Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
JHEP Rep ; 6(2): 100984, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38293685

ABSTRACT

Background & Aims: Lipid droplet (LD) accumulation in cells and tissues is understood to be an evolutionarily conserved tissue tolerance mechanism to prevent lipotoxicity caused by excess lipids; however, the presence of excess LDs has been associated with numerous diseases. Sepsis triggers the reprogramming of lipid metabolism and LD accumulation in cells and tissues, including the liver. The functions and consequences of sepsis-triggered liver LD accumulation are not well known. Methods: Experimental sepsis was induced by CLP (caecal ligation and puncture) in mice. Markers of hepatic steatosis, liver injury, hepatic oxidative stress, and inflammation were analysed using a combination of functional, imaging, lipidomic, protein expression and immune-enzymatic assays. To prevent LD formation, mice were treated orally with A922500, a pharmacological inhibitor of DGAT1. Results: We identified that liver LD overload correlates with liver injury and sepsis severity. Moreover, the progression of steatosis from 24 h to 48 h post-CLP occurs in parallel with increased cytokine expression, inflammatory cell recruitment and oxidative stress. Lipidomic analysis of purified LDs demonstrated that sepsis leads LDs to harbour increased amounts of unsaturated fatty acids, mostly 18:1 and 18:2. An increased content of lipoperoxides within LDs was also observed. Conversely, the impairment of LD formation by inhibition of the DGAT1 enzyme reduces levels of hepatic inflammation and lipid peroxidation markers and ameliorates sepsis-induced liver injury. Conclusions: Our results indicate that sepsis triggers lipid metabolism alterations that culminate in increased liver LD accumulation. Increased LDs are associated with disease severity and liver injury. Moreover, inhibition of LD accumulation decreased the production of inflammatory mediators and lipid peroxidation while improving tissue function, suggesting that LDs contribute to the pathogenesis of liver injury triggered by sepsis. Impact and Implications: Sepsis is a complex life-threatening syndrome caused by dysregulated inflammatory and metabolic host responses to infection. The observation that lipid droplets may contribute to sepsis-associated organ injury by amplifying lipid peroxidation and inflammation provides a rationale for therapeutically targeting lipid droplets and lipid metabolism in sepsis.

2.
Front Bioeng Biotechnol ; 11: 1203387, 2023.
Article in English | MEDLINE | ID: mdl-37362215

ABSTRACT

Introduction: Influenza A virus (IAV)-induced acute lung injury (ALI) is characterized by pronounced proinflammatory activation and respiratory lung dysfunction. In this study, we performed deep immune profiling on airway and circulating immune cells to examine the effect of immunomodulation and therapeutic outcomes of mesenchymal stem cells (MSCs) therapy in mice with IAV-induced ALI. Methods: Animals were inoculated intranasally with H1N1 IAV, followed by intravenous administration of vehicle, or human clinical-grade, bone marrow-derived MSCs 24-h later, and monitored for six days to evaluate the survival. In another set of animals, bronchoalveolar lavage (BAL) fluid and whole blood were collected three days after infection for flow or mass cytometry (CyTOF) immune profiling analysis. Results: Immune cell population and phenotypic shifts in blood were mapped by CyTOF. Increases were observed in granulocytes and myeloid-derived cells in blood from vehicle-treated animals. While MSC treatment accentuated changes in these populations, naïve B, antibody-secreting B cells, and T cells were decreased in MSC-treated animals at day 3. Compared to sham animals, IAV infection induced a significant 5.5-fold increase in BAL total cell counts, including CD4+ and CD8+ T cells, CD19+ B cells, CD11b + Ly6G + neutrophils, and CD11b + Ly6C + monocytes. MSC treatment significantly decreased BAL total cell counts in IAV-infected mice, specifically the number of infiltrating CD4+ T cells and CD11b + Ly6G + neutrophils. In contrast, there were increases in CD8+ T cells, B cells, and monocytes in the alveolar space in MSC-treated animals. Phenotypic immune cell profiling of blood and BAL revealed a significantly higher proportion of the monocyte population with the M2 phenotype (CD206) in MSC-treated animals; however, this failed to confer protective effects in the survival of infected mice or reduce viral titer in the lung. Further investigation revealed that MSCs were susceptible to IAV infection, leading to increased cell death and potentially affecting their efficacy. Conclusion: These findings provided in vivo evidence that MSCs promote the selective recruitment of immune cells to the site of infection during IAV infection, with reductions in proinflammatory phenotypes. However, MSCs offered no survival benefit in IAV-infected animals, possibly due to MSCs' H1N1 IAV susceptibility and subsequent cell death.

