Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.912
Filter
1.
Front Immunol ; 15: 1416961, 2024.
Article in English | MEDLINE | ID: mdl-38983862

ABSTRACT

Depression, projected to be the predominant contributor to the global disease burden, is a complex condition with diverse symptoms including mood disturbances and cognitive impairments. Traditional treatments such as medication and psychotherapy often fall short, prompting the pursuit of alternative interventions. Recent research has highlighted the significant role of gut microbiota in mental health, influencing emotional and neural regulation. Fecal microbiota transplantation (FMT), the infusion of fecal matter from a healthy donor into the gut of a patient, emerges as a promising strategy to ameliorate depressive symptoms by restoring gut microbial balance. The microbial-gut-brain (MGB) axis represents a critical pathway through which to potentially rectify dysbiosis and modulate neuropsychiatric outcomes. Preclinical studies reveal that FMT can enhance neurochemicals and reduce inflammatory markers, thereby alleviating depressive behaviors. Moreover, FMT has shown promise in clinical settings, improving gastrointestinal symptoms and overall quality of life in patients with depression. The review highlights the role of the gut-brain axis in depression and the need for further research to validate the long-term safety and efficacy of FMT, identify specific therapeutic microbial strains, and develop targeted microbial modulation strategies. Advancing our understanding of FMT could revolutionize depression treatment, shifting the paradigm toward microbiome-targeting therapies.


Subject(s)
Brain-Gut Axis , Depression , Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Humans , Depression/therapy , Depression/microbiology , Dysbiosis/therapy , Animals , Treatment Outcome
2.
Front Immunol ; 15: 1427739, 2024.
Article in English | MEDLINE | ID: mdl-39026681

ABSTRACT

Lactylation is a process where lactate, a cellular metabolism byproduct, is added to proteins, altering their functions. In the realm of macrophage activation, lactylation impacts inflammatory response and immune regulation. Understanding the effects of lactylation on macrophage activation is vital in lung diseases, as abnormal activation and function are pivotal in conditions like pneumonia, pulmonary fibrosis, COPD, and lung cancer. This review explores the concept of lactylation, its regulation of macrophage activation, and recent research progress in lung diseases. It offers new insights into lung disease pathogenesis and potential therapeutic targets.


Subject(s)
Lung Diseases , Macrophage Activation , Humans , Macrophage Activation/immunology , Animals , Lung Diseases/immunology , Lung Diseases/metabolism , Lactic Acid/metabolism , Macrophages/immunology , Macrophages/metabolism
3.
J Nanobiotechnology ; 22(1): 398, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970103

ABSTRACT

Diabetic wounds are characterized by incomplete healing and delayed healing, resulting in a considerable global health care burden. Exosomes are lipid bilayer structures secreted by nearly all cells and express characteristic conserved proteins and parent cell-associated proteins. Exosomes harbor a diverse range of biologically active macromolecules and small molecules that can act as messengers between different cells, triggering functional changes in recipient cells and thus endowing the ability to cure various diseases, including diabetic wounds. Exosomes accelerate diabetic wound healing by regulating cellular function, inhibiting oxidative stress damage, suppressing the inflammatory response, promoting vascular regeneration, accelerating epithelial regeneration, facilitating collagen remodeling, and reducing scarring. Exosomes from different tissues or cells potentially possess functions of varying levels and can promote wound healing. For example, mesenchymal stem cell-derived exosomes (MSC-exos) have favorable potential in the field of healing due to their superior stability, permeability, biocompatibility, and immunomodulatory properties. Exosomes, which are derived from skin cellular components, can modulate inflammation and promote the regeneration of key skin cells, which in turn promotes skin healing. Therefore, this review mainly emphasizes the roles and mechanisms of exosomes from different sources, represented by MSCs and skin sources, in improving diabetic wound healing. A deeper understanding of therapeutic exosomes will yield promising candidates and perspectives for diabetic wound healing management.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Wound Healing , Exosomes/metabolism , Humans , Animals , Mesenchymal Stem Cells/metabolism , Diabetes Mellitus/metabolism , Skin/metabolism , Oxidative Stress , Diabetes Complications
4.
Front Immunol ; 15: 1385319, 2024.
Article in English | MEDLINE | ID: mdl-38962004

