Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 264
Filter
1.
Rev Invest Clin ; 76(2): 65-79, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38718804

ABSTRACT

UNASSIGNED: Excess body weight has become a global epidemic and a significant risk factor for developing chronic diseases, which are the leading causes of worldwide morbidities. Adipose tissue (AT), primarily composed of adipocytes, stores substantial amounts of energy and plays a crucial role in maintaining whole-body glucose and lipid metabolism. This helps prevent excessive body fat accumulation and lipotoxicity in peripheral tissues. In addition, AT contains endothelial cells and a substantial population of immune cells (constituting 60-70% of non-adipocyte cells), including macrophages, T and B lymphocytes, and natural killer cells. These resident immune cells engage in crosstalk with adipocytes, contributing to the maintenance of metabolic and immune homeostasis in AT. An exacerbated inflammatory response or inadequate immune resolution can lead to chronic systemic low-grade inflammation, triggering the development of metabolic alterations and the onset of chronic diseases. This review aims to elucidate the regulatory mechanisms through which immune cells influence AT function and energy homeostasis. We also focus on the interactions and functional dynamics of immune cell populations, highlighting their role in maintaining the delicate balance between metabolic health and obesity-related inflammation. Finally, understanding immunometabolism is crucial for unraveling the pathogenesis of metabolic diseases and developing targeted immunotherapeutic strategies. These strategies may offer innovative avenues in the rapidly evolving field of immunometabolism. (Rev Invest Clin. 2024;76(2):65-79).


Subject(s)
Adipose Tissue , Inflammation , Metabolic Diseases , Obesity , Humans , Adipose Tissue/metabolism , Adipose Tissue/immunology , Obesity/immunology , Obesity/metabolism , Inflammation/immunology , Inflammation/metabolism , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Metabolic Diseases/etiology , Energy Metabolism/physiology , Adipocytes/metabolism , Adipocytes/immunology , Lipid Metabolism/physiology , Animals , Homeostasis
2.
J Cell Physiol ; 239(5): e31229, 2024 May.
Article in English | MEDLINE | ID: mdl-38426269

ABSTRACT

RNA-binding proteins (RBPs) play a crucial role in the regulation of posttranscriptional RNA networks, which can undergo dysregulation in many pathological conditions. Human antigen R (HuR) is a highly researched RBP that plays a crucial role as a posttranscriptional regulator. HuR plays a crucial role in the amplification of inflammatory signals by stabilizing the messenger RNA of diverse inflammatory mediators and key molecular players. The noteworthy correlations between HuR and its target molecules, coupled with the remarkable impacts reported on the pathogenesis and advancement of multiple diseases, position HuR as a promising candidate for therapeutic intervention in diverse inflammatory conditions. This review article examines the significance of HuR as a member of the RBP family, its regulatory mechanisms, and its implications in the pathophysiology of inflammation and cardiometabolic illnesses. Our objective is to illuminate potential directions for future research and drug development by conducting a comprehensive analysis of the existing body of research on HuR.


Subject(s)
Cardiovascular Diseases , ELAV-Like Protein 1 , Inflammation , Humans , ELAV-Like Protein 1/metabolism , ELAV-Like Protein 1/genetics , Inflammation/genetics , Inflammation/pathology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Animals , Gene Expression Regulation , Metabolic Diseases/genetics , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Signal Transduction , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
3.
Liver Transpl ; 30(6): 647-658, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38315054

ABSTRACT

This review discusses long-term complications from immunosuppressants after liver transplantation and the management of these complications. Common complications of calcineurin inhibitors include nephrotoxicity and metabolic diseases. Nephrotoxicity can be managed by targeting a lower drug level and/or adding an immunosuppressant of a different class. Metabolic disorders can be managed by treating the underlying condition and targeting a lower drug level. Gastrointestinal adverse effects and myelosuppression are common complications of antimetabolites that are initially managed with dose reduction or discontinuation if adverse events persist. Mammalian targets of rapamycin inhibitors are associated with myelosuppression, proteinuria, impaired wound healing, and stomatitis, which may require dose reduction or discontinuation. Induction agents and agents used for steroid-refractory rejection or antibody-mediated rejection are reviewed. Other rare complications of immunosuppressants are discussed as well.