3.
J Neuroinflammation ; 20(1): 61, 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36882750

ABSTRACT

Zika virus (ZIKV) infection is a global public health concern linked to adult neurological disorders and congenital diseases in newborns. Host lipid metabolism, including lipid droplet (LD) biogenesis, has been associated with viral replication and pathogenesis of different viruses. However, the mechanisms of LD formation and their roles in ZIKV infection in neural cells are still unclear. Here, we demonstrate that ZIKV regulates the expression of pathways associated with lipid metabolism, including the upregulation and activation of lipogenesis-associated transcription factors and decreased expression of lipolysis-associated proteins, leading to significant LD accumulation in human neuroblastoma SH-SY5Y cells and in neural stem cells (NSCs). Pharmacological inhibition of DGAT-1 decreased LD accumulation and ZIKV replication in vitro in human cells and in an in vivo mouse model of infection. In accordance with the role of LDs in the regulation of inflammation and innate immunity, we show that blocking LD formation has major roles in inflammatory cytokine production in the brain. Moreover, we observed that inhibition of DGAT-1 inhibited the weight loss and mortality induced by ZIKV infection in vivo. Our results reveal that LD biogenesis triggered by ZIKV infection is a crucial step for ZIKV replication and pathogenesis in neural cells. Therefore, targeting lipid metabolism and LD biogenesis may represent potential strategies for anti-ZIKV treatment development.


Subject(s)
Neuroblastoma , Zika Virus Infection , Zika Virus , Animals , Humans , Mice , Lipid Droplets , Virus Replication
4.
iScience ; 25(5): 104188, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35402859

ABSTRACT

Mesenchymal stem cells (MSCs) are being studied for the treatment of COVID-19-associated critical illness, due to their immunomodulatory properties. Here, we hypothesized that viral mimic-priming improves MSCs' abilities to rebalance the dysregulated immune responses in COVID-19. Transcriptome analysis of poly(I:C)-primed MSCs (pIC-MSCs) showed upregulation of pathways in antiviral and immunomodulatory responses. Together with increased expression of antiviral proteins such as MX1, IFITM3, and OAS1, these changes translated to greater effector functions in regulating monocytes and granulocytes while further enhancing MSCs' ability to block SARS-CoV-2 pseudovirus entry into epithelial cells. Most importantly, the addition of pIC-MSCs to COVID-19 patient whole blood significantly reduced inflammatory neutrophils and increased M2 monocytes while enhancing their phagocytic effector function. We reveal for the first time that MSCs can be primed by Toll-like receptor 3 agonist to improve their ability to rebalance the dysregulated immune responses seen in severe SARS-CoV-2 infection.

5.
Stem Cell Res Ther ; 12(1): 184, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33726829

ABSTRACT

BACKGROUND: Acute lung injury (ALI) and in its severe form, acute respiratory distress syndrome (ARDS), results in increased pulmonary vascular inflammation and permeability and is a major cause of mortality in many critically ill patients. Although cell-based therapies have shown promise in experimental ALI, strategies are needed to enhance the potency of mesenchymal stem cells (MSCs) to develop more effective treatments. Genetic modification of MSCs has been demonstrated to significantly improve the therapeutic benefits of these cells; however, the optimal vector for gene transfer is not clear. Given the acute nature of ARDS, transient transfection is desirable to avoid off-target effects of long-term transgene expression, as well as the potential adverse consequences of genomic integration. METHODS: Here, we explored whether a minicircle DNA (MC) vector containing human angiopoietin 1 (MC-ANGPT1) can provide a more effective platform for gene-enhanced MSC therapy of ALI/ARDS. RESULTS: At 24 h after transfection, nuclear-targeted electroporation using an MC-ANGPT1 vector resulted in a 3.7-fold greater increase in human ANGPT1 protein in MSC conditioned media compared to the use of a plasmid ANGPT1 (pANGPT1) vector (2048 ± 567 pg/mL vs. 552.1 ± 33.5 pg/mL). In the lipopolysaccharide (LPS)-induced ALI model, administration of pANGPT1 transfected MSCs significantly reduced bronchoalveolar lavage (BAL) neutrophil counts by 57%, while MC-ANGPT1 transfected MSCs reduced it by 71% (p < 0.001) by Holm-Sidak's multiple comparison test. Moreover, compared to pANGPT1, the MC-ANGPT1 transfected MSCs significantly reduced pulmonary inflammation, as observed in decreased levels of proinflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), and macrophage inflammatory protein-2 (MIP-2). pANGPT1-transfected MSCs significantly reduced BAL albumin levels by 71%, while MC-ANGPT1-transfected MSCs reduced it by 85%. CONCLUSIONS: Overall, using a minicircle vector, we demonstrated an efficient and sustained expression of the ANGPT1 transgene in MSCs and enhanced the therapeutic effect on the ALI model compared to plasmid. These results support the potential benefits of MC-ANGPT1 gene enhancement of MSC therapy to treat ARDS.


