Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.225
Filter
1.
Biomaterials ; 312: 122750, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39126779

ABSTRACT

Infiltration of immunosuppressive cells into the breast tumor microenvironment (TME) is associated with suppressed effector T cell (Teff) responses, accelerated tumor growth, and poor clinical outcomes. Previous studies from our group and others identified infiltration of immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) as critical contributors to immune dysfunction in the orthotopic claudin-low tumor model, limiting the efficacy of adoptive cellular therapy. However, approaches to target these cells in the TME are currently lacking. To overcome this barrier, polymeric micellular nanoparticles (PMNPs) were used for the co-delivery of small molecule drugs activating Toll-like receptors 7 and 8 (TLR7/8) and inhibiting PI3K delta (PI3Kδ). The immunomodulation of the TME by TLR7/8 agonist and PI3K inhibitor led to type 1 macrophage polarization, decreased MDSC accumulation and selectively decreased tissue-resident Tregs in the TME, while enhancing the T and B cell adaptive immune responses. PMNPs significantly enhanced the anti-tumor activity of local radiation therapy (RT) in mice bearing orthotopic claudin-low tumors compared to RT alone. Taken together, these data demonstrate that RT combined with a nanoformulated immunostimulant diminished the immunosuppressive TME resulting in tumor regression. These findings set the stage for clinical studies of this approach.


Subject(s)
Nanoparticles , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Tumor Microenvironment , Animals , Tumor Microenvironment/drug effects , Toll-Like Receptor 7/agonists , Female , Nanoparticles/chemistry , Mice , Toll-Like Receptor 8/agonists , Immunomodulation/drug effects , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Mice, Inbred BALB C , Micelles , Humans
2.
Front Immunol ; 15: 1447536, 2024.
Article in English | MEDLINE | ID: mdl-39224602

ABSTRACT

Mesenchymal stem cell derived extracellular vesicles (MSC EVs) are paracrine modulators of macrophage function. Scientific research has primarily focused on the immunomodulatory and regenerative properties MSC EVs derived from bone marrow. The dental pulp is also a source for MSCs, and their anatomical location and evolutionary function has primed them to be potent immunomodulators. In this study, we demonstrate that extracellular vesicles derived from dental pulp stem cells (DPSC EVs) have pronounced immunomodulatory effect on primary macrophages by regulating the NFκb pathway. Notably, the anti-inflammatory activity of DPSC-EVs is enhanced following exposure to an inflammatory stimulus (LPS). These inhibitory effects were also observed in vivo. Sequencing of the naïve and LPS preconditioned DPSC-EVs and comparison with our published results from marrow MSC EVs revealed that Naïve and LPS preconditioned DPSC-EVs are enriched with anti-inflammatory miRNAs, particularly miR-320a-3p, which appears to be unique to DPSC-EVs and regulates the NFκb pathway. Overall, our findings highlight the immunomodulatory properties of DPSC-EVs and provide vital clues that can stimulate future research into miRNA-based EV engineering as well as therapeutic approaches to inflammation control and disease treatment.


Subject(s)
Dental Pulp , Extracellular Vesicles , Immunomodulation , Inflammation , NF-kappa B , Dental Pulp/cytology , Dental Pulp/immunology , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Humans , Animals , Inflammation/immunology , Inflammation/metabolism , NF-kappa B/metabolism , Macrophages/immunology , Macrophages/metabolism , MicroRNAs/genetics , Lipopolysaccharides/pharmacology , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/immunology , Cells, Cultured , Signal Transduction , Stem Cells/immunology , Stem Cells/metabolism , Male
3.
Carbohydr Polym ; 346: 122586, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245482

ABSTRACT

Lentinan (LNT), a natural polysaccharide, has been reported to exhibit immunomodulatory effects in the intestine after oral administration. Herein, we aimed to investigate the lymphatic transport of LNT in Peyer's patches (PPs) by traceable fluorescent labeling and to explore whether/how LNT contacts related immune cells. Near-infrared imaging confirmed the absorption of LNT in the small intestinal segment and its accumulation within PPs after oral administration. Subsequently, tissue imaging confirmed that M cells are the main cells responsible for transporting LNT to PPs, and an M cell model was established to explore the involvement of Dectin-1 in the absorption process. Systematic in vitro and in vivo studies revealed that the Dectin-1 further mediates the uptake of LNT by mononuclear phagocytes in PPs. Moreover, LNT can promote the proliferation and differentiation of mononuclear phagocytes, thereby activating immune responses. In summary, this study elucidates the pharmacokinetic mechanisms by which LNT exerts oral immunomodulatory effects, providing a theoretical basis for the development and application of other polysaccharides.