Subject(s)
Graft Rejection , Immunosuppressive Agents , Liver Transplantation , Humans , Immunosuppressive Agents/adverse effects , Liver Transplantation/adverse effects , Graft Rejection/immunology , Graft Rejection/prevention & control , Calcineurin Inhibitors/adverse effects , Kidney Diseases/chemically induced , Kidney Diseases/immunology , Immunosuppression Therapy/adverse effects , Immunosuppression Therapy/methods , Metabolic Diseases/chemically induced , Metabolic Diseases/immunology , Metabolic Diseases/therapy , MTOR Inhibitors/adverse effects
4.
J Physiol Biochem ; 80(2): 249-260, 2024 May.
Article in English | MEDLINE | ID: mdl-38158555

ABSTRACT

N6-methyladenosine (m6A) is one of the most abundant epitranscriptomic modifications on eukaryotic mRNA. Evidence has highlighted that m6A is altered in response to inflammation-related factors and it is closely associated with various inflammation-related diseases. Multiple subpopulations of myeloid cells, such as macrophages, dendritic cells, and granulocytes, are crucial for the regulating of immune process in inflammation-related diseases. Recent studies have revealed that m6A plays an important regulatory role in the functional of multiple myeloid cells. In this review, we comprehensively summarize the function of m6A modification in myeloid cells from the perspective of myeloid cell production, activation, polarization, and migration. Furthermore, we discuss how m6A-mediated myeloid cell function affects the progression of inflammation-related diseases, including autoimmune diseases, chronic metabolic diseases, and malignant tumors. Finally, we discuss the challenges encountered in the study of m6A in myeloid cells, intended to provide a new direction for the study of the pathogenesis of inflammation-related diseases.


Subject(s)
Adenosine , Adenosine/analogs & derivatives , Inflammation , Myeloid Cells , Adenosine/metabolism , Humans , Inflammation/metabolism , Myeloid Cells/metabolism , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/immunology , Neoplasms/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/immunology , Metabolic Diseases/pathology
5.
Front Immunol ; 14: 1153915, 2023.
Article in English | MEDLINE | ID: mdl-37153549

ABSTRACT

Macrophage infiltration into adipose tissue is a key pathological factor inducing adipose tissue dysfunction and contributing to obesity-induced inflammation and metabolic disorders. In this review, we aim to present the most recent research on macrophage heterogeneity in adipose tissue, with a focus on the molecular targets applied to macrophages as potential therapeutics for metabolic diseases. We begin by discussing the recruitment of macrophages and their roles in adipose tissue. While resident adipose tissue macrophages display an anti-inflammatory phenotype and promote the development of metabolically favorable beige adipose tissue, an increase in pro-inflammatory macrophages in adipose tissue has negative effects on adipose tissue function, including inhibition of adipogenesis, promotion of inflammation, insulin resistance, and fibrosis. Then, we presented the identities of the newly discovered adipose tissue macrophage subtypes (e.g. metabolically activated macrophages, CD9+ macrophages, lipid-associated macrophages, DARC+ macrophages, and MFehi macrophages), the majority of which are located in crown-like structures within adipose tissue during obesity. Finally, we discussed macrophage-targeting strategies to ameliorate obesity-related inflammation and metabolic abnormalities, with a focus on transcriptional factors such as PPARγ, KLF4, NFATc3, and HoxA5, which promote macrophage anti-inflammatory M2 polarization, as well as TLR4/NF-κB-mediated inflammatory pathways that activate pro-inflammatory M1 macrophages. In addition, a number of intracellular metabolic pathways closely associated with glucose metabolism, oxidative stress, nutrient sensing, and circadian clock regulation were examined. Understanding the complexities of macrophage plasticity and functionality may open up new avenues for the development of macrophage-based treatments for obesity and other metabolic diseases.


