Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892159

ABSTRACT

Mesenchymal stem cells (MSCs) are known for their immunosuppressive properties. Based on the demonstrated anti-inflammatory effect of mouse MSCs from hair follicles (moMSCORS) in a murine wound closure model, this study evaluates their potential for preventing type 1 diabetes (T1D) in C57BL/6 mice. T1D was induced in C57BL/6 mice by repeated low doses of streptozotocin. moMSCORS were injected intravenously on weekly basis. moMSCORS reduced T1D incidence, the insulitis stage, and preserved insulin production in treated animals. moMSCORS primarily exerted immunomodulatory effects by inhibiting CD4+ T cell proliferation and activation. Ex vivo analysis indicated that moMSCORS modified the cellular immune profile within pancreatic lymph nodes and pancreatic infiltrates by reducing the numbers of M1 pro-inflammatory macrophages and T helper 17 cells and upscaling the immunosuppressive T regulatory cells. The proportion of pathogenic insulin-specific CD4+ T cells was down-scaled in the lymph nodes, likely via soluble factors. The moMSCORS detected in the pancreatic infiltrates of treated mice presumably exerted the observed suppressive effect on CD4+ through direct contact. moMSCORS alleviated T1D symptoms in the mouse, qualifying as a candidate for therapeutic products by multiple advantages: non-invasive sampling by epilation, easy access, permanent availability, scalability, and benefits of auto-transplantation.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Hair Follicle , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice, Inbred C57BL , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Mesenchymal Stem Cell Transplantation/methods , Diabetes Mellitus, Experimental/therapy , Male , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Pancreas/pathology , Pancreas/metabolism
2.
Eur J Pharmacol ; 971: 176509, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38493914

ABSTRACT

Acute respiratory distress syndrome (ARDS) became a focus of intensive research due to its death toll during the Covid-19 pandemic. An uncontrolled and excessive inflammatory response mediated by proinflammatory molecules such as high mobility group box protein 1 (HMGB1), IL-6, and TNF mounts as a response to infection. In this study, ethyl pyruvate (EP), a known inhibitor of HMGB1, was tested in the model of murine ARDS induced in C57BL/6 mice by intranasal administration of polyinosinic:polycytidylic acid (poly(I:C)). Intraperitoneal administration of EP ameliorated the ARDS-related histopathological changes in the lungs of poly(I:C)-induced ARDS and decreased numbers of immune cells in the lungs, broncho-alveolar lavage fluid and draining lymph nodes (DLN). Specifically, fewer CD8+ T cells and less activated CD4+ T cells were observed in DLN. Consequently, the lungs of EP-treated animals had fewer damage-inflicting CD8+ cells and macrophages. Additionally, the expression and production of proinflammatory cytokines, IL-17, IFN-γ and IL-6 were downregulated in the lungs. The expression of chemokine CCL5 which recruits immune cells into the lungs was also reduced. Finally, EP downregulated the expression of HMGB1 in the lungs. Our results imply that EP should be further evaluated as a potential candidate for ARDS therapy.


Subject(s)
HMGB1 Protein , Pyruvates , Respiratory Distress Syndrome , Humans , Animals , Mice , CD8-Positive T-Lymphocytes/metabolism , HMGB1 Protein/metabolism , Interleukin-6 , Pandemics , Disease Models, Animal , Mice, Inbred C57BL , Respiratory Distress Syndrome/drug therapy
3.
Front Immunol ; 14: 1271699, 2023.
Article in English | MEDLINE | ID: mdl-37915588

ABSTRACT

Innate lymphoid cells type 3 (ILC3s) are the first line sentinels at the mucous tissues, where they contribute to the homeostatic immune response in a major way. Also, they have been increasingly appreciated as important modulators of chronic inflammatory and autoimmune responses, both locally and systemically. The proper identification of ILC3 is of utmost importance for meaningful studies on their role in immunity. Flow cytometry is the method of choice for the detection and characterization of ILC3. However, the analysis of ILC3-related papers shows inconsistency in ILC3 phenotypic definition, as different inclusion and exclusion markers are used for their identification. Here, we present these discrepancies in the phenotypic characterization of human and mouse ILC3s. We discuss the pros and cons of using various markers for ILC3 identification. Furthermore, we consider the possibilities for the efficient isolation and propagation of ILC3 from different organs and tissues for in-vitro and in-vivo studies. This paper calls upon uniformity in ILC3 definition, isolation, and propagation for the increased possibility of confluent interpretation of ILC3's role in immunity.