Subject(s)
Lectins, C-Type , Lentinan , Peyer's Patches , Peyer's Patches/immunology , Peyer's Patches/drug effects , Peyer's Patches/metabolism , Animals , Lentinan/pharmacology , Lentinan/chemistry , Lectins, C-Type/metabolism , Mice , Administration, Oral , Phagocytes/drug effects , Phagocytes/metabolism , Phagocytes/immunology , Immunomodulation/drug effects , Male , Mice, Inbred BALB C , M Cells
4.
Curr Microbiol ; 81(10): 346, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39240286

ABSTRACT

The rise of methicillin-resistant Staphylococcus aureus (MRSA) poses a significant challenge in clinical settings due to its ability to evade conventional antibiotic treatments. This overview explores the potential of immunomodulatory strategies as alternative therapeutic approaches to combat MRSA infections. Traditional antibiotics are becoming less effective, necessitating innovative solutions that harness the body's immune system to enhance pathogen clearance. Recent advancements in immunotherapy, including the use of antimicrobial peptides, phage therapy, and mechanisms of immune cells, demonstrate promise in enhancing the body's ability to clear MRSA infections. However, the exact interactions between these therapies and immunomodulation are not fully understood, underscoring the need for further research. Hence, this review aims to provide a broad overview of the current understanding of non-traditional therapeutics and their impact on immune responses, which could lead to more effective MRSA treatment strategies. Additionally, combining immunomodulatory agents with existing antibiotics may improve outcomes, particularly for immunocompromised patients or those with chronic infections. As the landscape of antibiotic resistance evolves, the development of effective immunotherapeutic strategies could play a vital role in managing MRSA infections and reducing reliance on traditional antibiotics. Future research must focus on optimizing these approaches and validating their efficacy in diverse clinical populations to address the urgent need for effective MRSA management strategies.


Subject(s)
Immunomodulation , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Methicillin-Resistant Staphylococcus aureus/drug effects , Humans , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Immunotherapy/methods , Phage Therapy/methods , Animals , Immunomodulating Agents/pharmacology , Immunomodulating Agents/therapeutic use , Immunologic Factors
5.
Front Immunol ; 15: 1413179, 2024.
Article in English | MEDLINE | ID: mdl-39247182

ABSTRACT

Inflammation is a normal immune response in organisms, but it often triggers chronic diseases such as colitis and arthritis. Currently, the most widely used anti-inflammatory drugs are non-steroidal anti-inflammatory drugs, albeit they are accompanied by various adverse effects such as hypertension and renal dysfunction. Bioactive peptides (BAPs) provide therapeutic benefits for inflammation and mitigate side effects. Herein, this review focuses on the therapeutic effects of various BAPs on inflammation in different body parts. Emphasis is placed on the immunomodulatory mechanisms of BAPs in treating inflammation, such as regulating the release of inflammatory mediators, modulating MAPK and NF-κB signaling pathways, and reducing oxidative stress reactions for immunomodulation. This review aims to provide a reference for the function, application, and anti-inflammation mechanisms of BAPs.


Subject(s)
Inflammation , Peptides , Humans , Inflammation/drug therapy , Inflammation/immunology , Peptides/therapeutic use , Peptides/pharmacology , Animals , Signal Transduction/drug effects , Oxidative Stress/drug effects , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , NF-kappa B/metabolism , Inflammation Mediators/metabolism , Immunomodulation/drug effects
6.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39126007