Subject(s)
Adipose Tissue , Macrophages , Metabolic Diseases , Obesity , Adipose Tissue/immunology , Macrophages/classification , Macrophages/immunology , Obesity/immunology , Obesity/therapy , Metabolic Diseases/immunology , Metabolic Diseases/therapy , Humans , Inflammation/immunology , Inflammation/therapy , Adipogenesis/immunology , Cell Polarity
6.
J Atheroscler Thromb ; 29(3): 297-307, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34248111

ABSTRACT

Sterile chronic inflammation causes cardiometabolic disorders; however, the mechanisms are not fully understood. Previous studies have demonstrated the degradation of cells/tissues in the vasculature and metabolic organs in lifestyle-associated diseases, such as diabetes and hyperlipidemia, suggesting the release and/or accumulation of nucleic acids from damaged cells. DNA is indispensable for life; however, DNA fragments, especially those from pathogens, strongly induce inflammation by the activation of DNA sensors. Growing evidence suggests that DNA-sensing mechanisms, which are normally involved in self-defense against pathogens as the innate immune system, are associated with the progression of inflammatory diseases in response to endogenous DNA fragments. There are several types of DNA sensors in our bodies. Toll-like receptor 9 (TLR9)-one of the most studied DNA sensors-recognizes DNA fragments in endosome. In addition, stimulator of interferon genes (STING), which has recently been extensively investigated, recognizes cyclic GMP-AMP (cGAMP) generated from DNA fragments in the cytosol. Both TLR9 and STING are known to play pivotal roles in host defense as the innate immune system. However, recent studies have indicated that the activation of these DNA sensors in immune cells, such as macrophages, promotes inflammation leading to the development of vascular and metabolic diseases associated with lifestyle. In this review, we discuss recent advances in determining the roles of DNA sensors in these disease contexts. Revealing a novel mechanism of sterile chronic inflammation regulated by DNA sensors might facilitate clinical interventions for these health conditions.


Subject(s)
DNA/immunology , Immunity, Innate , Metabolic Diseases/immunology , Vascular Diseases/immunology , Animals , DNA/metabolism , Humans , Metabolic Diseases/metabolism , Signal Transduction , Vascular Diseases/metabolism
7.
J Leukoc Biol ; 111(2): 451-467, 2022 02.
Article in English | MEDLINE | ID: mdl-33884656

ABSTRACT

Periodontitis is a common chronic inflammatory disease that can result in tooth loss and poses a risk to systemic health. Lymphocytes play important roles in periodontitis through multiple mechanisms. Regulatory lymphocytes including regulatory B cells (Bregs) and T cells (Tregs) are the main immunosuppressive cells that maintain immune homeostasis, and are critical to our understanding of the pathogenesis of periodontitis and the development of effective treatments. In this review, we discuss the phenotypes, roles, and modulating strategies of regulatory lymphocytes including Bregs and Tregs in periodontitis and frequently cooccurring inflammatory diseases such as rheumatoid arthritis, Alzheimer disease, diabetes mellitus, and stroke. The current evidence suggests that restoring immune balance through therapeutic targeting of regulatory lymphocytes is a promising strategy for the treatment of periodontitis and other systemic inflammatory diseases.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Metabolic Diseases/pathology , Periodontitis/pathology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Metabolic Diseases/immunology , Periodontitis/immunology , Phenotype
8.
Front Immunol ; 12: 732913, 2021.
Article in English | MEDLINE | ID: mdl-34737743

