Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell Metab ; 36(6): 1302-1319.e12, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38838642

ABSTRACT

Glucagon-like peptide-1 receptor (GLP-1R) is a key regulator of glucose metabolism known to be expressed by pancreatic ß cells. We herein investigated the role of GLP-1R on T lymphocytes during immune response. Our data showed that a subset of T lymphocytes expresses GLP-1R, which is upregulated during alloimmune response, similarly to PD-1. When mice received islet or cardiac allotransplantation, an expansion of GLP-1Rpos T cells occurred in the spleen and was found to infiltrate the graft. Additional single-cell RNA sequencing (scRNA-seq) analysis conducted on GLP-1Rpos and GLP-1Rneg CD3+ T cells unveiled the existence of molecular and functional dissimilarities between both subpopulations, as the GLP-1Rpos are mainly composed of exhausted CD8 T cells. GLP-1R acts as a T cell-negative costimulatory molecule, and GLP-1R signaling prolongs allograft survival, mitigates alloimmune response, and reduces T lymphocyte graft infiltration. Notably, GLP-1R antagonism triggered anti-tumor immunity when tested in a preclinical mouse model of colorectal cancer.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Islets of Langerhans Transplantation , Mice, Inbred C57BL , Animals , Glucagon-Like Peptide-1 Receptor/metabolism , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Male , Heart Transplantation , Mice, Inbred BALB C , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Graft Survival/immunology
2.
Pharmacol Res ; 190: 106710, 2023 04.
Article in English | MEDLINE | ID: mdl-36871895

ABSTRACT

Diabetic kidney disease (DKD) is the first cause of end-stage kidney disease in patients with diabetes and its prevalence is increasing worldwide. It encompasses histological alterations that mainly affect the glomerular filtration unit, which include thickening of the basement membrane, mesangial cell proliferation, endothelial alteration, and podocyte injury. These morphological abnormalities further result in a persistent increase of urinary albumin-to-creatinine ratio and in a reduction of the estimated glomerular filtration rate. Several molecular and cellular mechanisms have been recognized, up to date, as major players in mediating such clinical and histological features and many more are being under investigation. This review summarizes the most recent advances in understanding cell death mechanisms, intracellular signaling pathways and molecular effectors that play a role in the onset and progression of diabetic kidney damage. Some of those molecular and cellular mechanisms have been already successfully targeted in preclinical models of DKD and, in some cases, strategies have been tested in clinical trials. Finally, this report sheds light on the relevance of novel pathways that may become therapeutic targets for future applications in DKD.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Podocytes , Humans , Diabetic Nephropathies/metabolism , Podocytes/pathology , Signal Transduction , Glomerular Filtration Rate , Diabetes Mellitus/metabolism
3.
Pharmacol Res ; 190: 106709, 2023 04.
Article in English | MEDLINE | ID: mdl-36842542

ABSTRACT

PURPOSE OF REVIEW: The purine nucleotide adenosine triphosphate (ATP) is released into extracellular spaces as extracellular ATP (eATP) as a consequence of cell injury or death and activates the purinergic receptors. Once released, eATP may facilitate T-lymphocyte activation and differentiation. The purpose of this review is to elucidate the role of ATP-mediated signaling in the immunological events related to type 1 diabetes (T1D). RECENT FINDINGS: T lymphocytes mediate immune response during the onset of T1D and promote pancreatic islet or whole pancreas rejection in transplantation. Recent data suggest a potential role for eATP in early steps of T1D onset and of allograft rejection. In different preclinical experimental models and clinical trials, several drugs targeting purinergic signaling have been employed to abrogate lymphocyte activation and differentiation, thus representing an achievable treatment to prevent/revert T1D or to induce long-term islet allograft function. SUMMARY: In preclinical and clinical settings, eATP-signaling inhibition induces immune tolerance in autoimmune disease and in allotransplantation. In this view, the purinergic system may represent a novel therapeutic target for auto- and allo-immunity.