ABSTRACT

The immune system comprises a complex yet tightly regulated network of cells and molecules that play a critical role in protecting the body from infection and disease. The activity and development of each immune cell is regulated in a myriad of ways including through the cytokine milieu, the availability of key receptors, via tailored intracellular signalling cascades, dedicated transcription factors and even by directly modulating gene accessibility and expression; the latter is more commonly known as epigenetic regulation. In recent years, epigenetic regulators have begun to emerge as key players involved in modulating the immune system. Among these, the lysine methyltransferase DOT1L has gained significant attention for its involvement in orchestrating immune cell formation and function. In this review we provide an overview of the role of DOT1L across the immune system and the implications of this role on health and disease. We begin by elucidating the general mechanisms of DOT1L-mediated histone methylation and its impact on gene expression within immune cells. Subsequently, we provide a detailed and comprehensive overview of recent studies that identify DOT1L as a crucial regulator of immune cell development, differentiation, and activation. Next, we discuss the potential mechanisms of DOT1L-mediated regulation of immune cell function and shed light on how DOT1L might be contributing to immune cell homeostasis and dysfunction. We then provide food for thought by highlighting some of the current obstacles and technical limitations precluding a more in-depth elucidation of DOT1L's role. Finally, we explore the potential therapeutic implications of targeting DOT1L in the context of immune-related diseases and discuss ongoing research efforts to this end. Overall, this review consolidates the current paradigm regarding DOT1L's role across the immune network and emphasises its critical role in governing the healthy immune system and its potential as a novel therapeutic target for immune-related diseases. A deeper understanding of DOT1L's immunomodulatory functions could pave the way for innovative therapeutic approaches which fine-tune the immune response to enhance or restore human health.


Subject(s)
Epigenesis, Genetic , Histone-Lysine N-Methyltransferase , Immune System , Humans , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Animals , Immune System/immunology , Immune System/metabolism , Immunomodulation , Histones/metabolism , Histones/immunology
5.
BMC Oral Health ; 24(1): 824, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033148

ABSTRACT

BACKGROUND: Excessive inflammation is a major cause of implant failure. The surface morphology, hydrophilicity, and loading of biomaterials are major properties modulating anti-inflammatory macrophage activation. This paper investigates the regulatory effects of modifying the surface of Titanium dioxide nanotubes (TNTs) with graphene oxide (GO) on the polarization of mouse monocyte macrophages (RAW264.7). METHODS: TNT was produced by the anodic oxidation of titanium. GO was subsequently electrodeposited on the TNT to obtain a TNT-GO composite. The samples were characterised through scanning electron microscopy (SEM), Raman spectroscopy, and X-ray diffraction. RAW264.7 cells were separately seeded onto the surface of three groups of samples: pure Ti, TNT, and TNT-GO. Under the condition of lipopolysaccharide stimulation, the influence of the sample surfaces on the gene expression profiles was investigated through RNA sequence analysis. In addition, cell spreading was observed through SEM, cell adhesion and proliferation were analysed using the CCK8 assay, and the expression of inflammation-related factors was investigated by ELISA and cellular immunofluorescence staining. The production of reactive oxygen species (ROS) in the RAW264.7 cells on the surface of the three groups was detected via immunofluorescence staining. RESULTS: The CCK8 results indicated that the adhesion and proliferation of the RAW264.7 cells were reduced on the TNT and TNT-GO surfaces. ELISA results revealed significant differences in the pro-inflammatory factors tumour necrosis factor-α and interleukin-6 secretion among the three groups at 24 h (p < 0.05). The secretion of pro-inflammatory factors significantly reduced and the expression of anti-inflammatory factor IL-10 increased on the TNT and TNT-GO surfaces. The RNA sequencing, ELISA, and cell immunofluorescence staining test results suggested that the inflammatory response of M1 polarization was reduced and the M2 polarization of macrophages was induced on the TNT-GO surface, which may be attributed to the reduction in ROS production. CONCLUSIONS: Under lipopolysaccharide stimulation, the inflammatory response of the RAW264.7 cells was reduced and the M2 polarization of macrophages was promoted on the TNT-GO surface, which may be caused by the reduced ROS production. Consequently, the designed TNT-GO material is promising for implants owing to its excellent inflammation regulation ability.


Subject(s)
Graphite , Macrophages , Nanotubes , Reactive Oxygen Species , Titanium , Graphite/pharmacology , Animals , Mice , Macrophages/drug effects , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Inflammation , Cell Adhesion/drug effects , Surface Properties , Lipopolysaccharides , Microscopy, Electron, Scanning , Cell Proliferation/drug effects , Spectrum Analysis, Raman , X-Ray Diffraction , Macrophage Activation/drug effects
6.
Mil Med Res ; 11(1): 48, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39034405

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the substantia nigra (SN). Activation of the neuroinflammatory response has a pivotal role in PD. Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach for various nerve injuries, but there are limited reports on their use in PD and the underlying mechanisms remain unclear. METHODS: We investigated the effects of clinical-grade hypoxia-preconditioned olfactory mucosa (hOM)-MSCs on neural functional recovery in both PD models and patients, as well as the preventive effects on mouse models of PD. To assess improvement in neuroinflammatory response and neural functional recovery induced by hOM-MSCs exposure, we employed single-cell RNA sequencing (scRNA-seq), assay for transposase accessible chromatin with high-throughput sequencing (ATAC-seq) combined with full-length transcriptome isoform-sequencing (ISO-seq), and functional assay. Furthermore, we present the findings from an initial cohort of patients enrolled in a phase I first-in-human clinical trial evaluating the safety and efficacy of intraspinal transplantation of hOM-MSC transplantation into severe PD patients. RESULTS: A functional assay identified that transforming growth factor-ß1 (TGF-ß1), secreted from hOM-MSCs, played a critical role in modulating mitochondrial function recovery in dopaminergic neurons. This effect was achieved through improving microglia immune regulation and autophagy homeostasis in the SN, which are closely associated with neuroinflammatory responses. Mechanistically, exposure to hOM-MSCs led to an improvement in neuroinflammation and neural function recovery partially mediated by TGF-ß1 via activation of the anaplastic lymphoma kinase/phosphatidylinositol-3-kinase/protein kinase B (ALK/PI3K/Akt) signaling pathway in microglia located in the SN of PD patients. Furthermore, intraspinal transplantation of hOM-MSCs improved the recovery of neurologic function and regulated the neuroinflammatory response without any adverse reactions observed in patients with PD. CONCLUSIONS: These findings provide compelling evidence for the involvement of TGF-ß1 in mediating the beneficial effects of hOM-MSCs on neural functional recovery in PD. Treatment and prevention of hOM-MSCs could be a promising and effective neuroprotective strategy for PD. Additionally, TGF-ß1 may be used alone or combined with hOM-MSCs therapy for treating PD.


Subject(s)
Disease Models, Animal , Mesenchymal Stem Cells , Olfactory Mucosa , Parkinson Disease , Transforming Growth Factor beta1 , Animals , Female , Humans , Male , Mice , Middle Aged , Mesenchymal Stem Cell Transplantation/methods , Parkinson Disease/complications , Parkinson Disease/therapy , Recovery of Function , Transforming Growth Factor beta1/metabolism
7.
Curr Rheumatol Rev ; 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39034724

ABSTRACT

BACKGROUND: Juvenile-onset systemic lupus erythematosus (jSLE) is an uncommon yet severe autoimmune/inflammatory condition affecting multiple bodily systems, typically manifest-ing before the age of 18. This disease exhibits significant complexity, displaying considerable variation among patients. Its effects can range in severity from minor to fatal, characterized by a pattern of recurring flare-ups and periods of remission, making its natural progression difficult to predict. AIM OF THE WORK: The aim of this work is to investigate the correlation between semaphorin 3A and systemic lupus erythematosus patients who follow up at Pediatric Rheumatology Unit Chil-dren's Hospital at Cairo University. PATIENTS & METHODS: This cross-sectional research was performed at the Pediatric Rheumatology Unit Cairo University Children's Hospital and included cases with jSLE under treatment and fol-low-up from the period of August 2021 to August 2022. RESULTS: Regarding demographic data of the studied subjects, highly significant variances were noted among the patient group & control group regarding age (years) & sex. However, there were non-significant variances among the patient group and control group concerning weight. In the current research, median (IQR) onset of disease was 2 (1 3) years, mean ± SD age at dis-ease diagnosis was 8.98 ± 2.13 years, median (IQR) disease duration 2 (1 3) years, family history was negative in 36 (90.0%) patients and consanguinity was negative in 28 (70.0%). The distribution of the manifestations within the patients group was as follow 7 (17.5%) with mu-cocutaneous, 7 (17.5%) with vasculitis, 4 (10.0%) with serositis, 11 (27.5%) with cardiac, 17 (42.5%) with renal, 11 (27.5%) with GIT, 5 (12.5%) with hematological, and 4 (10.0%) with neu-rological manifestations. In addition, there were 2 (5.0%) with arthritis, 31 (77.5%) with arthral-gia, and 2 (5.0%) with fever mean ± SD systolic BP was 115.95 ± 8.38 & mean ± SD diastolic BP was 75.60 ± 6.11. Regarding treatments in the patients' group, the median steroid dose was 15mg (5-25) with medi-an duration of 2 (1 3), 38 (95.0%) patients received hydroxychloroquine with mean ± SD hy-droxychloroquine dose of 205.26 mg ± 51.71. 23 (57.5%) patients received cyclophosphamide with mean ± SD number of cyclophosphamide doses 7.17 mg ± 2.42. Mycophenolate was re-ceived in 27 (67.5%) with mean ± SD dose of 614.07 mg ± 225.85. There were highly statistically significant differences between control group and patients' group concerning TLC, creatinine, & ESR. Highly statistically significant variance was noted among the control group and patients group concerning CRP. Regarding the patients' group, the mean ± SD serum C3 was 99.89 mg/dl ± 28.45, median (IQR) serum C4 was 14.5 mg/dl (8.8 25.5), and median (IQR) albumin creatinine ratio was 27 IU/ML (16 186). There was positive ANA with titre and pattern in 34 patients (85.0%), positive antids-DNA in 25 patients (62.5%), and positive anticardiolipin IgM and IgG in 5 patients (12.5%). Renal biopsy was found to be normal in 23 (57.5 percent), lupus nephritis class II, III in 3 (7.5 percent), lupus nephritis class III in 10 (25.0%), and lupus nephritis class IV in 4 (10.0%). Urine analysis results showed the following: normal in 28 (70.0%), albumin in 2 (5.0%), casts in 2 (5.0%), pus cell in 4 (10.0%), albumin + casts in 2 (5.0%) and albumin + pus cell in 2 (5.0%). Regarding semaphorin 3A level, a highly statistically significant variance was noted among the control & patients group concerning semaphorin 3A level found to be lower in cases than control with a p-value below 0.001. In patients' group, a negative correlation for semaphorin 3A with SBP, DBP, AST and ESR and also a positive correlation with steroid duration in the studied pa-tients. In addition, highly significant association between semaphorin 3A & positive CRP. How-ever, no significant relationship between semaphorin 3A & SLE manifestations except arthritis was found related to semaphorin 3A level. ROC curve shows that the semaphorin 3A cut-off point to predict SLE ≤ 3 with sensitivity = 47.50, specificity=92.50, PPV=86.4, and NPV=63.8. CONCLUSION: Reduced plasma Semaphorin 3A levels were found in this study; furthermore, their clinical relationship in SLE proposes their significant job in this illness. Furthermore, the ROC results demonstrated that Semaphorin 3A could be a new symptomatic biomarker in SLE with very high sensitivity for the determination of SLE, demonstrating that they might be helpful bi-omarkers for the evaluation of SLE. However, extra studies that focus on the potential role of Semaphorin 3A in SLE are needed.

8.
Colloids Surf B Biointerfaces ; 242: 114083, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39029246

ABSTRACT

Polysaccharides have garnered significant attention as potential nanoparticle carriers for targeted tumor therapy due to their excellent biodegradability and biocompatibility. Polyguluronic acid (PG) is a homogeneous acidic polysaccharide fragment derived from alginate, which is found in brown algae, possesses excellent bioactivities, unique properties. This study explored the immunomodulatory activity of PG and developed PG-based nanogels through modified disulfide bonds and Ca2+ dual crosslinking. We characterized their structure, assessed their drug-loading and release properties, and ultimately validated both the safety of the nanocarrier and the in vitro anti-tumor efficacy of the encapsulated drug. Results indicated that PG significantly enhanced the proliferative activity and phagocytosis of RAW264.7 cells while promoting reactive oxygen species (ROS) production and cytokine secretion. The study identified TLR4 as the primary receptor for PG recognition in RAW264.7 cells. Furthermore, PG-based drug-carrying nanogels were prepared, exhibiting uniform sizes of about 184 nm and demonstrating exceptional encapsulation efficiency (82.15 ± 0.82 %) and drug loading capacity (8.12 ± 0.08 %). In vitro release experiments showed that these nanogels could responsively release drugs under conditions of high glutathione (GSH) reduction, facilitating drug accumulation at tumor sites and enhancing therapeutic efficacy. This research not only expands the application of PG in drug delivery systems but also provides valuable insights into leveraging natural immunomodulatory polysaccharides as carriers for targeted drug delivery.

9.
Front Immunol ; 15: 1423510, 2024.
Article in English | MEDLINE | ID: mdl-38975338

ABSTRACT

Over the course of evolution, many proteins have undergone adaptive structural changes to meet the increasing homeostatic regulatory demands of multicellularity. Aminoacyl tRNA synthetases (aaRS), enzymes that catalyze the attachment of each amino acid to its cognate tRNA, are such proteins that have acquired new domains and motifs that enable non-canonical functions. Through these new domains and motifs, aaRS can assemble into large, multi-subunit complexes that enhance the efficiency of many biological functions. Moreover, because the complexity of multi-aminoacyl tRNA synthetase (mARS) complexes increases with the corresponding complexity of higher eukaryotes, a contribution to regulation of homeostatic functions in multicellular organisms is hypothesized. While mARS complexes in lower eukaryotes may enhance efficiency of aminoacylation, little evidence exists to support a similar role in chordates or other higher eukaryotes. Rather, mARS complexes are reported to regulate multiple and variegated cellular processes that include angiogenesis, apoptosis, inflammation, anaphylaxis, and metabolism. Because all such processes are critical components of immune homeostasis, it is important to understand the role of mARS complexes in immune regulation. Here we provide a conceptual analysis of the current understanding of mARS complex dynamics and emerging mARS complex roles in immune regulation, the increased understanding of which should reveal therapeutic targets in immunity and immune-mediated disease.


Subject(s)
Amino Acyl-tRNA Synthetases , Homeostasis , Homeostasis/immunology , Animals , Humans , Amino Acyl-tRNA Synthetases/immunology , Amino Acyl-tRNA Synthetases/metabolism , Immunomodulation
10.
Adv Exp Med Biol ; 1445: 11-36, 2024.
Article in English | MEDLINE | ID: mdl-38967747

ABSTRACT

Although V(D)J recombination and immunoglobulin (Ig) production are traditionally recognised to occur only in B lymphocytes and plasma cells, the expression of Igs in non-lymphoid cells, which we call non B cell-derived Igs (non B Igs), has been documented by growing studies. It has been demonstrated that non B-Igs can be widely expressed in most cell types, including, but not limited to, epithelial cells, cardiomyocytes, hematopoietic stem/progenitor cells, myeloid cells, and cells from immune-privileged sites, such as neurons and spermatogenic cells. In particular, malignant tumour cells express high level of IgG. Moreover, different from B-Igs that mainly localised on the B cell membrane and in the serum and perform immune defence function mainly, non B-Igs have been found to distribute more widely and play critical roles in immune defence, maintaining cell proliferation and survival, and promoting progression. The findings of non B-Igs may provide a wealthier breakthrough point for more therapeutic strategies for a wide range of immune-related diseases.


Subject(s)
Immunoglobulins , Humans , Animals , Immunoglobulins/genetics , Immunoglobulins/metabolism , Immunoglobulins/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Epithelial Cells/metabolism , Epithelial Cells/immunology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism
11.
Acta Pharm Sin B ; 14(6): 2732-2747, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38828144

ABSTRACT

The progression of ulcerative colitis (UC) is associated with immunologic derangement, intestinal hemorrhage, and microbiota imbalance. While traditional medications mainly focus on mitigating inflammation, it remains challenging to address multiple symptoms. Here, a versatile gas-propelled nanomotor was constructed by mild fusion of post-ultrasonic CaO2 nanospheres with Cu2O nanoblocks. The resulting CaO2-Cu2O possessed a desirable diameter (291.3 nm) and a uniform size distribution. It could be efficiently internalized by colonic epithelial cells and macrophages, scavenge intracellular reactive oxygen/nitrogen species, and alleviate immune reactions by pro-polarizing macrophages to the anti-inflammatory M2 phenotype. This nanomotor was found to penetrate through the mucus barrier and accumulate in the colitis mucosa due to the driving force of the generated oxygen bubbles. Rectal administration of CaO2-Cu2O could stanch the bleeding, repair the disrupted colonic epithelial layer, and reduce the inflammatory responses through its interaction with the genes relevant to blood coagulation, anti-oxidation, wound healing, and anti-inflammation. Impressively, it restored intestinal microbiota balance by elevating the proportions of beneficial bacteria (e.g., Odoribacter and Bifidobacterium) and decreasing the abundances of harmful bacteria (e.g., Prevotellaceae and Helicobacter). Our gas-driven CaO2-Cu2O offers a promising therapeutic platform for robust treatment of UC via the rectal route.

12.
Heliyon ; 10(11): e31747, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38828304

ABSTRACT

The study investigated the protective effects and mechanisms of probiotics in conjunction with an anti-PD-L1 antibody on the immune functions of septic mice. Sixty-four mice were assigned to sepsis groups receiving vehicle, probiotics, and anti-PD-L1 antibody individually or in combination, with healthy mice as controls. Sepsis was induced by cecal ligation and puncture (CLP), followed by intraperitoneal Lipopolysaccharide (LPS) injection. Blood and tissues were collected one day post-injection for detecting inflammation-related cytokines, Treg, PI3K/Akt pathway-related protein expression, and lung tissue pathology. The survival time of the remaining ten mice was recorded over seven days. Compared to healthy mice, septic mice given PBS exhibited significantly different serum levels of IL-6, IL-8, IL-17, IL-10, and IFN-γ (all p < 0.001). Treatment with anti-PD-L1 antibody combined with probiotics significantly increased the 7-day survival rate in septic mice, accompanied by decreased pro-inflammatory cytokines, increased anti-inflammatory cytokines, improved oxidative stress, reduced lung injury, and enhanced Th17/Treg balance. This combined therapy demonstrated superior efficacy compared to antibodies or probiotics alone. Additionally, it facilitated peripheral blood polymorphonuclear neutrophil apoptosis, enhancing protection by blocking PD-L1 function and inhibiting PI3K-dependent AKT phosphorylation. In conclusion, combining probiotics with an anti-PD-L1 antibody enhances protective effects in septic mice by reducing serum inflammatory factors, promoting neutrophil apoptosis, regulating Th17/Treg balance, and inhibiting the PI3K/Akt pathway.

13.
Front Immunol ; 15: 1415573, 2024.
Article in English | MEDLINE | ID: mdl-38835772

ABSTRACT

Efferocytosis, the process of engulfing and removing apoptotic cells, plays an essential role in preserving tissue health and averting undue inflammation. While macrophages are primarily known for this task, dendritic cells (DCs) also play a significant role. This review delves into the unique contributions of various DC subsets to efferocytosis, highlighting the distinctions in how DCs and macrophages recognize and handle apoptotic cells. It further explores how efferocytosis influences DC maturation, thereby affecting immune tolerance. This underscores the pivotal role of DCs in orchestrating immune responses and sustaining immune equilibrium, providing new insights into their function in immune regulation.


Subject(s)
Dendritic Cells , Macrophages , Phagocytosis , Dendritic Cells/immunology , Humans , Phagocytosis/immunology , Animals , Macrophages/immunology , Apoptosis/immunology , Immune Tolerance , Efferocytosis
14.
Front Immunol ; 15: 1401867, 2024.
Article in English | MEDLINE | ID: mdl-38846947

ABSTRACT

Tuberculosis (TB), caused by the bacterial pathogen Mycobacterium tuberculosis (MTB), remains one of the most prevalent and deadly infectious diseases worldwide. Currently, there are complex interactions between host cells and pathogens in TB. The onset, progression, and regression of TB are correlated not only with the virulence of MTB but also with the immunity of TB patients. Exosomes are cell-secreted membrane-bound nanovesicles with lipid bilayers that contain a variety of biomolecules, such as metabolites, lipids, proteins, and nucleic acids. Exosome-mediated cell-cell communication and interactions with the microenvironment represent crucial mechanisms through which exosomes exert their functional effects. Exosomes harbor a wide range of regulatory roles in physiological and pathological conditions, including MTB infection. Exosomes can regulate the immune response, metabolism, and cellular death to remodel the progression of MTB infection. During MTB infection, exosomes display distinctive profiles and quantities that may act as diagnostic biomarkers, suggesting that exosomes provide a revealing glimpse into the evolving landscape of MTB infections. Furthermore, exosomes derived from MTB and mesenchymal stem cells can be harnessed as vaccine platforms and drug delivery vehicles for the precise targeting and treatment of TB. In this review, we highlight the functions and mechanisms through which exosomes influence the progression of TB. Additionally, we unravel the critical significance of exosomal constituents in the diagnosis and therapeutic applications of TB, aiming to offer novel perspectives and strategies for combating TB.


Subject(s)
Biomarkers , Exosomes , Mycobacterium tuberculosis , Tuberculosis , Exosomes/immunology , Exosomes/metabolism , Humans , Tuberculosis/immunology , Tuberculosis/diagnosis , Tuberculosis/therapy , Tuberculosis/microbiology , Mycobacterium tuberculosis/immunology , Animals , Antitubercular Agents/therapeutic use
15.
Cancer Lett ; 597: 217084, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38925362

ABSTRACT

The intricate interplay among extracellular vesicles, cancer stemness properties, and the immune system significantly impacts hepatocellular carcinoma (HCC) progression, treatment response, and patient prognosis. Extracellular vesicles (EVs), which are membrane-bound structures, play a pivotal role in conveying proteins, lipids, and nucleic acids between cells, thereby serving as essential mediators of intercellular communication. Since a lot of current research focuses on small extracellular vesicles (sEVs), with diameters ranging from 30 nm to 200 nm, this review emphasizes the role of sEVs in the context of interactions between HCC stemness-bearing cells and the immune cells. sEVs offer promising opportunities for the clinical application of innovative diagnostic and prognostic biomarkers in HCC. By specifically targeting sEVs, novel therapeutics aimed at cancer stemness can be developed. Ongoing investigations into the roles of sEVs in cancer stemness and immune regulation in HCC will broaden our understanding and ultimately pave the way for groundbreaking therapeutic interventions.

16.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892139

ABSTRACT

Maternal obesity and over/undernutrition can have a long-lasting impact on offspring health during critical periods in the first 1000 days of life. Children born to mothers with obesity have reduced immune responses to stimuli which increase susceptibility to infections. Recently, maternal western-style diets (WSDs), high in fat and simple sugars, have been associated with skewing neonatal immune cell development, and recent evidence suggests that dysregulation of innate immunity in early life has long-term consequences on metabolic diseases and behavioral disorders in later life. Several factors contribute to abnormal innate immune tolerance or trained immunity, including changes in gut microbiota, metabolites, and epigenetic modifications. Critical knowledge gaps remain regarding the mechanisms whereby these factors impact fetal and postnatal immune cell development, especially in precursor stem cells in bone marrow and fetal liver. Components of the maternal microbiota that are transferred from mothers consuming a WSD to their offspring are understudied and identifying cause and effect on neonatal innate and adaptive immune development needs to be refined. Tools including single-cell RNA-sequencing, epigenetic analysis, and spatial location of specific immune cells in liver and bone marrow are critical for understanding immune system programming. Considering the vital role immune function plays in offspring health, it will be important to understand how maternal diets can control developmental programming of innate and adaptive immunity.


Subject(s)
Diet, Western , Fetal Development , Prenatal Exposure Delayed Effects , Humans , Female , Pregnancy , Diet, Western/adverse effects , Animals , Fetal Development/immunology , Prenatal Exposure Delayed Effects/immunology , Immune System/immunology , Immune System/metabolism , Epigenesis, Genetic , Gastrointestinal Microbiome/immunology , Immunity, Innate , Maternal Nutritional Physiological Phenomena , Fetus/immunology
17.
Cancers (Basel) ; 16(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38893071

ABSTRACT

Melanoma is commonly diagnosed in a younger population than most other solid malignancies and, in Australia and most of the world, is the leading cause of skin-cancer-related death. Melanoma is a cancer type with high immunogenicity; thus, immunotherapies are used as first-line treatment for advanced melanoma patients. Although immunotherapies are working well, not all the patients are benefitting from them. A lack of a comprehensive understanding of immune regulation in the melanoma tumour microenvironment is a major challenge of patient stratification. Overexpression of CD155 has been reported as a key factor in melanoma immune regulation for the development of therapy resistance. A more thorough understanding of the actions of current immunotherapy strategies, their effects on immune cell subsets, and the roles that CD155 plays are essential for a rational design of novel targets of anti-cancer immunotherapies. In this review, we comprehensively discuss current anti-melanoma immunotherapy strategies and the immune response contribution of different cell lineages, including tumour endothelial cells, myeloid-derived suppressor cells, cytotoxic T cells, cancer-associated fibroblast, and nature killer cells. Finally, we explore the impact of CD155 and its receptors DNAM-1, TIGIT, and CD96 on immune cells, especially in the context of the melanoma tumour microenvironment and anti-cancer immunotherapies.

18.
Biomolecules ; 14(6)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38927085

ABSTRACT

Ubiquitin-specific protease 5 (USP5) belongs to the ubiquitin-specific protease (USP) family, which uniquely recognizes unanchored polyubiquitin chains to maintain the homeostasis of monoubiquitin chains. USP5 participates in a wide range of cellular processes by specifically cleaving isopeptide bonds between ubiquitin and substrate proteins or ubiquitin itself. In the process of immune regulation, USP5 affects important cellular signaling pathways, such as NF-κB, Wnt/ß-catenin, and IFN, by regulating ubiquitin-dependent protein degradation. These pathways play important roles in immune regulation and inflammatory responses. In addition, USP5 regulates the activity and function of immunomodulatory signaling pathways via the deubiquitination of key proteins, thereby affecting the activity of immune cells and the regulation of immune responses. In the present review, the structure and function of USP5, its role in immune regulation, and the mechanism by which USP5 affects the development of diseases by regulating immune signaling pathways are comprehensively overviewed. In addition, we also introduce the latest research progress of targeting USP5 in the treatment of related diseases, calling for an interdisciplinary approach to explore the therapeutic potential of targeting USP5 in immune regulation.


Subject(s)
Signal Transduction , Humans , Animals , Endopeptidases/metabolism , Ubiquitination , Immunomodulation
19.
Immunol Res ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937380

ABSTRACT

T regulatory lymphocytes (Treg) expressing CCR5 exhibit strong suppression activity in various autoimmune disorders. However, there remains a lack of comprehensive understanding regarding their involvement in the development of type 1 diabetes (T1D). In this study, we examined the role of the CCR5/CCL5 axis in regulating inflammatory response and its impact on regulatory T cells in type 1 diabetes (T1D). We hypothesize that dysregulation of the CCR5/CCL5 axis contributes to the development and progression of T1D through modulation of Treg-dependent immune responses. We analyzed the expression levels of CCR5 on Tregs isolated from individuals with T1D, as well as the plasma concentration of its main ligands. We found that Tregs from T1D patients exhibited decreased expression of CCR5 compared to healthy controls. Additionally, we observed a correlation between the expression levels of CCR5 on Tregs and their immunosuppressive function in T1D patients. Our results indicate the impaired migratory capacity of CCR5 + Tregs, suggesting a possible link between the dysregulation of the CCR5/CCL5 axis and impaired immune regulation in T1D. In line with previous studies, our findings support the notion that dysregulation of the CCR5/CCL5 axis contributes to the development and progression of type 1 diabetes (T1D) by modulating Treg-dependent immune responses. The decreased expression of CCR5 on Tregs in T1D patients suggests a potential impairment in the migratory capacity of these cells, which could compromise their ability to suppress autoreactive T cells and maintain immune homeostasis. Furthermore, our study highlights the importance of CCR5 as a biomarker for identifying dysfunctional Tregs in T1D.

20.
Dev Comp Immunol ; 159: 105217, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38901503

ABSTRACT

Norepinephrine (NE) is involved in regulating cytokine expression and phagocytosis of immune cells in the innate immunity of vertebrates. In the present study, the modulation mechanism of NE on the biosynthesis of TNFs in oyster granulocytes was explored. The transcripts of CgTNF-1, CgTNF-2 and CgTNF-3 were highly expressed in granulocytes, and they were significantly up-regulated after LPS stimulation, while down-regulated after NE treatment. The phagocytic rate and apoptosis index of oyster granulocytes were also triggered by LPS stimulation and suppressed by NE treatment. The mRNA expressions of CgMAPK14 and CgRelish were significantly induced after NE treatment, and the translocation of CgRelish from cytoplasm to nucleus was observed. The concentration of intracellular Ca2+ in granulocytes was significantly up-regulated upon NE incubation, and this trend reverted after the treatment with DOX (specific antagonist for NE receptor, CgA1AR-1). No obvious significance was observed in intracellular cAMP concentrations in the PBS, NE and NE + DOX groups. Once CgA1AR-1 was blocked by DOX, the mRNA expressions of CgMAPK14 and CgRelish were significantly inhibited, and the translocation of CgRelish from cytoplasm to nucleus was also dramatically suppressed, while the mRNA expression of CgTNF-1 and the apoptosis index increased significantly to the same level with those in LPS group, respectively. These results collectively suggested that NE modulated TNF expression in oyster granulocyte through A1AR-p38 MAPK-Relish signaling pathway.


Subject(s)
Crassostrea , Granulocytes , Immunity, Innate , Lipopolysaccharides , Norepinephrine , p38 Mitogen-Activated Protein Kinases , Animals , Crassostrea/immunology , Norepinephrine/metabolism , Norepinephrine/pharmacology , Granulocytes/immunology , Granulocytes/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/immunology , Apoptosis , Signal Transduction , Phagocytosis , Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , Gene Expression Regulation , MAP Kinase Signaling System/immunology , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...