ABSTRACT

Obesity prevails worldwide to an increasing effect. For example, up to 42% of American adults are considered obese. Obese individuals are prone to a variety of complications of metabolic disorders including diabetes mellitus, hypertension, cardiovascular disease, and chronic kidney disease. Recent meta-analyses of clinical studies in patient cohorts in the ongoing coronavirus-disease 2019 (COVID-19) pandemic indicate that the presence of obesity and relevant disorders is linked to a more severe prognosis of COVID-19. Given the significance of obesity in COVID-19 progression, we provide a review of host metabolic and immune responses in the immunometabolic dysregulation exaggerated by obesity and the viral infection that develops into a severe course of COVID-19. Moreover, sequela studies of individuals 6 months after having COVID-19 show a higher risk of metabolic comorbidities including obesity, diabetes, and kidney disease. These collectively implicate an inter-systemic dimension to understanding the association between obesity and COVID-19 and suggest an interdisciplinary intervention for relief of obesity-COVID-19 complications beyond the phase of acute infection.


Subject(s)
COVID-19/immunology , COVID-19/metabolism , Obesity/immunology , Obesity/metabolism , COVID-19/complications , Disease Progression , Host-Pathogen Interactions/immunology , Humans , Immunity , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Obesity/complications , Prognosis , SARS-CoV-2/pathogenicity , Severity of Illness Index
9.
Front Immunol ; 12: 762564, 2021.
Article in English | MEDLINE | ID: mdl-34675940

ABSTRACT

Accumulating evidences support that amino acids direct the fate decision of immune cells. Glycine is a simple structural amino acid acting as an inhibitory neurotransmitter. Besides, glycine receptors as well as glycine transporters are found in macrophages, indicating that glycine alters the functions of macrophages besides as an inhibitory neurotransmitter. Mechanistically, glycine shapes macrophage polarization via cellular signaling pathways (e.g., NF-κB, NRF2, and Akt) and microRNAs. Moreover, glycine has beneficial effects in preventing and/or treating macrophage-associated diseases such as colitis, NAFLD and ischemia-reperfusion injury. Collectively, this review highlights the conceivable role of glycinergic signaling for macrophage polarization and indicates the potential application of glycine supplementation as an adjuvant therapy in macrophage-associated diseases.


Subject(s)
Glycine/immunology , Macrophages/immunology , Animals , Colitis/immunology , Glycine/metabolism , Humans , Metabolic Diseases/immunology , MicroRNAs , Neoplasms/immunology , Reperfusion Injury/immunology , Signal Transduction
10.
Eur J Endocrinol ; 184(6): 857-865, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34552304

ABSTRACT

OBJECTIVE: Obese and overweight body mass index (BMI) categories have been associated with increased immune-related adverse events (irAEs) in patients with cancer receiving immune checkpoint inhibitors (ICIs); however, the impact of being overweight in conjunction with related metabolic syndrome-associated factors on irAEs have not been investigated. We aimed to evaluate the impact of overweight and obese BMI according to metabolic disease burden on the development of irAEs. DESIGN AND METHODS: We conducted a retrospective observational study of patients receiving ICIs at a cancer center. Our main study outcome was development of ≥grade 2 (moderate) irAEs. Our main predictor was weight/metabolic disease risk category: (1) normal weight (BMI 18.5-24.9 kg/m2)/low metabolic risk (<2 metabolic diseases [diabetes, dyslipidemia, hypertension]), (2) normal weight/high metabolic risk (≥2 metabolic diseases), (3) overweight (BMI ≥25 kg/m2)/low metabolic risk, and (4) overweight/high metabolic risk. RESULTS: Of 411 patients in our cohort, 374 were eligible for analysis. Overall, 111 (30%) patients developed ≥grade 2 irAEs. In Cox analysis, overweight/low metabolic risk was significantly associated with ≥grade 2 irAEs (hazard ratio [HR]: 2.0, 95% confidence interval [95% CI]: 1.2-3.4) when compared to normal weight/low metabolic risk, while overweight/high metabolic risk (HR: 1.3, 95% CI: 0.7-2.2) and normal weight/high metabolic risk (HR: 1.5, 95% CI: 0.7-3.0) were not. CONCLUSIONS: Overweight patients with fewer metabolic comorbidities were at increased risk for irAEs. This study provides an important insight that BMI should be evaluated in the context of associated metabolic comorbidities in assessing risk of irAE development and ICI immune response.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/epidemiology , Immune Checkpoint Inhibitors/adverse effects , Metabolic Diseases/epidemiology , Neoplasms/drug therapy , Neoplasms/epidemiology , Adult , Aged , Aged, 80 and over , Body Mass Index , Cohort Studies , Drug-Related Side Effects and Adverse Reactions/immunology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Follow-Up Studies , Humans , Immune Checkpoint Inhibitors/administration & dosage , Male , Metabolic Diseases/complications , Metabolic Diseases/immunology , Metabolic Syndrome/complications , Metabolic Syndrome/epidemiology , Metabolic Syndrome/immunology , Middle Aged , Neoplasms/complications , Neoplasms/immunology , Obesity/complications , Obesity/epidemiology , Obesity/immunology , Overweight/complications , Overweight/epidemiology , Overweight/immunology , Retrospective Studies , Risk Assessment , Severity of Illness Index , Young Adult
11.
J Immunol ; 207(8): 1959-1963, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34544802