Subject(s)
Autoimmune Diseases , Diabetes Mellitus, Type 1 , Humans , Diabetes Mellitus, Type 1/drug therapy , Autoimmune Diseases/drug therapy , Transplantation, Homologous , T-Lymphocytes/metabolism , Adenosine Triphosphate/metabolism
4.
Pharmacol Res ; 182: 106320, 2022 08.
Article in English | MEDLINE | ID: mdl-35738455

ABSTRACT

In the last few years, a great interest has emerged in investigating the pleiotropic effects of Glucagon Like Peptide-1 Receptor Agonists (GLP-1RAs). While GLP-1RAs ability to lower plasma glucose and to induce weight loss has allowed them to be approved for the treatment of diabetes and obesity, consistent evidences from in vitro studies and preclinical models suggested that GLP-1RAs have anti-inflammatory properties and that may modulate the immune-system. Notably, such anti-inflammatory effects target different pathways in different tissues, underling the broad spectrum of GLP-1RAs actions. This review examines some of the currently proposed molecular mechanisms of GLP-1RAs actions and explores their potential benefits in reducing inflammatory responses, which may well suggest a future therapeutic use of GLP-1RAs in new indications.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Like Peptide-1 Receptor , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Hypoglycemic Agents/therapeutic use , Liraglutide/pharmacology , Obesity/drug therapy
5.
Nat Commun ; 13(1): 684, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115561

ABSTRACT

Loss of pancreatic beta cells is a central feature of type 1 (T1D) and type 2 (T2D) diabetes, but a therapeutic strategy to preserve beta cell mass remains to be established. Here we show that the death receptor TMEM219 is expressed on pancreatic beta cells and that signaling through its ligand insulin-like growth factor binding protein 3 (IGFBP3) leads to beta cell loss and dysfunction. Increased peripheral IGFBP3 was observed in established and at-risk T1D/T2D patients and was confirmed in T1D/T2D preclinical models, suggesting that dysfunctional IGFBP3/TMEM219 signaling is associated with abnormalities in beta cells homeostasis. In vitro and in vivo short-term IGFBP3/TMEM219 inhibition and TMEM219 genetic ablation preserved beta cells and prevented/delayed diabetes onset, while long-term IGFBP3/TMEM219 blockade allowed for beta cell expansion. Interestingly, in several patients' cohorts restoration of appropriate IGFBP3 levels was associated with improved beta cell function. The IGFBP3/TMEM219 pathway is thus shown to be a physiological regulator of beta cell homeostasis and is also demonstrated to be disrupted in T1D/T2D. IGFBP3/TMEM219 targeting may therefore serve as a therapeutic option in diabetes.


Subject(s)
Gene Expression Regulation , Homeostasis/genetics , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Secreting Cells/metabolism , Membrane Proteins/genetics , Signal Transduction/genetics , Adult , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Humans , Immunoblotting , Insulin-Like Growth Factor Binding Protein 3/metabolism , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
6.
JCI Insight ; 6(24)2021 12 22.
Article in English | MEDLINE | ID: mdl-34784300

ABSTRACT

A substantial proportion of patients who have recovered from coronavirus disease-2019 (COVID-19) experience COVID-19-related symptoms even months after hospital discharge. We extensively immunologically characterized patients who recovered from COVID-19. In these patients, T cells were exhausted, with increased PD-1+ T cells, as compared with healthy controls. Plasma levels of IL-1ß, IL-1RA, and IL-8, among others, were also increased in patients who recovered from COVID-19. This altered immunophenotype was mirrored by a reduced ex vivo T cell response to both nonspecific and specific stimulation, revealing a dysfunctional status of T cells, including a poor response to SARS-CoV-2 antigens. Altered levels of plasma soluble PD-L1, as well as of PD1 promoter methylation and PD1-targeting miR-15-5p, in CD8+ T cells were also observed, suggesting abnormal function of the PD-1/PD-L1 immune checkpoint axis. Notably, ex vivo blockade of PD-1 nearly normalized the aforementioned immunophenotype and restored T cell function, reverting the observed post-COVID-19 immune abnormalities; indeed, we also noted an increased T cell-mediated response to SARS-CoV-2 peptides. Finally, in a neutralization assay, PD-1 blockade did not alter the ability of T cells to neutralize SARS-CoV-2 spike pseudotyped lentivirus infection. Immune checkpoint blockade ameliorates post-COVID-19 immune abnormalities and stimulates an anti-SARS-CoV-2 immune response.