ABSTRACT

Diabetic retinopathy (DR) is one of the most prevalent secondary complications associated with diabetes. Specifically, Type 1 Diabetes Mellitus (T1D) has an immune component that may determine the evolution of DR by compromising the immune response of the retina, which is mediated by microglia. In the early stages of DR, the permeabilization of the blood-retinal barrier allows immune cells from the peripheral system to interact with the retinal immune system. The use of new bioactive molecules, such as 3-(2,4-dihydroxyphenyl)phthalide (M9), with powerful anti-inflammatory activity, might represent an advance in the treatment of diseases like DR by targeting the immune systems responsible for its onset and progression. Our research aimed to investigate the molecular mechanisms involved in the interaction of specific cells of the innate immune system during the progression of DR and the reduction in inflammatory processes contributing to the pathology. In vitro studies were conducted exposing Bv.2 microglial and Raw264.7 macrophage cells to proinflammatory stimuli for 24 h, in the presence or absence of M9. Ex vivo and in vivo approaches were performed in BB rats, an animal model for T1D. Retinal explants from BB rats were cultured with M9. Retinas from BB rats treated for 15 days with M9 via intraperitoneal injection were analyzed to determine survival, cellular signaling, and inflammatory markers using qPCR, Western blot, or immunofluorescence approaches. Retinal structure images were acquired via Spectral-Domain-Optical Coherence Tomography (SD-OCT). Our results show that the treatment with M9 significantly reduces inflammatory processes in in vitro, ex vivo, and in vivo models of DR. M9 works by inhibiting the proinflammatory responses during DR progression mainly affecting immune cell responses. It also induces an anti-inflammatory response, primarily mediated by microglial cells, leading to the synthesis of Arginase-1 and Hemeoxygenase-1(HO-1). Ultimately, in vivo administration of M9 preserves the retinal integrity from the degeneration associated with DR progression. Our findings demonstrate a specific interaction between both retinal and systemic immune cells in the progression of DR, with a differential response to treatment, mainly driven by microglia in the anti-inflammatory action. In vivo treatment with M9 induces a switch in immune cell phenotypes and functions that contributes to delaying the DR progression, positioning microglial cells as a new and specific therapeutic target in DR.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetic Retinopathy , Disease Models, Animal , Microglia , Animals , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/pathology , Diabetic Retinopathy/immunology , Rats , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/complications , Mice , Microglia/drug effects , Microglia/metabolism , Retina/drug effects , Retina/pathology , Retina/metabolism , RAW 264.7 Cells , Male , Benzofurans/pharmacology , Benzofurans/therapeutic use , Immunomodulation/drug effects , Inflammation/drug therapy , Inflammation/pathology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Rats, Inbred BB
7.
J Appl Biomater Funct Mater ; 22: 22808000241266665, 2024.
Article in English | MEDLINE | ID: mdl-39129373

ABSTRACT

The pro-inflammatory/anti-inflammatory polarized phenotypes of macrophages (M1/M2) can be used to predict the success of implant integration. Hence, activating and inducing the transformation of immunocytes that promote tissue repair appears to be a highly promising strategy for facilitating osteo-anagenesis. In a previous study, titanium implants were coated with a graphene oxide-hydroxyapatite (GO-HA) nanocomposite via electrophoretic deposition, and the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) was found to be significantly enhanced when the GO content was 2wt%. However, the effectiveness of the GO-HA nanocomposite coating in modifying the in vivo immune microenvironment still remains unclear. In this study, the effects of GO-HA coatings on osteogenesis were investigated based on the GO-HA-mediated immune regulation of macrophages. The HA-2wt%GO nanocomposite coatings exhibited good biocompatibility and favored M2 macrophage polarization. Meanwhile, they could also significantly upregulate IL-10 (anti-inflammatory factor) expression and downregulate TNF-α (pro-inflammatory factor) expression. Additionally, the microenvironment, which was established by M2 macrophages, favored the osteogenesis of BMSCs both in vivo and in vitro. These findings show that the GO-HA nanocomposite coating is a promising surface-modification material. Hence, this study provides a reference for the development of next-generation osteoimmunomodulatory biomaterials.


Subject(s)
Coated Materials, Biocompatible , Durapatite , Graphite , Macrophages , Mesenchymal Stem Cells , Osseointegration , Osteogenesis , Osseointegration/drug effects , Durapatite/chemistry , Durapatite/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/cytology , Animals , Graphite/chemistry , Graphite/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Osteogenesis/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Prostheses and Implants , Immunomodulation/drug effects , Nanocomposites/chemistry , RAW 264.7 Cells , Cell Differentiation/drug effects , Titanium/chemistry , Titanium/pharmacology , Male
8.
ACS Nano ; 18(32): 21512-21522, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39096486

ABSTRACT

Although minimally invasive interventional occluders can effectively seal heart defect tissue, they still have some limitations, including poor endothelial healing, intense inflammatory response, and thrombosis formation. Herein, a polyphenol-reinforced medicine/peptide glycocalyx-like coating was prepared on cardiac occluders. A coating consisting of carboxylated chitosan, epigallocatechin-3-gallate (EGCG), tanshinone IIA sulfonic sodium (TSS), and hyaluronic acid grafted with 3-aminophenylboronic acid was prepared. Subsequently, the mercaptopropionic acid-GGGGG-Arg-Glu-Asp-Val peptide was grafted by the thiol-ene "click" reaction. The coating showed good hydrophilicity and free radical-scavenging ability and could release EGCG-TSS. The results of biological experiments suggested that the coating could reduce thrombosis by promoting endothelialization, and promote myocardial repair by regulating the inflammatory response. The functions of regulating cardiomyocyte apoptosis and metabolism were confirmed, and the inflammatory regulatory functions of the coating were mainly dependent on the NF-kappa B and TNF signaling pathway.