Subject(s)
Immunity, Innate , Lymphocytes , Humans , Animals , Mice , Inflammation
4.
Molecules ; 28(8)2023 Apr 11.
Article in English | MEDLINE | ID: mdl-37110604

ABSTRACT

Recent data indicate the link between the number and function of T regulatory cells (Treg) in the gut immune tissue and initiation and development of autoimmunity associated with type 1 diabetes (T1D). Since type 3 innate lymphoid cells (ILC3) in the small intestine are essential for maintaining FoxP3+ Treg and there are no data about the possible role of ILC3 in T1D pathogenesis, the aim of this study was to explore ILC3-Treg link during the development of T1D. Mature diabetic NOD mice had lower frequencies of IL-2-producing ILC3 and Treg in small intestine lamina propria (SILP) compared to prediabetic NOD mice. Similarly, in multiple low doses of streptozotocin (MLDS)-induced T1D in C57BL/6 mice, hyperglycemic mice exhibited lower numbers of ILC3, IL-2+ ILC3 and Treg in SILP compared to healthy controls. To boost T1D severity, mice were treated with broad-spectrum antibiotics (ABX) for 14 days prior to T1D induction by MLDS. The higher incidence of T1D in ABX-treated mice was associated with significantly lower frequencies of IL-2+ ILC3 and FoxP3+ Treg in SILP compared with mice without ABX treatment. The obtained findings show that the lower proportions of IL-2-expressing ILC3 and FoxP3+ Treg in SILP coincided with diabetes progression and severity.


Subject(s)
Diabetes Mellitus, Type 1 , Mice , Animals , Diabetes Mellitus, Type 1/pathology , Mice, Inbred NOD , T-Lymphocytes, Regulatory , Interleukin-2 , Immunity, Innate , Mice, Inbred C57BL , Lymphocytes/pathology , Transcription Factors , Intestine, Small/pathology , Forkhead Transcription Factors/genetics
5.
Medicina (Kaunas) ; 58(6)2022 May 30.
Article in English | MEDLINE | ID: mdl-35744008

ABSTRACT

Background and Objectives: Reducing time of treatment during COVID-19 outbreaks has been recommended by the leading Radiation Oncology societies. Still minimizing radiation induced tissue toxicity is one of the most important issues in breast cancer patients. The study aimed to investigate compliance, clinical and dosimetry normal tissue toxicity, and cosmetic results between moderated and ultra-fractionated regimes for breast cancer patients during COVID-19 pandemic. Materials and Methods: This pilot prospective randomized study included 60 patients with early breast cancer after preserving surgery, 27 patients advocated to ultra-hypofractionated whole-breast three dimensional (3D) conformal radiotherapy of 26 Gy in 5 fractions over 1 week and 33 patients with moderate fractionated breast 3D conformal radiotherapy patients between March 2020 and July 2020, during the COVID pandemic outbreak. The compliance to treatment, dosimetric parameters, acute and late skin toxicity, subcutaneous tissue toxicity, cosmetic results and clinical follow up for 18 months for the two regimes were analyzed and compared. Results: When two regimes were compared 5 fraction group had significantly lower prevalence of newly infected cases of SARS-CoV-2 and thus delayed/interrupted treatment (p = 0.05), comparable grade 1 CTCAE v5, acute skin toxicity (p = 0.18), Grade 1 Radiation Morbidity Scoring Scheme (RESS) subcutaneous tissue toxicity (p = 0.18), Grade 1 RESS late skin toxicity (p = 0.88) and cosmetic results (p = 0.46). Dosimetric results reveled that patients in 5 fraction group received significantly lower median ipsilateral lung doses (p < 0.01) in addition to left breast cancer patients that received significantly lower median heart dose (p < 0.01) and median left anterior descending artery (LAD) dose (p < 0.01). Conclusion: Ultra-hypofractionated radiotherapy for breast cancer is comparable to moderate hypofractionation regimen regarding grade 1 acute skin toxicity, grade 1 subcutaneous tissue toxicity, late skin toxicity and cosmetic results. Application of ultra-hypofractionated radiotherapy with significantly lower radiation doses for lung and heart could be crucial in reducing the risk of acute/late pulmonary and heart radiation-induced toxicity.