Subject(s)
COVID-19/complications , Cytokines/immunology , Immune Checkpoint Inhibitors/pharmacology , Programmed Cell Death 1 Receptor/immunology , SARS-CoV-2/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , Case-Control Studies , Cytokines/drug effects , DNA Methylation , Female , Humans , Immunophenotyping , In Vitro Techniques , Interleukin 1 Receptor Antagonist Protein/drug effects , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-1beta/drug effects , Interleukin-1beta/immunology , Interleukin-8/drug effects , Interleukin-8/immunology , Male , MicroRNAs/metabolism , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Promoter Regions, Genetic , Post-Acute COVID-19 Syndrome
7.
Metabolism ; 121: 154804, 2021 08.
Article in English | MEDLINE | ID: mdl-34097917

ABSTRACT

AIMS/HYPOTHESIS: Inflammation has a major role in diabetic kidney disease. We thus investigated the role of the IL-8-CXCR1/2 axis in favoring kidney damage in diabetes. METHODS: Urinary IL-8 levels were measured in 1247 patients of the Joslin Kidney Study in type 2 diabetes (T2D). The expression of IL-8 and of its membrane receptors CXCR1/CXCR2 was quantified in kidney tissues in patients with T2D and in controls. The effect of CXCR1/2 blockade on diabetic kidney disease was evaluated in db/db mice. RESULTS: IL-8 urinary levels were increased in patients with T2D and diabetic kidney disease, with the highest urinary IL-8 levels found in the patients with the largest decline in glomerular filtration rate, with an increased albumin/creatine ratio and the worst renal outcome. Moreover, glomerular IL-8 renal expression was increased in patients with T2D, as compared to controls. High glucose elicits abundant IL-8 secretion in cultured human immortalized podocytes in vitro. Finally, in diabetic db/db mice and in podocytes in vitro, CXCR1/2 blockade mitigated albuminuria, reduced mesangial expansion, decreased podocyte apoptosis and reduced DNA damage. CONCLUSIONS/INTERPRETATION: The IL-8- CXCR1/2 axis may have a role in diabetic kidney disease by inducing podocyte damage. Indeed, targeting the IL-8-CXCR1/2 axis may reduce the burden of diabetic kidney disease.


Subject(s)
Diabetic Nephropathies/genetics , Interleukin-8/physiology , Receptors, CXCR/physiology , Adult , Animals , Case-Control Studies , Cells, Cultured , Cohort Studies , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Italy , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Podocytes/metabolism , Podocytes/pathology , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Signal Transduction/physiology
8.
Am J Transplant ; 21(10): 3280-3295, 2021 10.
Article in English | MEDLINE | ID: mdl-33764625

ABSTRACT

Despite much progress in improving graft outcome during cardiac transplantation, chronic allograft vasculopathy (CAV) remains an impediment to long-term graft survival. MicroRNAs (miRNAs) emerged as regulators of the immune response. Here, we aimed to examine the miRNA network involved in CAV. miRNA profiling of heart samples obtained from a murine model of CAV and from cardiac-transplanted patients with CAV demonstrated that miR-21 was most significantly expressed and was primarily localized to macrophages. Interestingly, macrophage depletion with clodronate did not significantly prolong allograft survival in mice, while conditional deletion of miR-21 in macrophages or the use of a specific miR-21 antagomir resulted in indefinite cardiac allograft survival and abrogated CAV. The immunophenotype, secretome, ability to phagocytose, migration, and antigen presentation of macrophages were unaffected by miR-21 targeting, while macrophage metabolism was reprogrammed, with a shift toward oxidative phosphorylation in naïve macrophages and with an inhibition of glycolysis in pro-inflammatory macrophages. The aforementioned effects resulted in an increase in M2-like macrophages, which could be reverted by the addition of L-arginine. RNA-seq analysis confirmed alterations in arginase-associated pathways associated with miR-21 antagonism. In conclusion, miR-21 is overexpressed in murine and human CAV, and its targeting delays CAV onset by reprogramming macrophages metabolism.