ABSTRACT

Previous studies indicate that IL-17A plays an important role in mediating the intestinal microbiota and systemic metabolic functions. However, it is not known where IL-17RA signaling occurs to mediate these effects. To investigate this question, we used intestinal epithelial-specific (Il17ra ΔIEC ) and liver-specific (Il17raΔLiver ) IL-17RA knockout mice as well as littermate control mice. Our results indicate that intestinal IL-17RA signaling helps mediate systemic metabolic functions upon exposure to prolonged high-fat diet. Il17ra ΔIEC mice display impaired glucose metabolism, altered hormone and adipokine levels, increased visceral adiposity, and greater hepatic lipid deposition when compared with their littermate controls. We show that IL-17RA-driven changes in microbiota composition are responsible for regulating systemic glucose metabolism. Altogether, our data elucidate the importance of intestinal IL-17RA signaling in regulating high-fat diet-mediated systemic glucose and lipid metabolism.


Subject(s)
Interleukin-17/metabolism , Intestinal Mucosa/physiology , Liver/physiology , Metabolic Diseases/immunology , Microbiota/immunology , Receptors, Interleukin-17/metabolism , Adipokines/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , Glucose/metabolism , Hormones/metabolism , Humans , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
12.
J Immunol ; 207(7): 1719-1724, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34544814

ABSTRACT

Metabolic diseases are common worldwide and include diseases of overnutrition, such as obesity, or undernutrition, such as kwashiorkor. Both the immune system and the microbiota contribute to a variety of metabolic diseases; however, these two processes have largely been studied independently of one another in this context. The gastrointestinal system houses the greatest density of microbes but also houses one of the largest collections of immune molecules, especially Abs. The IgA isotype dominates the Ab landscape at mucosal sites, and a number of studies have demonstrated the importance of this Ab to the stability of the microbiota. In this article, we review the literature that demonstrates how homeostatic Ab responses control microbiota composition and function to influence metabolic disease. We propose that many metabolic diseases may arise from disruptions to homeostatic immune control of gut commensals and that further understanding this interaction can offer a novel opportunity for therapeutic interventions.