Subject(s)
Glycocalyx , Hydrogels , Polyphenols , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Polyphenols/chemistry , Polyphenols/pharmacology , Glycocalyx/metabolism , Glycocalyx/chemistry , Glycocalyx/drug effects , Immunomodulation/drug effects , Regeneration/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Apoptosis/drug effects , Mice , Myocardium/metabolism , Catechin/chemistry , Catechin/analogs & derivatives , Catechin/pharmacology , Rats, Sprague-Dawley , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Male
9.
Int J Immunopathol Pharmacol ; 38: 3946320241276899, 2024.
Article in English | MEDLINE | ID: mdl-39162714

ABSTRACT

BACKGROUND: Induced Pluripotent Stem Cells (IPSCs) represent an innovative strategy for addressing challenging diseases, including various rheumatologic conditions. Aside from their regenerative capacities, some studies have shown the potential of these cells in the modulation of inflammatory responses. The underlying mechanisms by which they exert their effects have yet to be fully comprehended. Therefore, we aimed to explore the gene expression linked to the IGF pathway as well as IL-10 and TGF-ß, which are known to exert immunomodulatory effects. METHODS: A C57/Bl6 pregnant mouse was used for obtaining mouse embryonic fibroblasts (MEFs), then the IPSCs were induced using lentiviral vectors expressing the pluripotency genes (OCT4, SOX2, KLF1, and c-MYC). Cells were cultured for 72 h in DMEM high glucose plus leukemia inhibitory factor; Evaluating the gene expression was conducted using specific primers for Igf1, Igf2, Igfbp3, Igfbp4, Irs1, Il-10, and Tgf-ß genes, as well as SYBR green qPCR master mix. The data were analyzed using the 2-ΔΔCT method and were compared by employing the t test; the results were plotted using GraphPad PRISM software. MEFs were utilized as controls. RESULTS: Gene expression analyses revealed that Igf-1, Igf-bp3, Igf-bp4, and Il-10 were significantly overexpressed (p ≤ .01), while Igf-2 and Tgf-b genes were significantly downregulated in the lysates from IPSCs in comparison with the control MEFs. The Irs1 gene expression was not altered significantly. CONCLUSION: IPSCs are potentially capable of modulating inflammatory responses through the expression of various anti-inflammatory mediators from the IGF signaling, as well as IL-10. This discovery uncovers a previously unknown dimension of IPSCs' therapeutic effects, potentially leading to more advanced in vivo research and subsequent clinical trials.


Subject(s)
Induced Pluripotent Stem Cells , Interleukin-10 , Mice, Inbred C57BL , Animals , Interleukin-10/genetics , Interleukin-10/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice , Female , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Cells, Cultured , Fibroblasts/metabolism , Pregnancy , Immunomodulation/genetics
10.
J Extracell Vesicles ; 13(8): e12497, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39140452

ABSTRACT

Extracellular vesicles (EVs) derived from mesenchymal stem/stromal cells (MSCs) have been recognized as promising cytotherapeutics due to their demonstrated immunomodulatory effects in various preclinical models. The immunomodulatory capabilities of EVs stem from the proteins and genetic materials they carry from parent cells, but the cargo contents of EVs are significantly influenced by MSC tissues and donors, cellular age and culture conditions, resulting in functional variations. However, there are no surrogate assays available to validate the immunomodulatory potency of MSC-EVs before in vivo administration. In previous work, we discovered that microcarrier culture conditions enhance the immunomodulatory function of MSC-EVs, as well as the levels of immunosuppressive molecules such as TGF-ß1 and let-7b in MSC-EVs. Building on these findings, we investigated whether TGF-ß1 levels in MSC-EVs could serve as a surrogate biomarker for predicting their potency in vivo. Our studies revealed a strong correlation between TGF-ß1 and let-7b levels in MSC-EVs, as well as their capacity to suppress IFN-γ secretion in stimulated splenocytes, establishing biopotency and surrogate assays for MSC-EVs. Subsequently, we validated MSC-EVs generated from monolayer cultures (ML-EVs) or microcarrier cultures (MC-EVs) using murine models of experimental autoimmune uveoretinitis (EAU) and additional in vitro assays reflecting the Mode of Action of MSC-EVs in vivo. Our findings demonstrated that MC-EVs carrying high levels of TGF-ß1 exhibited greater efficacy than ML-EVs in halting disease progression in mice with EAU as well as inducing apoptosis and inhibiting the chemotaxis of retina-reactive T cells. Additionally, MSC-EVs suppressed the MAPK/ERK pathway in activated T cells, with treatment using TGF-ß1 or let-7b showing similar effects on the MAPK/ERK pathway. Collectively, our data suggest that MSC-EVs directly inhibit the infiltration of retina-reactive T cells toward the eyes, thereby halting the disease progression in EAU mice, and their immunomodulatory potency in vivo can be predicted by their TGF-ß1 levels.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Transforming Growth Factor beta1 , Uveitis , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Mice , Uveitis/therapy , Uveitis/immunology , Uveitis/metabolism , Transforming Growth Factor beta1/metabolism , MicroRNAs/metabolism , Autoimmune Diseases/therapy , Autoimmune Diseases/immunology , Disease Models, Animal , Immunomodulation , Mice, Inbred C57BL , Humans , Female
11.
Cells ; 13(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39120320