Subject(s)
Heart Transplantation , MicroRNAs , Allografts , Animals , Graft Rejection/genetics , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Humans , Macrophages , Mice , MicroRNAs/genetics
9.
Article in English | MEDLINE | ID: mdl-33188009

ABSTRACT

INTRODUCTION: Gestational diabetes mellitus (GDM) is the most frequent metabolic complication during pregnancy and is associated with development of short-term and long-term complications for newborns, with large-for-gestational-age (LGA) being particularly common. Interestingly, the mechanism behind altered fetal growth in GDM is only partially understood. RESEARCH DESIGN AND METHODS: A proteomic approach was used to analyze placental samples obtained from healthy pregnant women (n=5), patients with GDM (n=12) and with GDM and LGA (n=5). Effects of altered proteins on fetal development were tested in vitro in human embryonic stem cells (hESCs). RESULTS: Here, we demonstrate that the placental proteome is altered in pregnant women affected by GDM with LGA, with at least 37 proteins differentially expressed to a higher degree (p<0.05) as compared with those with GDM but without LGA. Among these proteins, 10 are involved in regulating tissue differentiation and/or fetal growth and development, with bone marrow proteoglycan (PRG2) and dipeptidyl peptidase-4 (DPP-4) being highly expressed. Both PRG2 and DPP-4 altered the transcriptome profile of stem cells differentiation markers when tested in vitro in hESCs, suggesting a potential role in the onset of fetal abnormalities. CONCLUSIONS: Our findings suggest that placental dysfunction may be directly responsible for abnormal fetal growth/development during GDM. Once established on a larger population, inhibitors of the pathways involving those altered factors may be tested in conditions such as GDM and LGA, in which therapeutic approaches are still lacking.


Subject(s)
Diabetes, Gestational , Fetal Macrosomia , Proteome , Cesarean Section , Female , Fetal Macrosomia/genetics , Humans , Infant, Newborn , Pregnancy , Proteome/genetics , Proteomics
10.
Acta Diabetol ; 57(7): 883-890, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32124076

ABSTRACT

AIMS/HYPOTHESIS: Impaired wound healing significantly impacts morbidity and mortality in diabetic patients, necessitating the development of novel treatments to improve the wound healing process. We here investigated the topical use of acellular embryonic stem cell extracts (EXTs) in wound healing in diabetic db/db mice. METHODS: Wounds were induced in diabetic db/db mice, which were subsequently treated with EXTs, with 3T3 fibroblast cell line protein extracts (3T3XTs) or with saline as a control. Pathology and mechanistic assays were then performed. RESULTS: The in vivo topical administration of EXTs facilitates wound closure, contraction and re-epithelialization. Moreover, EXTs reduced the number of wound-infiltrating CD45+ inflammatory cells and increased the rate of repair and of angiogenesis as compared to controls. Interestingly, the EXT effect was partly enhanced by the use of a collagen-based biocompatible scaffold. In vivo, topical administration of EXTs increased the percentage of regulatory T cells in the wounded tissue, while in vitro EXT treatment reduced T cell-mediated IFN-γ production. Proteomic screening revealed 82 proteins differentially segregating in EXTs as compared to 3T3 extracts, with APEX1 identified as a key player for the observed immunomodulatory effect of EXTs. CONCLUSIONS: EXTs are endowed with immunoregulatory and anti-inflammatory properties; their use improves wound healing in diabetic preclinical models.


Subject(s)
Cell Extracts/pharmacology , Cell Extracts/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Embryonic Stem Cells/chemistry , Wound Healing/drug effects , 3T3 Cells , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Embryonic Stem Cells/metabolism , Immunity, Innate/drug effects , Male , Mice , Mice, Transgenic , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology , Proteome/analysis , Proteome/metabolism , Proteomics , Wound Healing/physiology
11.
Curr Opin Nephrol Hypertens ; 28(1): 47-57, 2019 01.
Article in English | MEDLINE | ID: mdl-30418189