Subject(s)
Dysbiosis/immunology , Immunoglobulin A/metabolism , Metabolic Diseases/immunology , Microbiota/immunology , Mucous Membrane/immunology , Animals , Dysbiosis/microbiology , Host Microbial Interactions , Humans , Immunity, Mucosal , Immunomodulation , Metabolic Diseases/microbiology , Mucous Membrane/microbiology
13.
Biomolecules ; 11(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-34356595

ABSTRACT

Infectious and many non-infectious diseases share common molecular mechanisms. Among them, oxidative stress and the subsequent inflammatory reaction are of particular note. Metabolic disorders induced by external agents, be they bacterial or viral pathogens, excessive calorie intake, poor-quality nutrients, or environmental factors produce an imbalance between the production of free radicals and endogenous antioxidant systems; the consequence being the oxidation of lipids, proteins, and nucleic acids. Oxidation and inflammation are closely related, and whether oxidative stress and inflammation represent the causes or consequences of cellular pathology, both produce metabolic alterations that influence the pathogenesis of the disease. In this review, we highlight two key molecules in the regulation of these processes: Paraoxonase-1 (PON1) and chemokine (C-C motif) ligand 2 (CCL2). PON1 is an enzyme bound to high-density lipoproteins. It breaks down lipid peroxides in lipoproteins and cells, participates in the protection conferred by HDL against different infectious agents, and is considered part of the innate immune system. With PON1 deficiency, CCL2 production increases, inducing migration and infiltration of immune cells in target tissues and disturbing normal metabolic function. This disruption involves pathways controlling cellular homeostasis as well as metabolically-driven chronic inflammatory states. Hence, an understanding of these relationships would help improve treatments and, as well, identify new therapeutic targets.


Subject(s)
Aryldialkylphosphatase/metabolism , Chemokine CCL2/metabolism , Metabolic Diseases/metabolism , Aryldialkylphosphatase/physiology , Chemokine CCL2/physiology , Homeostasis , Humans , Inflammation , Ligands , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Metabolic Diseases/immunology , Metabolic Diseases/physiopathology , Oxidation-Reduction , Oxidative Stress
14.
Signal Transduct Target Ther ; 6(1): 247, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34210954

ABSTRACT

Inflammasomes are protein complexes of the innate immune system that initiate inflammation in response to either exogenous pathogens or endogenous danger signals. Inflammasome multiprotein complexes are composed of three parts: a sensor protein, an adaptor, and pro-caspase-1. Activation of the inflammasome leads to the activation of caspase-1, which cleaves pro-inflammatory cytokines such as IL-1ß and IL-18, leading to pyroptosis. Effectors of the inflammasome not only provide protection against infectious pathogens, but also mediate control over sterile insults. Aberrant inflammasome signaling has been implicated in the development of cardiovascular and metabolic diseases, cancer, and neurodegenerative disorders. Here, we review the role of the inflammasome as a double-edged sword in various diseases, and the outcomes can be either good or bad depending on the disease, as well as the genetic background. We highlight inflammasome memory and the two-shot activation process. We also propose the M- and N-type inflammation model, and discuss how the inflammasome pathway may be targeted for the development of novel therapy.


Subject(s)
Cardiovascular Diseases , Inflammasomes/immunology , Metabolic Diseases , Neoplasms , Neurodegenerative Diseases , Cardiovascular Diseases/immunology , Cardiovascular Diseases/therapy , Humans , Interleukin-18/immunology , Interleukin-1beta/immunology , Metabolic Diseases/immunology , Metabolic Diseases/therapy , Neoplasms/immunology , Neoplasms/therapy , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/therapy , Pyroptosis/immunology
15.
J Steroid Biochem Mol Biol ; 213: 105952, 2021 10.
Article in English | MEDLINE | ID: mdl-34274458

ABSTRACT

The impacts of glucocorticoids (GCs) are mainly mediated by a nuclear receptor (GR) existing in almost every tissue. The GR regulates a wide range of physiological functions, including inflammation, cell metabolism, and differentiation playing a major role in cellular responses to GCs and stress. Therefore, the dysregulation or disruption of GR can cause deficiencies in the adaptation to stress and the preservation of homeostasis. The number of GR polymorphisms associated with different diseases has been mounting per year. Tackling these clinical complications obliges a comprehensive understanding of the molecular network action of GCs at the level of the GR structure and its signaling pathways. Beyond genetic variation in the GR gene, epigenetic changes can enhance our understanding of causal factors involved in the development of diseases and identifying biomarkers. In this review, we highlight the relationships of GC receptor gene polymorphisms and epigenetics with different diseases.