ABSTRACT

Muscle-derived mesenchymal stromal cells (mdMSCs) hold great promise in regenerative medicine due to their immunomodulatory properties, multipotent differentiation capacity and ease of collection. However, traditional in vitro expansion methods use fetal bovine serum (FBS) and have numerous limitations including ethical concerns, batch-to-batch variability, immunogenicity, xenogenic contamination and regulatory compliance issues. This study investigates the use of 10% equine platelet lysate (ePL) obtained by plasmapheresis as a substitute for FBS in the culture of mdMSCs in innovative 2D and 3D models. Using muscle microbiopsies as the primary cell source in both models showed promising results. Initial investigations indicated that small variations in heparin concentration in 2D cultures strongly influenced medium coagulation with an optimal proliferation observed at final heparin concentrations of 1.44 IU/mL. The two novel models investigated showed that expansion of mdMSCs is achievable. At the end of expansion, the 3D model revealed a higher total number of cells harvested (64.60 ± 5.32 million) compared to the 2D culture (57.20 ± 7.66 million). Trilineage differentiation assays confirmed the multipotency (osteoblasts, chondroblasts and adipocytes) of the mdMSCs generated in both models with no significant difference observed. Immunophenotyping confirmed the expression of the mesenchymal stem cell (MSC) markers CD-90 and CD-44, with low expression of CD-45 and MHCII markers for mdMSCs derived from the two models. The generated mdMSCs also had great immunomodulatory properties. Specific immunological extraction followed by enzymatic detection (SIEFED) analysis demonstrated that mdMSCs from both models inhibited myeloperoxidase (MPO) activity in a strong dose-dependent manner. Moreover, they were also able to reduce reactive oxygen species (ROS) activity, with mdMSCs from the 3D model showing significantly higher dose-dependent inhibition compared to the 2D model. These results highlighted for the first time the feasibility and efficacy of using 10% ePL for mdMSC expansion in novel 2D and 3D approaches and also that mdMSCs have strong immunomodulatory properties that can be exploited to advance the field of regenerative medicine and cell therapy instead of using FBS with all its drawbacks.


Subject(s)
Blood Platelets , Cell Differentiation , Immunomodulation , Mesenchymal Stem Cells , Animals , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/immunology , Horses , Blood Platelets/metabolism , Cell Proliferation/drug effects , Cell Culture Techniques/methods , Cells, Cultured , Muscles , Immunophenotyping
12.
J Extracell Vesicles ; 13(8): e12472, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39092563

ABSTRACT

Recently, therapies utilizing extracellular vesicles (EVs) derived from mesenchymal stromal/stem cells (MSCs) have begun to show promise in clinical trials. However, EV therapeutic potential varies with MSC tissue source and in vitro expansion through passaging. To find the optimal MSC source for clinically translatable EV-derived therapies, this study aims to compare the angiogenic and immunomodulatory potentials and the protein and miRNA cargo compositions of EVs isolated from the two most common clinical sources of adult MSCs, bone marrow and adipose tissue, across different passage numbers. Primary bone marrow-derived MSCs (BMSCs) and adipose-derived MSCs (ASCs) were isolated from adult female Lewis rats and expanded in vitro to the indicated passage numbers (P2, P4, and P8). EVs were isolated from the culture medium of P2, P4, and P8 BMSCs and ASCs and characterized for EV size, number, surface markers, protein content, and morphology. EVs isolated from different tissue sources showed different EV yields per cell, EV sizes, and protein yield per EV. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of proteomics data and miRNA seq data identified key proteins and pathways associated with differences between BMSC-EVs and ASC-EVs, as well as differences due to passage number. In vitro tube formation assays employing human umbilical vein endothelial cells suggested that both tissue source and passage number had significant effects on the angiogenic capacity of EVs. With or without lipopolysaccharide (LPS) stimulation, EVs more significantly impacted expression of M2-macrophage genes (IL-10, Arg1, TGFß) than M1-macrophage genes (IL-6, NOS2, TNFα). By correlating the proteomics analyses with the miRNA seq analysis and differences observed in our in vitro immunomodulatory, angiogenic, and proliferation assays, this study highlights the trade-offs that may be necessary in selecting the optimal MSC source for development of clinical EV therapies.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Rats, Inbred Lew , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Female , Rats , Adipose Tissue/metabolism , Adipose Tissue/cytology , Neovascularization, Physiologic , Immunomodulation , Humans , Cells, Cultured , Cell Proliferation , Bone Marrow Cells/metabolism
13.
J Agric Food Chem ; 72(35): 19259-19273, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39177683