ABSTRACT

PURPOSE OF REVIEW: The leading cause of death in both chronic kidney disease (CKD) and renal transplant patients is cardiovascular events. Post-transplant diabetes mellitus (PTx-DM), which is a major cardiovascular risk factor, is a metabolic disorder that affects 5.5-60.2% of renal allograft recipients by 1-year posttransplant (PTx). PTx-DM has been associated with a negative impact on patient and graft outcomes and survival. RECENT FINDINGS: Individuals who develop PTx-DM are usually prone to this condition prior to and/or after developing CKD. Genetic factors, obesity, inflammation, medications and CKD all are risk factors for PTx-diabetes mellitus. The path to development of disease continues PTx frequently augmented by the use of diabetogenic maintenance immunosuppressive and some nonimmunosuppressive medications. These risk factors are usually associated with an increase in insulin resistance, a decrease in insulin gene expression and/or ß-cell dysfunction and apoptosis. SUMMARY: Some new anti-diabetes mellitus medications may help to improve the overall outcome; however, there is a real need for developing a preventive strategy. Identifying and targeting PTx-DM risk factors may help to guide the development of an effective programme. This could include the adoption of nondiabetogenic immunosuppressive protocols for high-risk patients.


Subject(s)
Diabetes Mellitus/etiology , Kidney Transplantation/adverse effects , Humans , Immunosuppressive Agents/therapeutic use , Insulin Resistance , Renal Insufficiency, Chronic/complications
13.
J Clin Invest ; 128(8): 3490-3503, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30010623

ABSTRACT

Purinergic receptor-7 (P2X7R) signaling controls Th17 and Th1 generation/differentiation, while NOD-like receptor P3 (NLRP3) acts as a Th2 transcriptional factor. Here, we demonstrated the existence of a P2X7R/NLRP3 pathway in T cells that is dysregulated by a P2X7R intracellular region loss-of-function mutation, leading to NLRP3 displacement and to excessive Th17 generation due to abrogation of the NLRP3-mediated Th2 program. This ultimately resulted in poor outcomes in cardiac-transplanted patients carrying the mutant allele, who showed abnormal Th17 generation. Transient NLRP3 silencing in nonmutant T cells or overexpression in mutant T cells normalized the Th profile. Interestingly, IL-17 blockade reduced Th17 skewing of human T cells in vitro and abrogated the severe allograft vasculopathy and abnormal Th17 generation observed in preclinical models in which P2X7R was genetically deleted. This P2X7R intracellular region mutation thus impaired the modulatory effects of P2X7R on NLRP3 expression and function in T cells and led to NLRP3 dysregulation and Th17 skewing, delineating a high-risk group of cardiac-transplanted patients who may benefit from personalized therapy.


Subject(s)
Graft Rejection/immunology , Heart Transplantation , Mutation , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Receptors, Purinergic P2X7/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Allografts , Female , Graft Rejection/genetics , Graft Rejection/pathology , Graft Rejection/therapy , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Precision Medicine , Receptors, Purinergic P2X7/genetics , Th17 Cells/pathology , Th2 Cells/pathology
14.
Rev Recent Clin Trials ; 13(2): 105-113, 2018.
Article in English | MEDLINE | ID: mdl-29173182

ABSTRACT

BACKGROUND: Systemic Lupus Erythematosus (SLE) is an autoimmune disorder characterized by chronic inflammation, which can result in a multitude of systemic or organ-limited manifestations, including the skin, lungs, heart, and kidney. SLE nephritis is present in an average of 38% of patients at the time of diagnosis, and may occur as the initial presentation of disease with progression to End-Stage Renal Disease (ESRD) in roughly 10-20% of patients. METHODS: A review of the current literature was undertaken to investigate the evolution of treatment of SLE nephritis based on randomized trials and robust observational studies. We aimed to provide a timeline of the development of current induction and maintenance therapy, as well as the development of novel targeted therapies, all leading to current guidelines. RESULTS: Based on all available current data on standard of care therapies for SLE nephritis, there is at best a complete remission rate of 50-60%, and roughly 13-25% of patients experience periods of relapse during maintenance therapy for SLE nephritis. Therefore, the need for newer, targeted therapies has been the focus of many current, ongoing clinical trials. CONCLUSION: Standard induction and maintenance therapies at present are anti-proliferative and nonspecific, that is, interfering with the process of autoantigen presentation and activation of autoreactive leukocytes. However, newer agents with specific T-cell, B-cell, or proteasome targets are currently being investigated.


