , Psoriasis/inmunología , Psoriasis/metabolismo , Psoriasis/etiología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/inmunología , Animales , Piel/patología , Piel/inmunología , Piel/metabolismo , Microambiente Celular/inmunología
15.
Front Immunol ; 15: 1371463, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38895114

RESUMEN

Osteoporosis represents a systemic imbalance in bone metabolism, augmenting the susceptibility to fractures among patients and emerging as a notable mortality determinant in the elderly population. It has evolved into a worldwide concern impacting the physical well-being of the elderly, imposing a substantial burden on both human society and the economy. Presently, the precise pathogenesis of osteoporosis remains inadequately characterized and necessitates further exploration. The advancement of osteoporosis is typically linked to the initiation of an inflammatory response. Cells in an inflammatory environment can cause inflammatory death including pyroptosis. Pyroptosis is a recently identified form of programmed cell death with inflammatory properties, mediated by the caspase and gasdermin families. It is regarded as the most inflammatory form of cell death in contemporary medical research. Under the influence of diverse cytokines, macrophages, and other immune cells may undergo pyroptosis, releasing inflammatory factors, such as IL-1ß and IL-18. Numerous lines of evidence highlight the pivotal role of pyroptosis in the pathogenesis of inflammatory diseases, including cancer, intestinal disorders, hepatic conditions, and cutaneous ailments. Osteoporosis progression is frequently associated with inflammation; hence, pyroptosis may also play a role in the pathogenesis of osteoporosis to a certain extent, making it a potential target for treatment. This paper has provided a comprehensive summary of pertinent research concerning pyroptosis and its impact on osteoporosis. The notion proposing that pyroptosis mediates osteoporosis via the inflammatory immune microenvironment is advanced, and we subsequently investigate potential targets for treating osteoporosis through the modulation of pyroptosis.


Asunto(s)
Inflamación , Osteoporosis , Piroptosis , Humanos , Piroptosis/inmunología , Osteoporosis/inmunología , Osteoporosis/metabolismo , Osteoporosis/etiología , Animales , Inflamación/inmunología , Microambiente Celular/inmunología
17.
J Nanobiotechnology ; 22(1): 315, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38840207

RESUMEN

Chronic hepatitis B poses a significant global burden, modulating immune cells, leading to chronic inflammation and long-term damage. Due to its hepatotropism, the hepatitis B virus (HBV) cannot infect other cells. The mechanisms underlying the intercellular communication among different liver cells in HBV-infected individuals and the immune microenvironment imbalance remain elusive. Exosomes, as important intercellular communication and cargo transportation tools between HBV-infected hepatocytes and immune cells, have been shown to assist in HBV cargo transportation and regulate the immune microenvironment. However, the role of exosomes in hepatitis B has only gradually received attention in recent years. Minimal literature has systematically elaborated on the role of exosomes in reshaping the immune microenvironment of the liver. This review unfolds sequentially based on the biological processes of exosomes: exosomes' biogenesis, release, transport, uptake by recipient cells, and their impact on recipient cells. We delineate how HBV influences the biogenesis of exosomes, utilizing exosomal covert transmission, and reshapes the hepatic immune microenvironment. And based on the characteristics and functions of exosomes, potential applications of exosomes in hepatitis B are summarized and predicted.


Asunto(s)
Exosomas , Virus de la Hepatitis B , Hepatitis B Crónica , Hepatocitos , Hígado , Exosomas/inmunología , Exosomas/metabolismo , Humanos , Virus de la Hepatitis B/inmunología , Hígado/inmunología , Hígado/virología , Animales , Hepatitis B Crónica/inmunología , Hepatocitos/virología , Hepatocitos/inmunología , Comunicación Celular , Microambiente Celular/inmunología , Hepatitis B/inmunología , Hepatitis B/virología
18.
ACS Biomater Sci Eng ; 10(6): 3923-3934, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38766805

RESUMEN

The repair of critical-sized bone defects continues to pose a challenge in clinics. Strontium (Sr), recognized for its function in bone metabolism regulation, has shown potential in bone repair. However, the underlying mechanism through which Sr2+ guided favorable osteogenesis by modulating macrophages remains unclear, limiting their application in the design of bone biomaterials. Herein, Sr-incorporated bioactive glass (SrBG) was synthesized for further investigation. The release of Sr ions enhanced the immunomodulatory properties and osteogenic potential by modulating the polarization of macrophages toward the M2 phenotype. In vivo, a 3D-printed SrBG scaffold was fabricated and showed consistently improved bone regeneration by creating a prohealing immunological microenvironment. RNA sequencing was performed to explore the underlying mechanisms. It was found that Sr ions might enhance the mitochondrial function of macrophage by activating PI3K/AKT/mTOR signaling, thereby favoring osteogenesis. Our findings demonstrate the relationship between the immunomodulatory role of Sr ions and the mitochondrial function of macrophages. By focusing on the mitochondrial function of macrophages, Sr2+-mediated immunomodulation sheds light on the future design of biomaterials for tissue regenerative engineering.