ABSTRACT

Probiotics are known to modulate host immune responses in the course of many diseases. Recently, bacterial extracellular vesicles (EVs), which contain bioactive proteins, lipids, nucleic acids, and metabolites released by bacteria, have been identified as potentially important mediators of bacteria-bacterium and bacteria-host interactions. With the deepening of research, it has been found that probiotic-derived EVs play a significant role in regulating host immune function and, thus, exerting health-promoting effects. Nevertheless, current research is in its early stages, and there remains a long way to go to bridge the gap between basic research and clinical practice. In this review, we describe the fundamental aspects of probiotic-derived EVs, including their biogenesis, cargo sorting mechanism, and transport capabilities. We further discussed the potential mechanisms of probiotic-derived EVs in regulating the host's gut microbiota and immune responses. Finally, we speculate about the potential of probiotic-derived EVs as new postbiotics for applications in functional food, disease treatment substitutes, and immune regulatory adjuvants.


Subject(s)
Bacteria , Extracellular Vesicles , Gastrointestinal Microbiome , Probiotics , Probiotics/pharmacology , Extracellular Vesicles/immunology , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Humans , Animals , Bacteria/immunology , Bacteria/metabolism , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry , Immunologic Factors/pharmacology , Immunomodulation
14.
Front Immunol ; 15: 1434027, 2024.
Article in English | MEDLINE | ID: mdl-39211038

ABSTRACT

A disintegrin and metalloproteinase domain 10 (ADAM10), a member of the ADAM family, is a cellular surface protein with potential adhesion and protease/convertase functions. The expression regulations in cancers by natural products [adenosine (AD) and its analogs, cordycepin (CD), and N6, N6-dimethyladenosine (m6 2A)], and immune regulation are unclear. As results, AD, CD, and m6 2A inhibited ADAM10 expression in various cancer cell lines, indicating their roles in anti-cancer agents. Further molecular docking with ADAM10 protein found the binding energies of all docking groups were <-7 kcal/mol for all small-molecules (AD, CD and m6 2A), suggesting very good binding activities. In addition, analysis of the immunomodulatory roles in cancer showed that ADAM10 was negatively correlated with immunomodulatory genes such as CCL27, CCL14, CCL25, CXCR5, HLA-B, HLA-DOB1, LAG3, TNFRSF18, and TNFRSF4 in bladder urothelial carcinoma, thymoma, breast invasive carcinoma, TGCT, kidney renal papillary cell carcinoma, SKCM and thyroid carcinoma, indicating the immune-promoting roles for ADAM10. LAG3 mRNA levels were reduced by both AD and CD in vivo. ADAM10 is also negatively associated with tumor immunosuppression and interrelated with the immune infiltration of tumors. Overall, the present study determined ADAM10 expression by AD, CD and m6 2A, and in AD or CD/ADAM10/LAG3 signaling in cancers, and suggested a potential method for immunotherapy of cancers by targeting ADAM10 using the small molecules AD, CD and m6 2A.


Subject(s)
ADAM10 Protein , Adenosine , Deoxyadenosines , Neoplasms , Humans , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/metabolism , Deoxyadenosines/pharmacology , Mice , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Amyloid Precursor Protein Secretases/metabolism , Molecular Docking Simulation , Membrane Proteins/metabolism , Membrane Proteins/genetics , Immunomodulation/drug effects , Female
15.
Front Immunol ; 15: 1421346, 2024.
Article in English | MEDLINE | ID: mdl-39211042

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a versatile environmental sensor and transcription factor found throughout the body, responding to a wide range of small molecules originating from the environment, our diets, host microbiomes, and internal metabolic processes. Increasing evidence highlights AhR's role as a critical regulator of numerous biological functions, such as cellular differentiation, immune response, metabolism, and even tumor formation. Typically located in the cytoplasm, AhR moves to the nucleus upon activation by an agonist where it partners with either the aryl hydrocarbon receptor nuclear translocator (ARNT) or hypoxia-inducible factor 1ß (HIF-1ß). This complex then interacts with xenobiotic response elements (XREs) to control the expression of key genes. AhR is notably present in various crucial immune cells, and recent research underscores its significant impact on both innate and adaptive immunity. This review delves into the latest insights on AhR's structure, activating ligands, and its multifaceted roles. We explore the sophisticated molecular pathways through which AhR influences immune and lymphoid cells, emphasizing its emerging importance in managing inflammatory diseases. Furthermore, we discuss the exciting potential of developing targeted therapies that modulate AhR activity, opening new avenues for medical intervention in immune-related conditions.