Subject(s)
Acute Lung Injury , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Acute Lung Injury/genetics , Acute Lung Injury/therapy , Humans , Lipopolysaccharides , Lung , Mice , Transgenes
6.
Sci Rep ; 9(1): 18078, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31792313

ABSTRACT

Mesenchymal stem cells (MSCs) have been shown to exert immunomodulatory effects in both acute and chronic diseases. In acute inflammatory conditions like sepsis, cell therapy must be administered within hours of diagnosis, requiring "off-the-shelf" cryopreserved allogeneic cell products. However, their immunomodulatory potency, particularly in abilities to modulate innate immune cells, has not been well documented. Herein we compared the stabilities and functionalities of cultured versus thawed, donor-matched MSCs in modulating immune responses in vitro and in vivo. Cultured and thawed MSCs exhibited similar surface marker profiles and viabilities at 0 hr; however, thawed MSCs exhibited higher levels of apoptotic cells beyond 4 hrs. In vitro potency assays showed no significant difference between the abilities of both MSCs (donor-matched) to suppress proliferation of activated T cells, enhance phagocytosis of monocytes, and restore endothelial permeability after injury. Most importantly, in animals with polymicrobial sepsis, both MSCs significantly improved the phagocytic ability of peritoneal lavage cells, and reduced plasma levels of lactate and selected inflammatory cytokines without significant difference between groups. These results show comparable in vitro and in vivo immunomodulatory efficacy of thawed and fresh MSC products, providing further evidence for the utility of a cryopreserved MSC product for acute inflammatory diseases.


Subject(s)
Immunomodulation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Sepsis/therapy , Animals , Apoptosis , Cells, Cultured , Cryopreservation , Female , Humans , Lymphocyte Activation , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Phagocytosis , Sepsis/immunology , T-Lymphocytes/immunology
7.
Sci Rep ; 9(1): 20304, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31889120

ABSTRACT

Mesenchymal stromal cells (MSCs) are a potential therapy for many chronic inflammatory diseases due to their regenerative, immunologic and anti-inflammatory properties. The two-way dialogue between MSCs and macrophages is crucial to tissue regeneration and repair. Previous research demonstrated that murine adipose-derived MSC conditioned medium (ASCcm) reprograms macrophages to an M2-like phenotype which protects from experimental colitis and sepsis. Here, our focus was to determine the molecular mechanism of lipid droplet biogenesis in macrophages re-educated using ASCcm. Adipose-derived MSC conditioned medium promotes phosphorylation of AKT/mTOR pathway proteins in macrophages. Furthermore, increased expression of PPARγ, lipid droplet biogenesis and PGE2 synthesis were observed in M2-like phenotype macrophages (high expression of arginase 1 and elevated IL-10). Treatment with mTOR inhibitor rapamycin or PPARγ inhibitor GW9662 suppressed lipid droplets and PGE2 secretion. However, these inhibitors had no effect on arginase-1 expression. Rapamycin, but not GW9662, inhibit IL-10 secretion. In conclusion, we demonstrate major effects of ASCcm to reprogram macrophage immunometabolism through mTOR and PPARγ dependent and independent pathways.