Subject(s)
Autoimmune Diseases/genetics , Bone Diseases/genetics , Cardiovascular Diseases/genetics , Epigenesis, Genetic , Mental Disorders/genetics , Metabolic Diseases/genetics , Receptors, Glucocorticoid/genetics , Adaptation, Physiological/genetics , Adaptation, Physiological/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Bone Diseases/immunology , Bone Diseases/pathology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/pathology , DNA Methylation , Glucocorticoids/immunology , Glucocorticoids/metabolism , Homeostasis/genetics , Homeostasis/immunology , Humans , Inflammation , Mental Disorders/immunology , Mental Disorders/pathology , Metabolic Diseases/immunology , Metabolic Diseases/pathology , Polymorphism, Genetic , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/immunology , Signal Transduction , Stress, Physiological/genetics , Stress, Physiological/immunology
16.
Nat Rev Immunol ; 21(10): 669-679, 2021 10.
Article in English | MEDLINE | ID: mdl-34285393

ABSTRACT

Cardiometabolic disorders were originally thought to be driven primarily by changes in lipid metabolism that cause the accumulation of lipids in organs, thereby impairing their function. Thus, in the setting of cardiovascular disease, statins - a class of lipid-lowering drugs - have remained the frontline therapy. In the past 20 years, seminal discoveries have revealed a central role of both the innate and adaptive immune system in driving cardiometabolic disorders. As such, it is now appreciated that immune-based interventions may have an important role in reducing death and disability from cardiometabolic disorders. However, to date, there have been a limited number of clinical trials exploring this interventional strategy. Nonetheless, elegant preclinical research suggests that immune-targeted therapies can have a major impact in treating cardiometabolic disease. Here, we discuss the history and recent advancements in the use of immunotherapies to treat cardiometabolic disorders.


Subject(s)
Cardiovascular Diseases/immunology , Cardiovascular Diseases/therapy , Immunotherapy , Metabolic Diseases/immunology , Metabolic Diseases/therapy , Animals , Humans
17.
Biomolecules ; 11(5)2021 05 11.
Article in English | MEDLINE | ID: mdl-34064822

ABSTRACT

Lipoxygenases (LOXs) are lipid metabolizing enzymes that catalyze the di-oxygenation of polyunsaturated fatty acids to generate active eicosanoid products. 12-lipoxygenases (12-LOXs) primarily oxygenate the 12th carbon of its substrates. Many studies have demonstrated that 12-LOXs and their eicosanoid metabolite 12-hydroxyeicosatetraenoate (12-HETE), have significant pathological implications in inflammatory diseases. Increased level of 12-LOX activity promotes stress (both oxidative and endoplasmic reticulum)-mediated inflammation, leading to damage in these tissues. 12-LOXs are also associated with enhanced cellular migration of immune cells-a characteristic of several metabolic and autoimmune disorders. Genetic depletion or pharmacological inhibition of the enzyme in animal models of various diseases has shown to be protective against disease development and/or progression in animal models in the setting of diabetes, pulmonary, cardiovascular, and metabolic disease, suggesting a translational potential of targeting the enzyme for the treatment of several disorders. In this article, we review the role of 12-LOXs in the pathogenesis of several diseases in which chronic inflammation plays an underlying role.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Inflammation/immunology , Metabolic Diseases/immunology , Animals , Arachidonate 12-Lipoxygenase/genetics , Humans , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Oxidation-Reduction
18.
Mol Cell Biochem ; 476(11): 3935-3950, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34181183