Subject(s)
Lupus Nephritis/diagnosis , Lupus Nephritis/therapy , Humans , Induction Chemotherapy , Kidney Failure, Chronic/etiology , Lupus Nephritis/complications , Maintenance Chemotherapy
15.
J Endocrinol Invest ; 40(8): 803-814, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28260183

ABSTRACT

INTRODUCTION: Although many approaches have been tested to overcome the insulin dependence caused by the pancreatic ß-cells destruction observed in individuals affected by type 1 diabetes (T1D), medical research has largely failed to halt the onset or to reverse T1D. METHODS: In this work, the state of the art of immunotherapy will be examined, and the most important achievement in the field will be critically discussed. Particularly, we will focus on the clinical aspect, thus avoiding the tedious preclinical work done in NOD mice, which has been so poorly translated to the bedside. CONCLUSIONS: Stem cell therapies achieved thus this far the most promising results, while immune ablation and standard immunosuppressants did not maintain the premises of preclinical results. The next step will be to generate a feasible and safe clinical approach in order to cure the thousands of patients affected by T1D.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Immunotherapy , Animals , Humans
17.
Am J Nephrol ; 45(4): 310-319, 2017.
Article in English | MEDLINE | ID: mdl-28237979

ABSTRACT

BACKGROUND: Despite the stable incidence of end-stage renal disease (ESRD), it continues to be associated with an unacceptably high cardiovascular risk. SUMMARY: ESRD is characterized by enhanced oxidative stress and severe inflammation, which boost cardiovascular risk, thus increasing cardiovascular-associated mortality rate. While substantial effort has been made in the technological innovation of dialytic techniques, few significant advances have been made to reduce inflammation in patients with ESRD. Indeed, this contrasts with the extensive scientific breakthroughs made in the basic field of science in targeting inflammation. There is thus a pressing need for clinical trials to test the effect of reducing inflammation in patients with ESRD. Here, we will revisit the negative effect of ESRD on inflammation and explore the impact of enhanced inflammation on cardiovascular outcomes and survival in patients with ESRD. Finally, we will discuss the need for clinical trials that target inflammation in ESRD, as well as weigh potential disadvantages and offer novel innovative approaches. Key Message: We will try to understand why the issue of inflammation has not been successfully addressed thus far in patients with ESRD, while at the same time weighing the potential disadvantages and offering novel innovative approaches for targeting inflammation in patients with ESRD.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cardiovascular Diseases/prevention & control , Complement Inactivating Agents/therapeutic use , Inflammation/therapy , Kidney Failure, Chronic/therapy , Oxidative Stress/drug effects , Antioxidants/therapeutic use , C-Reactive Protein/antagonists & inhibitors , C-Reactive Protein/immunology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/mortality , Clinical Trials as Topic , Complement Inactivating Agents/economics , Complement System Proteins/immunology , Cytokines/antagonists & inhibitors , Cytokines/immunology , Humans , Incidence , Inflammation/epidemiology , Inflammation/immunology , Kidney Failure, Chronic/epidemiology , Kidney Failure, Chronic/immunology , Oxidative Stress/immunology , Renal Dialysis/adverse effects , Risk Factors
18.
PLoS One ; 12(1): e0169077, 2017.
Article in English | MEDLINE | ID: mdl-28052095

ABSTRACT

BACKGROUND: Alteration of certain metabolites may play a role in the pathophysiology of renal allograft disease. METHODS: To explore metabolomic abnormalities in individuals with a failing kidney allograft, we analyzed by liquid chromatography-mass spectrometry (LC-MS/MS; for ex vivo profiling of serum and urine) and two dimensional correlated spectroscopy (2D COSY; for in vivo study of the kidney graft) 40 subjects with varying degrees of chronic allograft dysfunction stratified by tertiles of glomerular filtration rate (GFR; T1, T2, T3). Ten healthy non-allograft individuals were chosen as controls. RESULTS: LC-MS/MS analysis revealed a dose-response association between GFR and serum concentration of tryptophan, glutamine, dimethylarginine isomers (asymmetric [A]DMA and symmetric [S]DMA) and short-chain acylcarnitines (C4 and C12), (test for trend: T1-T3 = p<0.05; p = 0.01; p<0.001; p = 0.01; p = 0.01; p<0.05, respectively). The same association was found between GFR and urinary levels of histidine, DOPA, dopamine, carnosine, SDMA and ADMA (test for trend: T1-T3 = p<0.05; p<0.01; p = 0.001; p<0.05; p = 0.001; p<0.001; p<0.01, respectively). In vivo 2D COSY of the kidney allograft revealed significant reduction in the parenchymal content of choline, creatine, taurine and threonine (all: p<0.05) in individuals with lower GFR levels. CONCLUSIONS: We report an association between renal function and altered metabolomic profile in renal transplant individuals with different degrees of kidney graft function.