Subject(s)
Adipose Tissue/cytology , Cell Communication , Lipid Droplets/metabolism , Lipid Metabolism , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Signal Transduction , Models, Biological , PPAR gamma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
8.
Sci Rep ; 7: 46444, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28406244

ABSTRACT

Atherosclerosis is a chronic inflammatory cardiovascular disease that is responsible of high mortality worldwide. Evidence indicates that maladaptive autoimmune responses in the arterial wall play critical roles in the process of atherosclerosis. Cortistatin is a neuropeptide expressed in the vascular system and atherosclerotic plaques that regulates vascular calcification and neointimal formation, and inhibits inflammation in different experimental models of autoimmune diseases. Its role in inflammatory cardiovascular disorders is largely unexplored. The aim of this study is to investigate the potential therapeutic effects of cortistatin in two well-established preclinical models of atherosclerosis, and the molecular and cellular mechanisms involved. Systemic treatment with cortistatin reduced the number and size of atherosclerotic plaques in carotid artery, heart, aortic arch and aorta in acute and chronic atherosclerosis induced in apolipoprotein E-deficient mice fed a high-lipid diet. This effect was exerted at multiple levels. Cortistatin reduced Th1/Th17-driven inflammatory responses and increased regulatory T cells in atherosclerotic arteries and lymphoid organs. Moreover, cortistatin reduced the capacity of endothelial cells to bind and recruit immune cells to the plaque and impaired the formation of foam cells by enhancing cholesterol efflux from macrophages. Cortistatin emerges as a new candidate for the treatment of the clinical manifestations of atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Foam Cells/drug effects , Hyperlipidemias/complications , Neuropeptides/administration & dosage , Plaque, Atherosclerotic/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Foam Cells/metabolism , Hyperlipidemias/genetics , Mice , Mice, Knockout, ApoE , Neuropeptides/pharmacology , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
9.
Ann N Y Acad Sci ; 1351: 89-98, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25951888

ABSTRACT

Cortistatin is a neuropeptide isolated from cortical brain regions, showing high structural homology and sharing many functions with somatostatin. However, cortistatin exerts unique functions in the central nervous and immune systems, including decreasing locomotor activity, inducing sleep-promoting effects, and deactivating inflammatory and T helper (TH )1/TH 17-driven responses in preclinical models of sepsis, arthritis, multiple sclerosis, and colitis. Besides its release by cortical and hippocampal interneurons, cortistatin is produced by macrophages, lymphocytes, and peripheral nociceptive neurons in response to inflammatory stimuli, supporting a physiological role of cortistatin in the immune and nociceptive systems. Cortistatin-deficient mice have been shown to have exacerbated nociceptive responses to neuropathic and inflammatory pain sensitization. However, a paradoxical effect has been observed in studies of immune disorders, in which, despite showing competent inflammatory/autoreactive responses, cortistatin-deficient mice were partially resistant to systemic autoimmunity and inflammation. This unexpected phenotype was associated with elevated circulating glucocorticoids and anxiety-like behavior. These findings support cortistatin as a novel multimodal therapeutic approach to treat autoimmunity and clinical pain and identify it as a key endogenous component of the neuroimmune system related to stress responses.


Subject(s)
Anti-Inflammatory Agents/metabolism , Autoimmune Diseases/immunology , Cerebral Cortex/metabolism , Neuropeptides/metabolism , Sleep/physiology , Animals , Disease Models, Animal , Humans , Inflammation/immunology , Macrophages/immunology , Mice , Motor Activity/physiology , Nociceptors/immunology , Somatostatin/metabolism , Th1 Cells/immunology , Th17 Cells/immunology
10.
Arthritis Rheumatol ; 66(2): 327-39, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24504805

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) is an autoimmune disease caused by loss of immunologic self tolerance and characterized by chronic joint inflammation. Cells isolated from human amniotic membrane (HAMCs) were recently found to display immunosuppressive properties. The aim of this study was to characterize the effect of HAMCs on antigen-specific T cell responses in RA patients and to evaluate their therapeutic potential in a preclinical experimental model of RA. METHODS: We investigated the effects of HAMCs on collagen-reactive T cell proliferation and cytokine production, on the production of mediators of inflammation by synoviocytes, and on the generation of Treg cells in peripheral blood mononuclear cells and synovial membrane cells isolated from RA patients. Mice with collagen-induced arthritis (CIA) were treated with HAMCs after disease onset, and clinical scores and joint levels of mediators of inflammation were evaluated. We determined Th1/Th17-mediated autoreactive responses in the mice by measuring the proliferation and the cytokine profile of lymph node cells restimulated with collagen. RESULTS: Treatment with HAMCs suppressed synovial inflammatory responses and antigen-specific Th1/Th17 activation in cells isolated from RA patients. Moreover, HAMCs stimulated the generation of human CD4+CD25+FoxP3+ Treg cells with a capacity to suppress collagen-specific T cell responses. Systemic infusion of HAMCs significantly reduced the incidence and severity of CIA by down-regulating the 2 deleterious components of disease: Th1-driven autoimmunity and inflammation. In mice with CIA, HAMC treatment decreased the production of various inflammatory cytokines and chemokines in the joints, impaired antigen-specific Th1/Th17 cell expansion in the lymph nodes, and generated peripheral antigen-specific Treg cells. HAMCs also protected the mice from experimental sepsis, inflammatory bowel disease, and autoimmune encephalomyelitis. CONCLUSION: HAMCs have emerged as attractive candidates for a cell-based therapy for RA.


Subject(s)
Amnion/cytology , Arthritis, Experimental/therapy , Cell- and Tissue-Based Therapy/methods , Colitis/therapy , Encephalomyelitis/therapy , Sepsis/therapy , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Proliferation , Cells, Cultured , Colitis/metabolism , Colitis/pathology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis/metabolism , Encephalomyelitis/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Sepsis/metabolism , Sepsis/pathology , Synovial Membrane/metabolism , Synovial Membrane/pathology , T-Lymphocytes/pathology , Treatment Outcome
11.
Brain Behav Immun ; 37: 152-63, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24321213

ABSTRACT

Adrenomedullin is a neuropeptide known for its cardiovascular activities and anti-inflammatory effects. Here, we investigated the effect of adrenomedullin in a model of experimental autoimmune encephalomyelitis (EAE) that mirrors chronic progressive multiple sclerosis. A short-term systemic treatment with adrenomedullin reduced clinical severity and incidence of EAE, the appearance of inflammatory infiltrates in spinal cord and the subsequent demyelination and axonal damage. This effect was exerted at multiple levels affecting both early and late events of the disease. Adrenomedullin decreased the presence/activation of encephalitogenic Th1 and Th17 cells and down-regulated several inflammatory mediators in peripheral lymphoid organs and central nervous system. Noteworthy, adrenomedullin inhibited the production by encephalitogenic cells of osteopontin and of Granulocyte Macrophage Colony-Stimulating Factor (GM-CSF), two critical cytokines in the development of EAE. At the same time, adrenomedullin increased the number of IL-10-producing regulatory T cells with suppressive effects on the progression of EAE. Furthermore, adrenomedullin generated dendritic cells with a semi-mature phenotype that impaired encephalitogenic responses in vitro and in vivo. Finally, adrenomedullin regulated glial activity and favored an active program of neuroprotection/regeneration. Therefore, the use of adrenomedullin emerges as a novel multimodal therapeutic approach to treat chronic progressive multiple sclerosis.


Subject(s)
Adrenomedullin/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Brain/drug effects , Brain/immunology , Chronic Disease , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Spinal Cord/drug effects , Spinal Cord/immunology , Spleen/drug effects , Spleen/immunology
12.
J Immunol ; 191(5): 2144-54, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23918980

ABSTRACT

Cortistatin is a cyclic-neuropeptide produced by brain cortex and immune cells that shows potent anti-inflammatory activity. In this article, we investigated the effect of cortistatin in two models of experimental autoimmune encephalomyelitis (EAE) that mirror chronic and relapsing-remitting multiple sclerosis. A short-term systemic treatment with cortistatin reduced clinical severity and incidence of EAE, the appearance of inflammatory infiltrates in spinal cord, and the subsequent demyelination and axonal damage. This effect was associated with a reduction of the two deleterious components of the disease, namely, the autoimmune and inflammatory response. Cortistatin decreased the presence/activation of encephalitogenic Th1 and Th17 cells in periphery and nervous system, and downregulated various inflammatory mediators, whereas it increased the number of regulatory T cells with suppressive effects on the encephalitogenic response. Moreover, cortistatin regulated glial activity and favored an active program of neuroprotection/regeneration. We further used cortistatin-deficient mice to investigate the role of endogenous cortistatin in the control of immune responses. Surprisingly, cortistatin-deficient mice were partially resistant to EAE and other inflammatory disorders, despite showing competent inflammatory/autoreactive responses. This unexpected phenotype was associated with elevated circulating glucocorticoids and an anxiety-like behavior. Our findings provide a powerful rationale for the assessment of the efficacy of cortistatin as a novel multimodal therapeutic approach to treat multiple sclerosis and identify cortistatin as a key endogenous component of neuroimmune system.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Neuropeptides/metabolism , T-Lymphocytes/drug effects , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology
13.
Brain Behav Immun ; 30: 54-60, 2013 May.
Article in English | MEDLINE | ID: mdl-23376169

ABSTRACT

Ghrelin is an important gastrointestinal hormone that regulates feeding and metabolism. Moreover, ghrelin is produced by immune cells and shows potent anti-inflammatory activities. Here, we investigated its effect in two models of experimental autoimmune encephalomyelitis (EAE) that mirror chronic and relapsing-remitting multiple sclerosis. A short systemic treatment with ghrelin after the disease onset reduced clinical severity and incidence of both forms of EAE, which was associated with a decrease in inflammatory infiltrates in spinal cord and in the subsequent demyelination. This therapeutic effect was exerted through the reduction of the autoimmune and inflammatory components of the disease. Ghrelin decreased the presence/activation of encephalitogenic Th1 and Th17 cells in periphery and nervous system, down-regulated various inflammatory mediators, and induced regulatory T cells. In summary, our findings provide a powerful rationale for the assessment of the efficacy of ghrelin as a novel therapeutic approach for treating multiple sclerosis through distinct immunomodulatory mechanisms and further support the concept that the neuroendocrine and immune systems crosstalk to finely tune the final immune response of our body.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Ghrelin/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Th1 Cells/drug effects , Th17 Cells/drug effects , Animals , Cytokines/metabolism , Down-Regulation/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Forkhead Transcription Factors/metabolism , Ghrelin/pharmacology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Mice , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/metabolism , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
14.
Arthritis Rheum ; 65(5): 1390-401, 2013 May.
Article in English | MEDLINE | ID: mdl-23371349

ABSTRACT

OBJECTIVE: To investigate the role of the antiinflammatory neuropeptide cortistatin in chronic pain evoked by joint inflammation. METHODS: Thermal and mechanical hyperalgesia was evoked in mouse knee joints by intraplantar injection of tumor necrosis factor α and intraarticular infusion of Freund's complete adjuvant, and the analgesic effects of cortistatin, administered centrally, peripherally, and systemically, were assessed. In addition, the effects of cortistatin on the production of nociceptive peptides and the activation of pain signaling were assayed in dorsal root ganglion cultures and in inflammatory pain models. The role of endogenous cortistatin in pain sensitization and perpetuation of chronic inflammatory states was evaluated in cortistatin-deficient mice. Finally, the effect of noxious/inflammatory stimuli in the production of cortistatin by the peripheral nociceptive system was assayed in vitro and in vivo. RESULTS: Expression of cortistatin was observed in peptidergic nociceptors of the peripheral nociceptive system, and endogenous cortistatin was found to participate in the tuning of pain sensitization, especially in pathologic inflammatory conditions. Results showed that cortistatin acted both peripherally and centrally to reduce the tactile allodynia and heat hyperalgesia evoked by arthritis and peripheral tissue inflammation in mice, via mechanisms that were independent of its antiinflammatory action. These mechanisms involved direct action on nociceptive neurons and regulation of central sensitization. The analgesic effects of cortistatin in murine arthritic pain were linked to binding of the neuropeptide to somatostatin and ghrelin receptors, activation of the G protein subunit Gαi , impairment of ERK signaling, and decreased production of calcitonin gene-related peptide in primary nociceptors. CONCLUSION: These findings indicate that cortistatin is an antiinflammatory factor with potent analgesic effects that may offer a new approach to pain therapy in pathologic inflammatory states, including osteoarthritis and rheumatoid arthritis.


Subject(s)
Analgesia , Arthritis/drug therapy , Hyperalgesia/drug therapy , Neuropeptides/pharmacology , Pain/drug therapy , Animals , Arthritis/chemically induced , Arthritis/metabolism , Calcitonin Gene-Related Peptide/metabolism , Central Nervous System Sensitization , Disease Models, Animal , Drug Therapy, Combination , Female , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/toxicity , GTP-Binding Protein alpha Subunits/metabolism , Ghrelin/metabolism , Ghrelin/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Injections, Intra-Articular , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/deficiency , Neuropeptides/metabolism , Pain/chemically induced , Pain/metabolism , Pain Threshold , Protein Binding , Receptors, Ghrelin/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Somatostatin/pharmacology , Stifle/drug effects , Stifle/metabolism , Stifle/physiopathology , Tumor Necrosis Factor-alpha/toxicity
15.
Gut ; 62(8): 1131-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22637701

ABSTRACT

OBJECTIVE: To investigate the effect of adipose-derived mesenchymal stromal cells (ASCs) on the activation state of macrophages (MΦ) in vitro, and the potential therapeutic effect of these cells in experimental colitis and sepsis. DESIGN: Murine bone marrow-derived macrophages were cultured with ASCs or with ASC conditioned media (ASC-MΦ) and characterised for the expression of several regulatory macrophage markers, including enzymes and cytokines, and for their immunomodulatory capacity in vitro. The therapeutic effect was investigated of ASC-MΦ in two models of experimental inflammatory colitis induced by trinitrobenzene sulphonic acid and dextran sodium sulphate, and in polymicrobial sepsis induced by caecal ligation and puncture. RESULTS: ASC-MΦ showed a phenotype that clearly differed from the classically activated macrophages or the alternatively activated macrophages induced by interleukin (IL)-4, characterised by high arginase activity, increased production of IL-10 upon restimulation and potent immunosuppressive activity on T cells and macrophages. Activation of cyclo-oxygenase-2 on ASCs seems to be critically involved in inducing this phenotype. Systemic infusion of ASC-MΦ inhibited colitis in mice, reducing mortality and weight loss while lowering the colonic and systemic levels of inflammatory cytokines. Importantly, therapeutic injection of ASC-MΦ in established chronic colitis alleviated its progression and avoided disease recurrence. Moreover, ASC-MΦ protected from severe sepsis by reducing the infiltration of inflammatory cells into various organs and by downregulating the production of several inflammatory mediators, where ASC-MΦ-derived IL-10 played a critical role. CONCLUSION: ASCs induce a distinct regulatory activation state of macrophages which possess potent immunomodulatory ability and therapeutic potential in inflammatory bowel diseases and sepsis.


Subject(s)
Adipose Tissue/cytology , Colitis/prevention & control , Macrophages/transplantation , Mesenchymal Stem Cells/immunology , Sepsis/prevention & control , Acute Disease , Animals , Bone Marrow Cells/immunology , Cells, Cultured , Coculture Techniques , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Culture Media, Conditioned , Cytokines/biosynthesis , Disease Models, Animal , Humans , Immunosuppression Therapy/methods , Inflammation Mediators/metabolism , Macrophage Activation/immunology , Male , Mice , Mice, Inbred BALB C , Sepsis/immunology , Systemic Inflammatory Response Syndrome/prevention & control , Treatment Outcome
16.
Article in English | MEDLINE | ID: mdl-23094830

ABSTRACT

Vasoactive intestinal peptide (VIP) is a well-known immunoregulatory neuropeptide produced by the immune system in response to inflammation, autoimmunity or alloantigens as a natural endogenous mechanism of induction of tolerance. VIP has been proven therapeutically effective in various experimental models of autoimmune disorders and recently in human sarcoidosis. Numerous studies clearly show that VIP exerts its immunomodulatory effects by downregulating both inflammatory and Th1 responses. Recent evidences suggest that new actors enter in scene to play a role in this scenario of tolerance. By inducing antigen-specific regulatory T cells and tolerogenic dendritic cells, VIP seems to reinforce/reinstall immune tolerance, especially under autoimmune conditions. Transplantation is also a condition where VIP-related therapies emerge as promising tools for clinical application. Induction of alloantigen-specific tolerance is critical to achieve organ transplant tolerance and to avoid graft-versus-host responses following allogeneic hematopoietic transplantation. This review will focus on describing the capacity of VIP to induce suppressive/regulatory immune cells and how we can manage this cell-based therapeutic strategy to induce transplant tolerance in subjects free of immunosuppressive drugs.


Subject(s)
Organ Transplantation/methods , Transplantation Immunology/drug effects , Vasoactive Intestinal Peptide/therapeutic use , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/methods , Humans , Immune Tolerance/drug effects , T-Lymphocytes/immunology
17.
Article in English | MEDLINE | ID: mdl-23094829

ABSTRACT

A hallmark in most neurological disorders is a massive neuronal cell death, in which uncontrolled immune response is usually involved, leading to neurodegeneration. The vasoactive intestinal peptide (VIP) is a pleiotropic peptide that combines neuroprotective and immunomodulatory actions. Alterations on VIP/VIP receptors in patients with neurodenegerative diseases, together with its involvement in the development of embryonic nervous tissue, and findings found in VIP-deficient mutant mice, have showed the relevance of this endogenous peptide in normal physiology and in pathologic states of the central nervous system (CNS). In this review, we will summarize the role of VIP in normal CNS and in neurological disorders. The studies carried out with this peptide have demonstrated its therapeutic effect and render it as an attractive candidate to be considered in several neurological disorders linked to neuroinflammation or abnormal neural development.


Subject(s)
Nervous System Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Vasoactive Intestinal Peptide/therapeutic use , Alzheimer Disease/physiopathology , Animals , Autistic Disorder/physiopathology , Brain/drug effects , Brain/physiology , Brain Injuries/physiopathology , Developmental Disabilities/drug therapy , Down Syndrome/physiopathology , Encephalitis/drug therapy , Female , Fetal Alcohol Spectrum Disorders/physiopathology , Humans , Mice , Multiple Sclerosis/physiopathology , Neuroprotective Agents/administration & dosage , Parkinson Disease/physiopathology , Pregnancy , Receptors, Vasoactive Intestinal Peptide/drug effects , Vasoactive Intestinal Peptide/administration & dosage , Vasoactive Intestinal Peptide/physiology
18.
Neuroendocrinology ; 94(2): 89-100, 2011.
Article in English | MEDLINE | ID: mdl-21734355

ABSTRACT

Although necessary to eliminate pathogens, inflammation can lead to serious deleterious effects in the host if left unchecked. During the inflammatory response, further damage may arise from potential autoimmune responses occurring when the immune cells and molecules that respond to pathogen-derived antigens also react to self-antigens. In this sense, the identification of endogenous factors that control exacerbated immune responses is a key goal for the development of new therapeutic approaches for inflammatory and autoimmune diseases. Some neuropeptides that are produced during the ongoing inflammatory response have emerged as endogenous anti-inflammatory agents that could collaborate in tuning the balanced steady state of the immune system. These neuropeptides participate in maintaining immune tolerance through two distinct mechanisms: by regulating the balance between pro-inflammatory and anti-inflammatory factors, and by inducing the emergence of regulatory T cells with suppressive activity against autoreactive T cell effectors. Indeed, a functioning neuropeptide system contributes to general health, and alterations in the levels of these neuropeptides and/or their receptors lead to changes in susceptibility to inflammatory and autoimmune diseases. Recently, we found that some neuropeptides also have antimicrobial and antiparasitic actions, suggesting that they could act as primary mediators of innate defense, even in the most primitive organisms. In this review, we use the vasoactive intestinal peptide as example of an immunomodulatory neuropeptide to summarize the most relevant data found for other neuropeptides with similar characteristics, including adrenomedullin, urocortin, cortistatin and ghrelin.


Subject(s)
Immune System/physiology , Neuropeptides/immunology , Neuropeptides/physiology , Animals , Homeostasis , Humans , Immunologic Factors/physiology , Immunologic Factors/therapeutic use , Immunosuppressive Agents/pharmacology , Inflammation/physiopathology , Neuropeptides/therapeutic use , Neurosecretory Systems/physiology
19.
Curr Pharm Des ; 16(28): 3158-72, 2010.
Article in English | MEDLINE | ID: mdl-20687881

ABSTRACT

Because there are no particular molecular signatures of self, autoimmunity is the inevitable evolutionary price of being able to make effective responses against a wide variety of pathogens by the immune system. Without the various phenomena referred to as immune tolerance, the organism would surely self-destruct. Considerable evidence suggests that various endogenous neuropeptides play a major role in the education of our immune system to be self-tolerant. The fact that neuropeptides regulate various layers involved in maintenance of tolerance, including regulation of the balance between pro-inflammatory and anti-inflammatory responses and between self-reactive Th1/Th17 cells and regulatory T cells, makes them attractive candidates for the development of new therapies for the treatment of autoimmune disorders. Here we use the vasoactive intestinal peptide of a prototype of immunomodulatory neuropeptide to review the most relevant data found for other neuropeptides with similar characteristics, including melanocyte-stimulating hormone, urocortin, adrenomedullin, neuropeptide Y, cortistatin and ghrelin. We also evaluate the challenges that must be overcome before achieving their clinical application and offer our opinion on how a physiologically functional neuropeptide system contributes to general health.


Subject(s)
Autoimmune Diseases/drug therapy , Autoimmunity/drug effects , Immunologic Factors/therapeutic use , Neuroimmunomodulation/drug effects , Neuropeptides/pharmacology , Neuropeptides/therapeutic use , Animals , Autoimmune Diseases/immunology , Autoimmunity/immunology , Humans , Immune Tolerance/drug effects , Immune Tolerance/immunology , Models, Biological , Vasoactive Intestinal Peptide/pharmacology , Vasoactive Intestinal Peptide/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...