ABSTRACT

Extracellular matrix (ECM) plays an important role in the structural organization of tissue and delivery of external cues to the cell. Biglycan, a class I small leucine-rich proteoglycans (SLRP), is a key component of the ECM that participates in scaffolding the collagen fibrils and mediates cell signaling. Dysregulation of biglycan expression can result in wide range of clinical conditions such as metabolic disorder, inflammatory disorder, musculoskeletal defects and malignancies. In this review, we aim to update our current understanding regarding the link between altered expression of biglycan and different clinicopathological states. Biglycan interacts with toll like receptors (TLR)-2 and TLR-4 on the immune cells which initiates inflammation and aggravates inflammatory disorders. ECM unbound soluble biglycan acts as a DAMP (danger associated molecular pattern) resulting in sterile inflammation. Dysregulation of biglycan expression is also observed in inflammatory metabolic conditions such as atherosclerosis and obesity. In cancer, high-biglycan expression facilitates tumor growth, invasion and metastasis which is associated with poor clinical outcome. As a pivotal structural component of the ECM, biglycan strengthens the musculoskeletal system and its absence is associated with musculoskeletal defects. Thus, SLRP biglycan is a potential marker which is significantly altered in different clinicopathological states.


Subject(s)
Biglycan/metabolism , Inflammation/immunology , Metabolic Diseases/immunology , Neoplasms/metabolism , Small Leucine-Rich Proteoglycans/metabolism , Animals , Biomarkers/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Neoplasms/immunology , Neoplasms/pathology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
19.
Nat Commun ; 12(1): 2598, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972511

ABSTRACT

The intestinal immune system is an important modulator of glucose homeostasis and obesity-associated insulin resistance. Dietary factors, the intestinal microbiota and their metabolites shape intestinal immunity during obesity. The intestinal immune system in turn affects processes such as intestinal permeability, immune cell trafficking, and intestinal hormone availability, impacting systemic insulin resistance. Understanding these pathways might identify mechanisms underlying treatments for insulin resistance, such as metformin and bariatric surgery, or aid in developing new therapies and vaccination approaches. Here, we highlight evolving concepts centered on intestinal immunity, diet, and the microbiota to provide a working model of obesity-related metabolic disease.


Subject(s)
Gastrointestinal Microbiome/immunology , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Obesity/metabolism , Animals , B-Lymphocytes/immunology , Cytokines/metabolism , Diet Therapy , Humans , Immune System/cytology , Immune System/metabolism , Inflammation/immunology , Inflammation/metabolism , Insulin Resistance/immunology , Metabolic Diseases/microbiology , Metabolic Diseases/therapy , Obesity/diet therapy , Obesity/immunology , Obesity/therapy , T-Lymphocytes/immunology
20.
Nutrients ; 13(3)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807621

ABSTRACT

Flavanols are natural occurring polyphenols abundant in fruits and vegetables to which have been attributed to beneficial effects on health, and also against metabolic diseases, such as diabetes, obesity and metabolic syndrome. These positive properties have been associated to the modulation of different molecular pathways, and importantly, to the regulation of immunological reactions (pro-inflammatory cytokines, chemokines, adhesion molecules, nuclear factor-κB [NF-κB], inducible enzymes), and the activity of cells of the immune system. In addition, flavanols can modulate the composition and function of gut microbiome in a prebiotic-like manner, resulting in the positive regulation of metabolic pathways and immune responses, and reduction of low-grade chronic inflammation. Moreover, the biotransformation of flavanols by gut bacteria increases their bioavailability generating a number of metabolites with potential to affect human metabolism, including during metabolic diseases. However, the exact mechanisms by which flavanols act on the microbiota and immune system to influence health and disease remain unclear, especially in humans where these connections have been scarcely explored. This review seeks to summarize recent advances on the complex interaction of flavanols with gut microbiota, immunity and inflammation focus on metabolic diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Flavonoids/pharmacokinetics , Gastrointestinal Microbiome/drug effects , Immune System/drug effects , Metabolic Diseases/therapy , Biological Availability , Diet/methods , Humans , Inflammation , Metabolic Diseases/immunology , Metabolic Diseases/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...