Asunto(s)
Vidrio , Macrófagos , Mitocondrias , Transducción de Señal , Estroncio , Animales , Ratones , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/química , Regeneración Ósea/efectos de los fármacos , Microambiente Celular/efectos de los fármacos , Microambiente Celular/inmunología , Vidrio/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/inmunología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Osteogénesis/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Estroncio/farmacología , Estroncio/química , Andamios del Tejido/química , Serina-Treonina Quinasas TOR/metabolismo
19.
Front Immunol ; 15: 1365422, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38807593

RESUMEN

Autogenous arteriovenous fistula (AVF) is the preferred dialysis access for receiving hemodialysis treatment in end-stage renal disease patients. After AVF is established, vascular remodeling occurs in order to adapt to hemodynamic changes. Uremia toxins, surgical injury, blood flow changes and other factors can induce inflammatory response, immune microenvironment changes, and play an important role in the maintenance of AVF vascular remodeling. This process involves the infiltration of pro-inflammatory and anti-inflammatory immune cells and the secretion of cytokines. Pro-inflammatory and anti-inflammatory immune cells include neutrophil (NEUT), dendritic cell (DC), T lymphocyte, macrophage (Mφ), etc. This article reviews the latest research progress and focuses on the role of immune microenvironment changes in vascular remodeling of AVF, in order to provide a new theoretical basis for the prevention and treatment of AVF failure.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Microambiente Celular , Fallo Renal Crónico , Diálisis Renal , Remodelación Vascular , Animales , Humanos , Derivación Arteriovenosa Quirúrgica/efectos adversos , Microambiente Celular/inmunología , Fallo Renal Crónico/terapia , Fallo Renal Crónico/inmunología
20.
Aging Cell ; 23(6): e14140, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38481058

RESUMEN

Weakened germinal center responses by the aged immune system result in diminished immunity against pathogens and reduced efficacy of vaccines. Prolonged contacts between activated B cells and CD4+ T cells are crucial to germinal center formation and T follicular helper cell (Tfh) differentiation, but it is unclear how aging impacts the quality of this interaction. Peptide immunization confirmed that aged mice have decreased expansion of antigen-specific germinal center B cells and reduced antibody titers. Furthermore, aging was associated with accumulated Tfh cells, even in naïve mice. Despite increased numbers, aged Tfh had reduced expression of master transcription factor BCL6 and increased expression of the ectonucleotidase CD39. In vitro activation revealed that proliferative capacity was maintained in aged CD4+ T cells, but not the costimulatory molecule CD40L. When activated in vitro by aged antigen-presenting cells, young CD4+ naïve T cells generated reduced numbers of activated cells with upregulated CD40L. To determine the contribution of cell-extrinsic influences on antigen-specific Tfh induction, young, antigen-specific B and CD4+ T cells were adoptively transferred into aged hosts prior to peptide immunization. Transferred cells had reduced expansion and differentiation into germinal center B cell and Tfh and reduced antigen-specific antibody titers when compared to young hosts. Young CD4+ T cells transferred aged hosts differentiated into Tfh cells with reduced PD-1 and BCL6 expression, and increased CD39 expression, though they maintained their mitochondrial capacity. These results highlight the role of the lymphoid microenvironment in modulating CD4+ T cell differentiation, which contributes to impaired establishment and maintenance of germinal centers.


Asunto(s)
Ligando de CD40 , Diferenciación Celular , Proteínas Proto-Oncogénicas c-bcl-6 , Animales , Ratones , Envejecimiento/inmunología , Ligando de CD40/metabolismo , Ligando de CD40/inmunología , Diferenciación Celular/inmunología , Microambiente Celular/inmunología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Células T Auxiliares Foliculares/inmunología , Células T Auxiliares Foliculares/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Masculino , Femenino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...