Subject(s)
Breast Neoplasms , COVID-19 , Radiation Injuries , Radiotherapy, Conformal , Breast Neoplasms/epidemiology , Breast Neoplasms/radiotherapy , Breast Neoplasms/surgery , Female , Humans , Pandemics , Prospective Studies , Radiotherapy, Conformal/adverse effects , Radiotherapy, Conformal/methods , SARS-CoV-2
6.
Inflamm Res ; 71(2): 169-182, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34999919

ABSTRACT

Ethyl pyruvate (EP) has potent influence on redox processes, cellular metabolism, and inflammation. It has been intensively studied in numerous animal models of systemic and organ-specific disorders whose pathogenesis involves a strong immune component. Here, basic chemical and biological properties of EP are discussed, with an emphasis on its redox and metabolic activity. Further, its influence on myeloid and T cells is considered, as well as on intracellular signaling beyond its effect on immune cells. Also, the effects of EP on animal models of chronic inflammatory and autoimmune disorders are presented. Finally, a possibility to apply EP as a treatment for such diseases in humans is discussed. Scientific papers cited in this review were identified using the PubMed search engine that relies on the MEDLINE database. The reference list covers the most important findings in the field in the past twenty years.


Subject(s)
Autoimmune Diseases/drug therapy , Inflammation/drug therapy , Pyruvates/therapeutic use , Animals , Disease Models, Animal , Humans , Myeloid Cells/drug effects , Pyruvates/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , T-Lymphocytes/drug effects
7.
Stem Cell Rev Rep ; 18(6): 2028-2044, 2022 08.
Article in English | MEDLINE | ID: mdl-35080748

ABSTRACT

Wound healing of acute full-thickness injuries and chronic non-healing ulcers leads to delayed wound closure, prolonged recovery period and hypertrophic scarring, generating a demand for an autologous cell therapy and a relevant pre-clinical research models for wound healing. In this study, an immunocompetent model for wound healing was employed using a syngeneic murine cell line of mesenchymal stem cells cultured from the mouse whisker hair follicle outer root sheath (named moMSCORS). moMSCORS were isolated using an air-liquid interface method, expanded in vitro and characterized according to the MSC definition criteria - cell viability, in vitro proliferation, MSC phenotype and multi-lineage differentiations. Moreover, upon applying moMSCORS in an in vivo full-thickness wound model in the syngeneic C57BL/6 mice, the treated wounds displayed different morphology to that of the untreated wound beds. Quantitative evaluation of angiogenesis, granulation and wound closure involving clinical scoring and software-based quantification indicated a lower degree of inflammation in the treated wounds. Histological staining of treated wounds by the means of H&E, Alcian Blue, PicroSirius Red and αSMA immune labelling showed lower cellularity, less collagen filaments as well as thinner dermal and epidermal layers compared with the untreated wounds, indicating a general reduction of hypertrophic scars. The decreased inflammation, accelerated wound closure and non-hypertrophic scarring, which were facilitated by moMSCORS, hereby address a common problem of hypertrophic scars and non-physiological tissue properties upon wound closure, and additionally offer an in vivo model for the autologous cell-based wound healing.


Subject(s)
Mesenchymal Stem Cells , Skin Diseases , Animals , Cicatrix , Hair Follicle , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Skin Diseases/metabolism , Wound Healing/physiology
8.
Life Sci ; 288: 120184, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34838848

ABSTRACT

AIMS: Rosmarinic acid (RA) is a polyphenol that occurs in plants of the Lamiaceae family. Phenethyl ester of RA (PERA), a novel RA derivative, has been developed and evaluated in vivo in an animal model of type 1 diabetes (T1D). METHODS: T1D was induced in male C57BL/6 mice using multiple low doses of streptozotocin (STZ) administered intraperitoneally for 5 consecutive days. Intraperitoneal administration of PERA (2.5 mg/kg bw) began from the first STZ injection and continued for 20 days. KEY FINDINGS: PERA-treated mice exhibited lower incidence of T1D (monitored up to 38 days from the disease induction), and fluorescent histochemical analysis showed that their pancreatic islets expressed more insulin. PERA treatment significantly down-regulated the proportions of CD11b+ and CD11c+ myeloid cells in the immune cell infiltrates in the pancreatic islets early during T1D pathogenesis (on day 9 after T1D induction), while on day 15, PERA significantly reduced the proportions of CD11c+, CD8+, Th1 and Th17 cells. Simultaneously, it was found that the cells from the pancreatic infiltrates of PERA-treated mice produced significantly less reactive oxygen species than cells from the control group. SIGNIFICANCE: These findings suggest that PERA efficiently prevented T1D development in mice. Interestingly, PERA attenuated the inflammatory process in the islets through temporally specific interference with the innate and adaptive immune response and therefore shows great promise for further clinical evaluation as a novel T1D therapeutic.


Subject(s)
Autoimmunity , Cinnamates/pharmacology , Depsides/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/prevention & control , Esters/chemistry , Islets of Langerhans/drug effects , Animals , Cinnamates/chemistry , Depsides/chemistry , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Phenylethyl Alcohol/chemistry , Rosmarinic Acid
9.
Biomolecules ; 11(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34944410

ABSTRACT

Ethyl pyruvate (EP) has profound anti-inflammatory and immunomodulatory properties. Here, its effects were determined on experimental autoimmune myocarditis (EAM) induced in mice by heart-specific myosin-alpha heavy chain peptide immunization. EP was applied intraperitoneally, daily, starting with the immunization. Severity of EAM was determined by histological assessment of immune cell infiltrates into the heart. Cells were phenotypically characterized by flow cytometry. Concentration of cytokines in cell culture supernatants and sera was determined by ELISA. EP reduced the infiltration of immune cells into the heart and lessened heart inflammation. Smaller number of total immune cells, as well as of CD11b+ and CD11c+ cells were isolated from the hearts of EP-treated mice. A reduced number of antigen-presenting cells, detected by anti-CD11c, MHC class II and CD86 antibodies, as well as of T helper (Th)1 and Th17 cells, detected by anti-CD4, IFN-γ and IL-17 antibodies, was determined in mediastinal lymph nodes draining the heart, in parallel. In the spleen, only the number of CD11c+ cells were reduced, but not of the other examined populations, thus implying limited systemic effect of EP. Reduced production of IFN-γ and IL-17 by myosin-alpha heavy chain peptide-restimulated cells of the lymph nodes draining the site of immunization was observed in EP-treated mice. Our results clearly imply that EP restrains autoimmunity in EAM. Therapeutic application of EP in the treatment of myocarditis in humans should be addressed in the forthcoming studies.


Subject(s)
Autoimmune Diseases/drug therapy , Cytokines/metabolism , Myocarditis/immunology , Pyruvates/administration & dosage , Animals , Antigen Presentation , Cells, Cultured , Culture Media/chemistry , Disease Models, Animal , Lymph Nodes/drug effects , Lymph Nodes/immunology , Male , Mice , Myocarditis/drug therapy , Phenotype , Pyruvates/pharmacology , Th1 Cells/immunology , Th17 Cells/immunology
10.
Cytokine ; 138: 155372, 2021 02.
Article in English | MEDLINE | ID: mdl-33246771

ABSTRACT

Obesity, a global health problem nowadays, is a state of low-grade chronic inflammation of adipose tissue (AT) associated with increased adipocyte growth and proliferation and immune cell polarization towards an inflammatory phenotype within the stromal vascular fraction (SVF). Pro-inflammatory cells in the AT produce mediators of inflammation (IL-1ß, TNF, macrophage migration inhibitory factor - MIF), thereby surpassing the anti-inflammatory response mediated by IL-10 and TGF-ß, cytokines produced by regulatory T (Treg) cells. In this study we demonstrate that the absence of the pro-inflammatory cytokine MIF led to obesity and inflammation in the visceral AT (VAT) in 6 months old MIF-/- mice. Besides the increment of pro-inflammatory AT macrophages and the enhanced production of TNF and IL-1ß, VAT of MIF-/- mice contained increased numbers of Treg cells. In situ proliferation of Treg cells did not differ between MIF-/- and wild type mice, but Treg cells isolated from the VAT of MIF-deficient mice, and not from the cervical lymph nodes, exhibited lower expression and production of IL-10 and TGF-ß. Additionally, SVF cells had significantly lower levels of STAT3 and IL-33, altogether indicating that VAT Treg cells in MIF-/- mice, albeit abundantly present, are not fully functional. These results indicate that MIF is a new regulator of VAT Treg cell function, necessary for their immunosuppressive activities.


Subject(s)
Adipose Tissue/metabolism , Gene Deletion , Inflammation/genetics , Interleukin-10/metabolism , Interleukin-33/metabolism , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Obesity/genetics , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/cytology , Transforming Growth Factor beta1/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism , Adipocytes/cytology , Animals , Immunosuppression Therapy , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Inbred C57BL , Stromal Vascular Fraction
11.
Molecules ; 25(18)2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32916780

ABSTRACT

Ethyl pyruvate (EP), a stable form of pyruvate, has shown beneficial effects in animal models of shock, ischemia/reperfusion injury, and sepsis due to its potent anti-oxidant and anti-inflammatory properties. Our recent study demonstrated that EP application prevented the clinical manifestation of type 1 diabetes in mice by augmenting regulatory T cell (Treg) number and function. Our present study shows that EP increases Treg proliferation and suppressive function (perforin and IL-10 expression) during in vitro differentiation from conventional CD4+CD25- T cells. Enhanced expansion of Treg after EP treatment correlated with increased ATP levels and relied on increased glycolysis. Inhibition of oxidative phosphorylation did not attenuate EP stimulatory effects, suggesting that this metabolic pathway was not mandatory for EP-driven Treg proliferation. Moreover, EP lowered the expression of carnitine palmitoyltransferase I, an enzyme involved in fatty acid oxidation. Further, the stimulatory effect of EP on Treg proliferation was not mediated through inhibition of the mTOR signaling pathway. When given in vivo either intraperitoneally or orally to healthy C57BL/6 mice, EP increased the number of Treg within the peritoneal cavity or gut-associated lymphoid tissue, respectively. In conclusion, EP promotes in vitro Treg proliferation through increased glycolysis and enhances Treg proliferation when administered in vivo.


Subject(s)
Cell Proliferation/drug effects , Glycolysis/drug effects , Pyruvates/pharmacology , T-Lymphocytes, Regulatory/cytology , Adenosine Triphosphate/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cell Differentiation , Fatty Acids/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Spleen/metabolism , T-Lymphocytes, Regulatory/drug effects
12.
Food Funct ; 11(9): 7793-7803, 2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32808624

ABSTRACT

Chokeberry (Aronia melanocarpa) fruit extracts (CE) are rich in polyphenols and usually exhibit immunomodulatory, anti-viral and anti-bacterial effects. We have previously shown that the CE used in this study activated macrophages and stimulated effector T cell differentiation in vitro. When applied orally to healthy mice, CE increased the proportion of CD11c+ dendritic cells in the gut-associated lymphoid tissue. CE-pretreated BALB/c mice readily eradicated orally ingested Listeria monocytogenes as evidenced by a slighter decrease in body weight and number of bacteria recovered from the spleen and reduced spleen size compared to the control infected mice. CE pretreatment in infected mice resulted in higher proportions of CD11b+ macrophages and CD8+ cytotoxic T cells both in the gut and the spleen. Phagocytosis, reactive oxygen species production and the proportions of activated CD86+ macrophages (CD11b+) and dendritic cells (CD11c+) were also enhanced in CE-pretreated infected mice. Furthermore, the expression of inducible nitric oxide synthase and IL-6 was increased in CE-pretreated infected mice and similar results were obtained in peritoneal macrophages in vitro. This effect of CE was associated with increased phosphorylation of IκB and Notch1 production. Finally, CE pretreatment elevated the proportion of perforin-producing cells in the spleen compared to control infected mice. This study demonstrates that prophylactic treatment with CE leads to more rapid eradication of bacterial infection with L. monocytogenes predominantly through increased activity of myeloid cells in the gut and in the spleen.


Subject(s)
Fruit/chemistry , Immunologic Factors/pharmacology , Listeria monocytogenes , Listeriosis/immunology , Photinia/chemistry , Plant Extracts/pharmacology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunomodulation , Intestine, Small/immunology , Listeria monocytogenes/isolation & purification , Listeriosis/drug therapy , Listeriosis/microbiology , Lymphoid Tissue/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/metabolism , Phagocytosis , Phytotherapy , Reactive Oxygen Species/metabolism , Spleen/immunology , Spleen/microbiology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
13.
Eur J Pharmacol ; 864: 172721, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31586630

ABSTRACT

Type 1 diabetes (T1D) is a multifactorial autoimmune disease that develops as a consequence of macrophage- and T cell-dependent pancreatic ß-cell death. Multiple approaches for induction of anti-inflammatory/regulatory mechanisms that would attenuate T1D have been utilized, with little or no beneficial effects. To achieve prolonged stimulation of regulatory immune cells, we orally introduced microparticles (MPs) loaded with all-trans retinoic acid (ATRA) and transforming growth factor-ß (TGF-ß) to C57BL/6 mice treated with multiple low doses of streptozotocin (MLDS) for T1D induction. Disease incidence was significantly lower in ATRA/TGF-ß MPs-treated mice, as was the degree of immune cell infiltration into the pancreatic islets. In Peyer's patches (PP), ATRA/TGF-ß MPs up-regulated tolerogenic dendritic cells (tolDCs) (CD11c+CD11b-CD103+), while the proportion of mature dendritic cells was not altered. This was accompanied by reduced Th1 and Th17 proportions and up-regulation of regulatory T cells (Tregs - CD4+CD25highFoxP3+). The immune cell composition in the pancreatic lymph nodes was similar to PP. Further, the proportion of effector Tbet+CD25med cells was decreased, while the proportion of Tbet+ Treg cells that specifically inhibit Th1 response was increased. Moreover, ATRA/TGF-ß MPs treatment resulted in increased Treg proliferation and frequency of CTLA-4+PD1+ and CD39+IL-10+ Tregs, suggestive of their higher suppressive capacity. Reduced pancreatic infiltration may have been a consequence of lower cell capacity for matrix degradation. In conclusion, oral application of ATRA/TGF-ß MPs ameliorated T1D through potentiation of tolDCs and Tregs, inhibition of Th1 response and prevention of the immune cell entrance into the islets.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Drug Carriers/chemistry , Microspheres , Transforming Growth Factor beta/pharmacology , Tretinoin/administration & dosage , Tretinoin/pharmacology , Administration, Oral , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/therapeutic use , Tretinoin/therapeutic use
14.
Sci Rep ; 8(1): 6337, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29679061

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a multifunctional protein that is involved in the development of gut-related inflammation. To investigate the role of MIF in the function of the intestinal barrier, we have explored intestinal permeability and gut-associated immune response in MIF-deficient (MIF-KO) mice. The absence of MIF provoked impairment of tight and adherens epithelial junctions in the colon through the disturbance of E-cadherin, zonula occludens-1, occludin and claudin-2 expression, which lead to the increase of intestinal barrier permeability. In these circumstances the diversity and content of gut microbiota in MIF-KO mice was considerably different compared to wild type mice. This change in microbiota was accompanied by an increased intestinal IgA concentration and a higher production of pro-inflammatory cytokines TNF and IFN-γ in mesenteric lymph nodes of MIF-KO mice. The forced changes of microbiota executed by antibiotics prevented the "leakage" of the barrier in MIF-KO mice, probably through up-regulation of occludin expression and normalization of cellular pore diameters. In addition, cytokine secretion was normalized after the treatment with antibiotics. These results suggest that MIF participates in the maintenance of physiological microbiota diversity and immunosurveillance, which in turn enables the proper intestinal barrier function.


Subject(s)
Intestinal Mucosa/metabolism , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/physiology , Adherens Junctions/metabolism , Animals , Colon/metabolism , Female , Gastrointestinal Microbiome , Inflammation/metabolism , Interferon-gamma/metabolism , Intestines/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Occludin/metabolism , Permeability , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
J Alzheimers Dis ; 61(2): 619-630, 2018.
Article in English | MEDLINE | ID: mdl-29254086

ABSTRACT

Alzheimer's disease (AD) is characterized by accumulation of amyloid-ß plaques that further promotes microglia-mediated neuroinflammatory responses and inflammation in the brain. Emerging data are revealing the relation between gut-associated lymphoid tissue (GALT) cells and CNS, as effector cells primed in the gut might home to the brain. This study aimed to determine cell composition of GALT in 5xFAD mice, an established model for AD. Immune cells isolated from Peyer's patches (PP) and mesenteric lymph nodes (MLN) were stained with surface and intracellular markers for T helper (Th) subpopulations, B lymphocytes and macrophages and analyzed cytofluorimetrically, while cytokine expression and production were determined by qPCR and ELISA, respectively. Inflammation was detected in GALT of 5xFAD mice with established AD pathology. Although the production of IFN-γ, IL-4, and IL-10 was comparable between the strains, lower IL-17 production was observed in PP and MLN cells. This phenomenon could not be attributed to a lower abundance of Th17 cells, or cytokines that initiate their formation or propagation (TGF-ß, IL-6, and IL-23). Also, reduced IL-17 production was not a consequence of altered Il-17 mRNA transcription or deficiency of Rorγt, a key transcription factor for IL-17. However, the expression of miR-155 (a non-coding micro RNA that promotes the development of Th17 cells), was significantly lower in MLN cells of 5xFAD mice. In contrast, mice without AD neuropathology did not have inflammation in GALT or altered Th17 numbers, nor decreased IL-17 production. In conclusion, the observed changes in GALT of 5xFAD mice mirror the disease progression and might reflect inadequate immune surveillance in the gut and lead to enhanced AD pathology.


Subject(s)
Alzheimer Disease/pathology , B-Lymphocytes/immunology , Interleukin-17/metabolism , MicroRNAs/metabolism , Th17 Cells/immunology , Alzheimer Disease/immunology , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Female , Lymph Nodes/pathology , Male , Mice , Mice, Transgenic , MicroRNAs/genetics , Peyer's Patches/pathology
16.
Front Immunol ; 9: 3130, 2018.
Article in English | MEDLINE | ID: mdl-30687329

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease in which a strong inflammatory response causes the death of insulin-producing pancreatic ß-cells, while inefficient regulatory mechanisms allow that response to become chronic. Ethyl pyruvate (EP), a stable pyruvate derivate and certified inhibitor of an alarmin-high mobility group box 1 (HMGB1), exerts anti-oxidant and anti-inflammatory properties in animal models of rheumatoid arthritis and encephalomyelitis. To test its therapeutic potential in T1D, EP was administered intraperitoneally to C57BL/6 mice with multiple low-dose streptozotocin (MLDS)-induced T1D. EP treatment decreased T1D incidence, reduced the infiltration of cells into the pancreatic islets and preserved ß-cell function. Apart from reducing HMGB1 expression, EP treatment successfully interfered with the inflammatory response within the local pancreatic lymph nodes and in the pancreas. Its effect was restricted to boosting the regulatory arm of the immune response through up-regulation of tolerogenic dendritic cells (CD11c+CD11b-CD103+) within the pancreatic infiltrates and through the enhancement of regulatory T cell (Treg) levels (CD4+CD25highFoxP3+). These EP-stimulated Treg displayed enhanced suppressive capacity reflected in increased levels of CTLA-4, secreted TGF-ß, and IL-10 and in the more efficient inhibition of effector T cell proliferation compared to Treg from diabetic animals. Higher levels of Treg were a result of increased differentiation and proliferation (Ki67+ cells), but also of the heightened potency for migration due to increased expression of adhesion molecules (CD11a and CD62L) and CXCR3 chemokine receptor. Treg isolated from EP-treated mice had the activated phenotype and T-bet expression more frequently, suggesting that they readily suppressed IFN-γ-producing cells. The effect of EP on Treg was also reproduced in vitro. Overall, our results show that EP treatment reduced T1D incidence in C57BL/6 mice predominantly by enhancing Treg differentiation, proliferation, their suppressive capacity, and recruitment into the pancreas.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/metabolism , Lymphocyte Activation/immunology , Pyruvates/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adaptive Immunity/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Biomarkers , Cell Differentiation/immunology , Cell Movement/immunology , Cell Proliferation , Diabetes Mellitus, Experimental , Disease Models, Animal , Immunity, Innate , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Lymphocyte Activation/drug effects , Mice , Nitric Oxide/metabolism , Pyruvates/pharmacology , Reactive Oxygen Species/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...