Subject(s)
Inflammation , Receptors, Aryl Hydrocarbon , Signal Transduction , Receptors, Aryl Hydrocarbon/metabolism , Humans , Animals , Inflammation/immunology , Inflammation/metabolism , Immunomodulation , Basic Helix-Loop-Helix Transcription Factors
16.
Int Immunopharmacol ; 140: 112889, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39128418

ABSTRACT

This study aimed to characterize PANoptosis-related genes with immunoregulatory features in osteoarthritis (OA) and investigate their potential diagnostic and therapeutic implications. Gene expression data from OA patients and healthy controls were obtained from the Gene Expression Omnibus (GEO) database. Differential expression analysis and functional enrichment analysis were conducted to identify PANoptosis-related genes (PRGs) associated with OA pathogenesis. A diagnostic model was developed using LASSO regression, and the diagnostic value of key PRGs was evaluated using Receiver Operating Characteristic Curve (ROC) analysis. The infiltration of immune cells and potential small molecule agents were also examined. A total of 39 differentially expressed PANoptosis-related genes (DE-PRGs) were identified, with functional enrichment analysis revealing their involvement in inflammatory response regulation and immune modulation pathways. Seven key PRGs, including CDKN1A, EZH2, MEG3, NR4A1, PIK3R2, S100A8, and SYVN1, were selected for diagnostic model construction, demonstrating high predictive performance in both training and validation datasets. The correlation between key PRGs and immune cell infiltration was explored. Additionally, molecular docking analysis identified APHA-compound-8 as a potential therapeutic agent targeting key PRGs. This study identified and analyzed PRGs in OA, uncovering their roles in immune regulation. Seven key PRGs were used to construct a diagnostic model with high predictive performance. The identified PRGs' correlation with immune cell infiltration was elucidated, and APHA-compound-8 was highlighted as a potential therapeutic agent. These findings offer novel diagnostic markers and therapeutic targets for OA, warranting further in vivo validation and exploration of clinical applications.


Subject(s)
Molecular Docking Simulation , Osteoarthritis , Humans , Osteoarthritis/genetics , Osteoarthritis/immunology , Gene Expression Profiling , Databases, Genetic , Immunomodulation/genetics
17.
Virulence ; 15(1): 2395831, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39185619

ABSTRACT

Salmonella enterica serovar Typhi (S. Typhi) causes typhoid fever, a systemic infection that affects millions of people worldwide. S. Typhi can invade and survive within host cells, such as intestinal epithelial cells and macrophages, by modulating their immune responses. However, the immunomodulatory capability of S. Typhi in relation to TolC-facilitated efflux pump function remains unclear. The role of TolC, an outer membrane protein that facilitates efflux pump function, in the invasion and immunomodulation of S. Typhi, was studied in human intestinal epithelial cells and macrophages. The tolC deletion mutant of S. Typhi was compared with the wild-type and its complemented strain in terms of their ability to invade epithelial cells, survive and induce cytotoxicity in macrophages, and elicit proinflammatory cytokine production in macrophages. The tolC mutant, which has a defective outer membrane, was impaired in invading epithelial cells compared to the wild-type strain, but the intracellular presence of the tolC mutant exhibited greater cytotoxicity and induced higher levels of proinflammatory cytokines (IL-1ß and IL-8) in macrophages compared to the wild-type strain. These effects were reversed by complementing the tolC mutant with a functional tolC gene. Our results suggest that TolC plays a role in S. Typhi to efficiently invade epithelial cells and suppress host immune responses during infection. TolC may be a potential target for the development of novel therapeutics against typhoid fever.


Subject(s)
Bacterial Outer Membrane Proteins , Epithelial Cells , Macrophages , Salmonella typhi , Typhoid Fever , Salmonella typhi/pathogenicity , Salmonella typhi/immunology , Salmonella typhi/genetics , Humans , Macrophages/microbiology , Macrophages/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/immunology , Epithelial Cells/microbiology , Epithelial Cells/immunology , Typhoid Fever/immunology , Typhoid Fever/microbiology , Immunomodulation , Cytokines/metabolism , Cytokines/immunology , Microbial Viability , Interleukin-8/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/immunology , Cell Line
18.
In Vivo ; 38(5): 2152-2164, 2024.
Article in English | MEDLINE | ID: mdl-39187336