Subject(s)
Kidney Transplantation , Metabolomics/methods , Adult , Chromatography, Liquid , Creatinine/urine , Female , Glomerular Filtration Rate/physiology , Humans , Male , Middle Aged , Multivariate Analysis , Tandem Mass Spectrometry
19.
Acta Diabetol ; 52(5): 917-27, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25808641

ABSTRACT

AIMS: Mesenchymal stem cells (MSCs) are multipotent cells with immunomodulatory properties. We tested the ability of MSCs to delay islet allograft rejection. METHODS: Mesenchymal stem cells were generated in vitro from C57BL/6 and BALB/c mice bone marrow, and their immunomodulatory properties were tested in vitro. We then tested the effect of a local or systemic administration of heterologous and autologous MSCs on graft survival in a fully allogeneic model of islet transplantation (BALB/c islets into C57BL/6 mice). RESULTS: In vitro, autologous, but not heterologous, MSCs abrogated immune cell proliferation in response to alloantigens and skewed the immune response toward a Th2 profile. A single dose of autologous MSCs co-transplanted under the kidney capsule with allogeneic islets delayed islet rejection, reduced graft infiltration, and induced long-term graft function in 30 % of recipients. Based on ex vivo analysis of recipient splenocytes, the use of autologous MSCs did not appear to have any systemic effect on the immune response toward graft alloantigens. The systemic injection of autologous MSCs or the local injection of heterologous MSCs failed to delay islet graft rejection. CONCLUSION: Autologous, but not heterologous, MSCs showed multiple immunoregulatory properties in vitro and delayed allograft rejection in vivo when co-transplanted with islets; however, they failed to prevent rejection when injected systemically. Autologous MSCs thus appear to produce a local immunoprivileged site, which promotes graft survival.


Subject(s)
Graft Rejection/prevention & control , Islets of Langerhans Transplantation/methods , Mesenchymal Stem Cell Transplantation/methods , Animals , Bone Marrow Cells/immunology , Bone Marrow Transplantation/methods , Cell Proliferation , Cytokines/metabolism , Graft Rejection/immunology , Graft Survival , Lymphocyte Culture Test, Mixed , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/ultrastructure , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/cytology , Th17 Cells/immunology
20.
Acta Diabetol ; 51(6): 897-904, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24894496

ABSTRACT

Diabetic nephropathy is the leading and possibly the most devastating complication of diabetes, with a prevalence ranging from 25 to 40 % in diabetic individuals, and as such represents an important challenge for public health worldwide. As a major cause of end-stage renal disease, diabetic nephropathy also accounts for a large proportion of deaths in diabetic individuals. To date, therapeutic options for overt diabetic nephropathy include medical interventions to reduce blood glucose levels and to control blood pressure and proteinuria. Recent evidence suggests a strong role for inflammation in the development and progression of diabetic nephropathy. Various immune cells, cytokines and chemokines have been implicated in the onset of diabetic nephropathy, while immune-related transcription factors and adhesion molecules have been correlated with the establishment of a renal proinflammatory microenvironment. Both inflammation and immune activation may promote severe distress in the kidney, with subsequent increased local fibrosis, ultimately leading to the development of end-stage renal disease. Stem cells are undifferentiated cells capable of regenerating virtually any organ or tissue and bearing important immunoregulatory and anti-inflammatory properties. Due to the aforementioned considerations, significant interest has been ignited with regard to the use of stem cells as novel therapeutics for diabetic nephropathy. Here, we will be examining in detail how anti-inflammatory properties of different populations of stem cells may offer novel therapy for the treatment of diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/therapy , Stem Cell Transplantation , Stem Cells/immunology , Transplantation Immunology , Animals , Diabetes Mellitus/immunology , Diabetes Mellitus/therapy , Diabetic Nephropathies/immunology , Humans , Stem Cell Transplantation/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...