ABSTRACT

BACKGROUND/AIM: Oral squamous cell carcinoma (OSCC) presents a significant health challenge, requiring effective treatments. Magnolol, a compound with potential anticancer properties, warrants investigation in OSCC treatment. Here, we aimed to assess the efficacy of magnolol in inhibiting progression of OSCC and to explore the underlying mechanisms of its action. MATERIALS AND METHODS: We evaluated the effect of magnolol on tumor progression using the MOC1-bearing orthotopic model. We examined its impact on pathology and toxicity through hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC), and biochemical analysis. We also investigated the immunoregulatory effects of magnolol in the MOC1-bearing model using flow cytometry. RESULTS: At high doses, magnolol significantly reduced tumor volume (p<0.0001 for comparisons between treated with magnolol and untreated groups) and weight loss by 70% in vivo. It also induced caspase-dependent apoptosis, evidenced by 2.42-, 2-, and 2.2-fold increases in the expression of caspase-3, -8, and -9, respectively, in mouse tumors treated with high 60 mg/kg of magnolol compared to untreated (p<0.0001 for all comparisons). Magnolol demonstrated no toxicity, maintaining body weight and normal biochemical parameters, including liver and kidney function. Pathological evaluations showed no adverse effects on organs in all treatment groups. Moreover, high doses of magnolol enhanced natural killer cells (by 3%), dendritic cells (20-25%), and cytotoxic T cells (20-40%) while reducing myeloid-derived suppressor cells and regulatory T cells by 1.5 times. CONCLUSION: Magnolol demonstrates potential as a therapeutic agent for OSCC, offering antitumor efficacy and immunomodulatory benefits.


Subject(s)
Apoptosis , Biphenyl Compounds , Carcinoma, Squamous Cell , Lignans , Mouth Neoplasms , Lignans/pharmacology , Lignans/therapeutic use , Animals , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Mice , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Apoptosis/drug effects , Cell Line, Tumor , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Humans , Disease Models, Animal , Xenograft Model Antitumor Assays , Tumor Burden/drug effects , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunomodulation/drug effects
19.
Front Immunol ; 15: 1425816, 2024.
Article in English | MEDLINE | ID: mdl-39188713

ABSTRACT

The ketogenic diet (KD) is marked by a substantial decrease in carbohydrate intake and an elevated consumption of fats and proteins, leading to a metabolic state referred to as "ketosis," where fats become the primary source of energy. Recent research has underscored the potential advantages of the KD in mitigating the risk of various illnesses, including type 2 diabetes, hyperlipidemia, heart disease, and cancer. The macronutrient distribution in the KD typically entails high lipid intake, moderate protein consumption, and low carbohydrate intake. Restricting carbohydrates to below 50 g/day induces a catabolic state, prompting metabolic alterations such as gluconeogenesis and ketogenesis. Ketogenesis diminishes fat and glucose accumulation as energy reserves, stimulating the production of fatty acids. Neurodegenerative diseases, encompassing Alzheimer's disease, Parkinson's disease are hallmarked by persistent neuroinflammation. Evolving evidence indicates that immune activation and neuroinflammation play a significant role in the pathogenesis of these diseases. The protective effects of the KD are linked to the generation of ketone bodies (KB), which play a pivotal role in this dietary protocol. Considering these findings, this narrative review seeks to delve into the potential effects of the KD in neuroinflammation by modulating the immune response. Grasping the immunomodulatory effects of the KD on the central nervous system could offer valuable insights into innovative therapeutic approaches for these incapacitating conditions.


Subject(s)
Diet, Ketogenic , Neuroinflammatory Diseases , Humans , Animals , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/diet therapy , Neuroinflammatory Diseases/metabolism , Ketone Bodies/metabolism , Immunomodulation
20.
Med Oncol ; 41(9): 231, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39162936

ABSTRACT

Colorectal cancer (CRC), the third most common cancer worldwide, develops mainly due to the accumulation of genetic and epigenetic changes over many years. Substantial evidence suggests that gut microbiota plays a significant role in the initiation, progression, and control of CRC, depending on the balance between beneficial and pathogenic microorganisms. Nonetheless, gut microbiota composition by regulating the host immune response may either promote or inhibit CRC. Thus, modification of gut microbiota potentially impacts clinical outcomes of immunotherapy. Previous studies have indicated that therapeutic strategies such as probiotics, prebiotics, and postbiotics enhance the intestinal immune system and improve the efficacy of immunotherapeutic agents, potentially serving as a complementary strategy in cancer immunotherapy. This review discusses the role of the gut microbiota in the onset and development of CRC in relation to the immune response. Additionally, we focus on the effect of strategies manipulating gut microbiome on the immune response and efficacy of immunotherapy against CRC. We demonstrate that manipulation of gut microbiome can enhance immune response and outcomes of immunotherapy through downregulating Treg cells and other immunosuppressive cells while improving the function of T cells within the tumor; however, further research, especially clinical trials, are needed to evaluate its efficacy in cancer treatment.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Immunomodulation , Immunotherapy , Probiotics , Humans , Colorectal Neoplasms/immunology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/therapy , Gastrointestinal Microbiome/immunology , Probiotics/therapeutic use , Immunomodulation/immunology , Immunotherapy/methods , Prebiotics/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL