Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 258
Filter
1.
J Neuroinflammation ; 18(1): 265, 2021 Nov 13.
Article in English | MEDLINE | ID: mdl-34772416

ABSTRACT

BACKGROUND: Lymphocytes have dichotomous functions in ischemic stroke. Regulatory T cells are protective, while IL-17A from innate lymphocytes promotes the infarct growth. With recent advances of T cell-subtype specific transgenic mouse models it now has become possible to study the complex interplay of T cell subpopulations in ischemic stroke. METHODS: In a murine model of experimental stroke we analyzed the effects of IL-10 on the functional outcome for up to 14 days post-ischemia and defined the source of IL-10 in ischemic brains based on immunohistochemistry, flow cytometry, and bone-marrow chimeric mice. We used neutralizing IL-17A antibodies, intrathecal IL-10 injections, and transgenic mouse models which harbor a deletion of the IL-10R on distinct T cell subpopulations to further explore the interplay between IL-10 and IL-17A pathways in the ischemic brain. RESULTS: We demonstrate that IL-10 deficient mice exhibit significantly increased infarct sizes on days 3 and 7 and enlarged brain atrophy and impaired neurological outcome on day 14 following tMCAO. In ischemic brains IL-10 producing immune cells included regulatory T cells, macrophages, and microglia. Neutralization of IL-17A following stroke reversed the worse outcome in IL-10 deficient mice and intracerebral treatment with recombinant IL-10 revealed that IL-10 controlled IL-17A positive lymphocytes in ischemic brains. Importantly, IL-10 acted differentially on αß and γδ T cells. IL-17A producing CD4+ αß T cells were directly controlled via their IL-10-receptor (IL-10R), whereas IL-10 by itself had no direct effect on the IL-17A production in γδ T cells. The control of the IL-17A production in γδ T cells depended on an intact IL10R signaling in regulatory T cells (Tregs). CONCLUSIONS: Taken together, our data indicate a key function of IL-10 in restricting the detrimental IL-17A-signaling in stroke and further supports that IL-17A is a therapeutic opportunity for stroke treatment.


Subject(s)
Interleukin-10/therapeutic use , Interleukin-17/antagonists & inhibitors , Ischemic Stroke/drug therapy , Animals , Antibodies, Neutralizing/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Immunohistochemistry , Infarction, Middle Cerebral Artery/prevention & control , Injections, Spinal , Interleukin-10/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Interleukin-10/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Treatment Outcome
2.
Theranostics ; 11(20): 9988-10000, 2021.
Article in English | MEDLINE | ID: mdl-34815799

ABSTRACT

Rationale: Tailored inflammation control is badly needed for the treatment of kinds of inflammatory diseases, such as atherosclerosis. IL-10 is a potent anti-inflammatory cytokine, while systemic and repeated delivery could cause detrimental side-effects due to immune repression. In this study, we have developed a nano-system to deliver inflammation-responsive Il-10 mRNA preferentially into macrophages for tailored inflammation control. Methods:Il-10 was engineered to harbor a modified HCV-IRES (hepatitis C virus internal ribosome entry site), in which the two miR-122 recognition sites were replaced by two miR-155 recognition sites. The translational responsiveness of the engineered mRNA to miR-155 was tested by Western blot or ELISA. Moreover, the engineered Il-10 mRNA was passively encapsulated into exosomes by forced expression in donor cells. Therapeutic effects on atherosclerosis and the systemic leaky expression effects in vivo of the functionalized exosomes were analyzed in ApoE-/- (Apolipoprotein E-deficient) mice. Results: The engineered IRES-Il-10 mRNA could be translationally activated in cells when miR-155 was forced expressed or in M1 polarized macrophages with endogenous miR-155 induced. In addition, the engineered IRES-Il-10 mRNA, when encapsulated into the exosomes, could be efficiently delivered into macrophages and some other cell types in the plaque in ApoE-/- mice. In the recipient cells of the plaque, the encapsulated Il-10 mRNA was functionally translated into protein, with relatively low leaky in other tissues/organs without obvious inflammation. Consistent with the robust Il-10 induction in the plaque, exosome-based delivery of the engineered Il-10 could alleviate the atherosclerosis in ApoE-/- mice. Conclusion: Our study established a potent platform for controlled inflammation control via exosome-based systemic and repeated delivery of engineered Il-10 mRNA, which could be a promising strategy for atherosclerosis treatment.


Subject(s)
Atherosclerosis/therapy , Drug Delivery Systems/methods , Interleukin-10/pharmacology , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , China , Exosomes/immunology , Exosomes/physiology , Genetic Engineering/methods , HEK293 Cells , Humans , Inflammation/metabolism , Interleukin-10/administration & dosage , Interleukin-10/genetics , Internal Ribosome Entry Sites/genetics , Macrophage Activation , Macrophages/metabolism , Male , Mice , MicroRNAs/genetics , Plaque, Atherosclerotic/metabolism , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/pharmacology
3.
PLoS One ; 16(10): e0259114, 2021.
Article in English | MEDLINE | ID: mdl-34705865

ABSTRACT

INTRODUCTION: Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of autoantibodies such as anti-Sm. Studies in patients with SLE and murine models of lupus reveal that the most critical anti-Sm autoantibodies are predominantly direct against D1(83-119), D2, and B´/B epitopes. OBJECTIVES: The present study aimed to analyze the induction of antigen-specific tolerance after prophylactic immunization with a DNA vaccine encoding the epitopes: D183-119, D2, B´/B, and B´/BCOOH in co-vaccination with IFN-γ or IL-10 in a murine model of lupus induced by pristane. MATERIAL AND METHODS: To obtain endotoxin-free DNA vaccines, direct cloning techniques using pcDNA were performed: D183-119, D2, B´/B, B´/BCOOH, IFN-γ, or IL-10. Lupus was induced by 0.5 mL of pristane via intraperitoneal in BALB/c female mice. Immunoprecipitation with K562 cells was metabolically labeled with 35S and ELISA to detect serum antibodies or mice IgG1, IgG2a isotypes. ELISA determined IL-10 and IFN-γ from splenocytes supernatants. Proteinuria was assessed monthly, and lupus nephritis was evaluated by immunofluorescence, and electron microscopy. RESULTS: The prophylactic co-vaccination with D2/IL-10 reduced the expression of kidney damage observed by electron microscopy, direct immunofluorescence, and H & E, along with reduced level of anti-nRNP/Sm antibodies (P = 0.048). CONCLUSION: The prophylactic co-vaccination of IL-10 with D2 in pristane-induced lupus ameliorates the renal damage maybe by acting as prophylactic DNA tolerizing therapy.


Subject(s)
Interleukin-10 , Lupus Erythematosus, Systemic/prevention & control , Vaccines, DNA , Animals , Autoantibodies/immunology , Autoantigens/immunology , Female , Interleukin-10/administration & dosage , Interleukin-10/pharmacology , Mice , Mice, Inbred BALB C , Therapies, Investigational , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/pharmacology
4.
Front Immunol ; 12: 667897, 2021.
Article in English | MEDLINE | ID: mdl-34108968

ABSTRACT

A therapy that includes an oral vaccine for type 1 diabetes (T1D) using live attenuated Salmonella MvP728 (ΔhtrA/ΔpurD), cytokines (IL10 and TGFß) and preproinsulin (PPI) antigen in combination with a sub-therapeutic dose of anti-CD3 mAb was developed by our team. The vaccine combination therapy reduced insulitis and prevented and reversed diabetes in non-obese diabetic (NOD) mice. Here, we show the effectiveness of an alternative Salmonella mutant (ΔmsbB) as a carrier strain, which is anticipated to have lower risks of an inflammatory response and septicemia as a result of modification in the lipopolysaccharide (LPS) via detoxification of lipid A. This mutant strain proved to have highly reduced pathogenic side effects. Salmonella strain ΔmsbB expressed autoantigens and in combination with cytokines and anti-CD3 mAb, successfully prevented and reversed T1D to levels comparable to the previously used carrier strain ΔhtrA/ΔpurD. Additionally, the Salmonella msbB mutant resulted in higher rates of host cell infection. These results further demonstrate the potential of an oral Salmonella-based combined therapy in the treatment of early T1D.


Subject(s)
Acyltransferases/genetics , Bacterial Proteins/genetics , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/prevention & control , Genetic Vectors , Mutation , Salmonella/genetics , Vaccines, DNA/administration & dosage , Administration, Oral , Animals , Antibodies, Monoclonal/administration & dosage , Biomarkers/blood , CD3 Complex/antagonists & inhibitors , CD3 Complex/immunology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Female , Insulin/administration & dosage , Insulin/genetics , Interleukin-10/administration & dosage , Interleukin-10/genetics , Mice , Mice, Inbred NOD , Protein Precursors/administration & dosage , Protein Precursors/genetics , RAW 264.7 Cells , Salmonella/immunology , Salmonella/pathogenicity , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/genetics , Vaccines, Attenuated/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology
5.
FASEB J ; 35(8): e21754, 2021 08.
Article in English | MEDLINE | ID: mdl-34191338

ABSTRACT

To obtain a successful pregnancy, the establishment of maternal-fetal tolerance and successful placentation are required to be established. Disruption of this immune balance and/or inadequate placental perfusion is believed to be associated with a lot of pregnancy-related complications, such as recurrent spontaneous abortion, pre-eclampsia, and fetal intrauterine growth restriction. Extravillous trophoblasts (EVTs) have the unique ability to instruct decidual immune cells (DICs) to develop a regulatory phenotype for fetal tolerance. Utilizing immortalized human first trimester extravillous trophoblast cells and primary EVTs, we found that DICs promote EVT function and placental development. We have previously shown that checkpoints T-cell immunoglobulin mucin-3 (Tim-3) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are important for DIC function. In the present study, we showed that blockade of Tim-3 and CTLA-4 pathways leaded to the abnormal DICs-EVTs interaction, poor placental development, and increased fetal loss. Treatment with IL-4 and IL-10 could rescue the adverse effects of targeting Tim-3 and CTLA-4 on the pregnancy outcome. Hence, the reproductive safety must be a criterion considered in the assessment of immuno-therapeutic agents. In addition, IL-4 and IL-10 may represent novel therapeutic strategies to prevent pregnancy loss induced by checkpoint inhibition.


Subject(s)
CTLA-4 Antigen/immunology , Decidua/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Interleukin-10/immunology , Interleukin-4/immunology , Trophoblasts/immunology , Animals , CTLA-4 Antigen/antagonists & inhibitors , Cell Communication/immunology , Cells, Cultured , Decidua/cytology , Embryo Loss/immunology , Female , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Humans , Immune Tolerance , Interleukin-10/administration & dosage , Interleukin-4/administration & dosage , Male , Maternal-Fetal Exchange/immunology , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred DBA , Models, Immunological , Placentation/immunology , Pregnancy , Pregnancy Outcome , Signal Transduction/immunology , Trophoblasts/cytology
6.
Int J Cancer ; 149(2): 403-408, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33709428

ABSTRACT

Interleukin (IL)-10 has anti-inflammatory and CD8+ T-cell-stimulating properties. Pegilodecakin (pegylated recombinant human IL-10) induces intratumoral antigen-specific CD8 + T-cells and upregulates IFNγ and major histocompatibility complexes (MHC) I and II. Pegilodecakin has single-agent activity with manageable toxicity in advanced renal cell carcinama (aRCC) (data cutoff 24 March 2016). Pegilodecakin with pembrolizumab or nivolumab revealed clinical activity in aRCC (data cutoff 1 July 2018). Here, we report for the first time the results of pegilodecakin+ pazopanib, and final results for monotherapy and long-term follow-up with pegilodecakin + anti-programmed cell death 1 (anti-PD-1) inhibitors (data cutoff 19 February 2019). Phase 1/1b multi-cohort dose escalation IVY study enrolled 353 patients. Sixty-six patients with aRCC were treated with pegilodecakin alone or with pazopanib or anti-PD-1 inhibitor in cohorts A, G, H and I (data cutoff 19 February 2019). Primary endpoints included safety and tolerability. Secondary endpoint was tumor response by immune-related response criteria (irRC). Pegilodecakin plus nivolumab or pembrolizumab yielded median progression-free survival (mPFS) of 13.9 months and 6-month PFS probability of 60%, 76% 1-year overall survival (OS) probability and 61% 2-year OS probability. Pegilodecakin monotherapy produced mPFS of 1.8 months, 6-month PFS probability 25%, 1-year OS 50%, and 2-year OS 17%. Median OS was not reached in both combinations. Objective response rates (ORRs) were 33% with pazopanib and 43% with anti-PD-1. Most common Grade 3/4 treatment-related adverse events included anemia, thrombocytopenia and hypertriglyceridemia. In these heavily pretreated renal cell carcinama cohorts of IVY, pegilodecakin+anti-PD-1 inhibitor showed promising clinical activity. Safety profile of pegilodecakin alone and with anti-PD-1 inhibitors was consistent as previously reported.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Renal Cell/drug therapy , Indazoles/administration & dosage , Interleukin-10/administration & dosage , Kidney Neoplasms/drug therapy , Polyethylene Glycols/administration & dosage , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Administration Schedule , Female , Humans , Indazoles/therapeutic use , Interleukin-10/therapeutic use , Male , Middle Aged , Nivolumab/administration & dosage , Nivolumab/therapeutic use , Polyethylene Glycols/therapeutic use , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Survival Analysis , Treatment Outcome , Young Adult
7.
J Clin Oncol ; 39(10): 1108-1118, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33555926

ABSTRACT

PURPOSE: SEQUOIA compared efficacy and safety of adding pegilodecakin (PEG), a pegylated recombinant human interleukin (IL)-10, with folinic acid, fluorouracil, and oxaliplatin (FOLFOX) in patients following progression on first-line gemcitabine-containing therapy with metastatic pancreatic ductal adenocarcinoma (PDAC). PATIENTS AND METHODS: SEQUOIA, a randomized, global phase III study, compared FOLFOX with PEG + FOLFOX as second line in gemcitabine-refractory PDAC. Patients were randomly assigned 1:1 (PEG + FOLFOX:FOLFOX) and stratified by prior gemcitabine and region. Eligible patients had only one prior gemcitabine-containing treatment. Primary end point was overall survival (OS). Secondary end points included progression-free survival (PFS), response evaluation per Response Evaluation Criteria in Solid Tumor (RECIST) 1.1, and safety. Exploratory analyses included biomarkers related to immune activation. RESULTS: Between March 1, 2017, and September 9, 2019, 567 patients were randomly assigned PEG + FOLFOX (n = 283) or FOLFOX (n = 284). Most (94.7%) patients received prior gemcitabine plus nab paclitaxel. OS was similar comparing PEG + FOLFOX versus FOLFOX (median: 5.8 v 6.3 months; hazard ratio = 1.045; 95% CI, 0.863 to 1.265). Also, PFS (median 2.1 v 2.1 months; hazard ratio = 0.981; 95% CI, 0.808 to 1.190) and objective response rate (4.6% v 5.6%) were similar between the treatment arms. Most common (≥ 35%) treatment-emergent adverse events in PEG + FOLFOX versus FOLFOX were thrombocytopenia (55% v 20%), anemia (40% v 16%), fatigue (61% v 45%), neutropenia (39% v 28%), abdominal pain (37% v 29%), nausea (45% v 41%), neuropathy (37% v 38%), and decreased appetite (35% v 31%). Exploratory analyses revealed increases in total IL-18, interferon (IFN)-γ, and granzyme B and decreases in transforming growth factor (TGF)-ß with the addition of PEG. CONCLUSION: PEG added to FOLFOX did not improve efficacy in advanced gemcitabine-refractory PDAC. Safety findings were consistent as previously observed from PEG with chemotherapy; toxicity was manageable and tolerable. Exploratory pharmacodynamic results were consistent with immunostimulatory signals of the IL-10R pathway.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Pancreatic Ductal/pathology , Deoxycytidine/therapeutic use , Disease Progression , Drug Resistance, Neoplasm , Fatigue/chemically induced , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Interleukin-10/administration & dosage , Interleukin-10/adverse effects , Kaplan-Meier Estimate , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Middle Aged , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Oxaliplatin/administration & dosage , Oxaliplatin/adverse effects , Pancreatic Neoplasms/pathology , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Thrombocytopenia/chemically induced , Gemcitabine
8.
Invest New Drugs ; 39(1): 182-192, 2021 02.
Article in English | MEDLINE | ID: mdl-32910338

ABSTRACT

Background Treatment options for pancreatic ductal adenocarcinoma (PDAC) are limited and checkpoint blockade inhibitors have been disappointing in this disease. Pegilodecakin has demonstrated single agent anti-tumor activity in immune-sensitive tumors. Phase 1 and preclinical data indicate synergy of pegilodecakin with 5-FU and platins. We assessed the safety and activity of pegilodecakin+FOLFOX in patients with PDAC. Methods IVY (NCT02009449) was an open-label phase 1b trial in the United States. Here we report on all enrolled patients from cohort C. Heavily pretreated patients were treated with pegilodecakin (self-administered subcutaneously daily at 2.5, 5, or 10 µg/kg) + 5-flurouracil/leucovorin/oxaliplatin (FOLFOX), dosed per manufacturers prescribing information, until tumor progression. Eligible patients had measurable disease per immune-related response criteria (irRC), were ≥ 18 years of age, and had ECOG performance status of 0 or 1. Patients were evaluated for primary(safety) and secondary (tumor response per irRC) endpoints. Results From 5 August 2014-12 July 2016, 39 patients enrolled in cohort C. All patients were evaluable for safety. In this advanced population, regimen had manageable toxicities with no immune-related adverse events (irAEs) greater than grade 1. The most common grade 3/4/5 TEAEs were thrombocytopenia (21[53.8%] of 39) and anemia (17[43.6%] of 39). In evaluable PDAC patients, the best overall response of pegilodecakin+FOLFOX was 3(14%) with CRs in 2(9%) patients. Conclusions Pegilodecakin+FOLFOX had an acceptable tolerability profile in PDAC, with no substantial irAEs seen, and promising efficacy with the combination yielding a 2-year OS of 24% (95% CI 10-42). These data led to the phase 3 study with pegilodecakin+FOLFOX as second-line therapy of PDAC (SEQUOIA).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Interleukin-10/therapeutic use , Pancreatic Neoplasms/drug therapy , Polyethylene Glycols/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/immunology , Dose-Response Relationship, Drug , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/immunology , Fluorouracil/therapeutic use , Humans , Interleukin-10/administration & dosage , Interleukin-10/adverse effects , Interleukin-10/immunology , Kaplan-Meier Estimate , Leucovorin/administration & dosage , Leucovorin/adverse effects , Leucovorin/immunology , Leucovorin/therapeutic use , Middle Aged , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/immunology , Organoplatinum Compounds/therapeutic use , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Progression-Free Survival , Response Evaluation Criteria in Solid Tumors , Pancreatic Neoplasms
9.
Am J Physiol Renal Physiol ; 319(3): F359-F365, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32686523

ABSTRACT

Interleukin-10 (IL-10) is an anti-inflammatory cytokine that induces nitric oxide (NO) production. IL-10 supplementation has been previously shown to lower blood pressure (BP) in male hypertensive mice, but the effect of exogenous IL-10 in hypertensive female rodents has not been studied. For the present study, we hypothesized that chronic infusion of IL-10 in hypertensive rats would lower BP concomitant with an increase in renal NO synthase (NOS) activity. Male and female spontaneously hypertensive rats (SHRs; 12 wk old) were randomized to receive IL-10 infusion by subcutaneous minipump (3.5 µg·kg-1·day-1) or serve as sham controls (n = 4-6 rats per treatment per sex). BP was measured by tail cuff before and after 2 wk of treatment. Renal T cells and IL-10 were measured by flow cytometry, and NOS activity was determined by conversion of radiolabeled arginine to radiolabeled citrulline. Female SHRs had greater IL-10+ renal cells than male SHRs and greater expression of the IL-10 receptor at baseline. BP did not change in female SHRs treated with IL-10, but BP significantly decreased following IL-10 infusion in male SHRs. Contrary to our hypothesis, NOS enzymatic activity decreased with IL-10 treatment in the renal inner medulla and cortex of both sexes. Renal regulatory T cells also decreased in both sexes after IL-10 treatment. In conclusion, despite male SHRs having less IL-10 and IL-10 receptor expression in the kidney compared with female SHRs, exogenous IL-10 selectively decreased BP only in male SHRs. Furthermore, our data suggest that exogenous IL-10-induced decreases in BP in male SHRs are not dependent on upregulating renal NOS activity.


Subject(s)
Blood Pressure/drug effects , Interleukin-10/pharmacology , Animals , Female , Gene Expression Regulation/drug effects , Inflammation/metabolism , Infusion Pumps , Infusions, Subcutaneous , Interleukin-10/administration & dosage , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Male , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Random Allocation , Rats , Rats, Inbred SHR , Receptors, Interleukin-10/genetics , Receptors, Interleukin-10/metabolism , Sex Factors , T-Lymphocytes/cytology
10.
Front Immunol ; 11: 1103, 2020.
Article in English | MEDLINE | ID: mdl-32582188

ABSTRACT

A combination treatment (CT) of proinsulin and IL-10 orally delivered via genetically modified Lactococcus lactis bacteria combined with low-dose anti-CD3 (aCD3) therapy successfully restores glucose homeostasis in newly diagnosed non-obese diabetic (NOD) mice. Tolerance is accompanied by the accumulation of Foxp3+ regulatory T cells (Tregs) in the pancreas. To test the potential of this therapy outside the window of acute diabetes diagnosis, we substituted autoimmune diabetic mice, with disease duration varying between 4 and 53 days, with syngeneic islets at the time of therapy initiation. Untreated islet recipients consistently showed disease recurrence after 8.2 ± 0.7 days, while 32% of aCD3-treated and 48% of CT-treated mice remained normoglycemic until 6 weeks after therapy initiation (P < 0.001 vs. untreated controls for both treatments, P < 0.05 CT vs. aCD3 therapy). However, mice that were diabetic for more than 2 weeks before treatment initiation were less efficient at maintaining normoglycemia than those treated within 2 weeks of diabetes diagnosis, particularly in the aCD3-treated group. The complete elimination of endogenous beta cell mass with alloxan at the time of diabetes diagnosis pointed toward the significance of continuous feeding of the islet antigen proinsulin at the time of aCD3 therapy for treatment success. The CT providing proinsulin protected 69% of mice, compared to 33% when an irrelevant antigen (ovalbumin) was combined with aCD3 therapy, or to 27% with aCD3 therapy alone. Sustained tolerance was accompanied with a reduction of IGRP+CD8+ autoreactive T cells and an increase in insulin-reactive (InsB12-20 or InsB13-2) Foxp3+CD4+ Tregs, with a specific accumulation of Foxp3+ Tregs around the insulin-containing islet grafts after CT with proinsulin. The combination of proinsulin and IL-10 via oral Lactococcus lactis with low-dose aCD3 therapy can restore tolerance to beta cells in autoimmune diabetic mice, also when therapy is started outside the window of acute diabetes diagnosis, providing persistence of insulin-containing islets or prolonged beta cell function.


Subject(s)
CD3 Complex/antagonists & inhibitors , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/drug effects , Interleukin-10/administration & dosage , Proinsulin/administration & dosage , Animals , Diabetes Mellitus, Experimental/immunology , Genetic Vectors , Humans , Lactococcus lactis , Mice , Mice, Inbred NOD , Self Tolerance/drug effects , Self Tolerance/immunology
11.
Life Sci ; 256: 117985, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32562692

ABSTRACT

AIMS: To assess the combination therapy of anti-CD20 mabs and adenovirus-mediated interleukin-10 (IL-10) gene delivery on the prevention of type 1 diabetes (T1D) in non-obese diabetes (NOD) mice. MAIN METHODS: In present study, we simultaneously blocked the B cell interactions and recovered the Th cell subset proportion by using through anti-CD20 Mab and adenovirus-mediated gene delivery of IL-10, respectively. After 9 consecutive days of combination therapy, various measurements, including hematoxylin-eosin staining (HE), terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labelling assay (TUNEL), immunohistochemistry, ELISA, PCR and western blot were applied to further assess the efficacy. KEY FINDINGS: The results suggested that the combination intervention reduced the T1D-associated morbidity of NOD mice, promote insulin secretion, control blood glucose and ease pancreatitis. Moreover, the combination therapy might play a protective role in pancreatic ß cells by suppressing the expression of TNF-α and Fas, blocking the Caspase-8 and Caspase-3 apoptotic pathways and activating the Bcl-2 anti-apoptotic pathway. Finally, the combination intervention may up-regulate the gene expression of CK-19 and PDX-1 and further accelerate the differentiation and proliferation of pancreatic ß cells. SIGNIFICANCE: Therefore, the combination intervention with anti-CD20 mabs and the IL-10 gene plays a role in the prevention of T1D to some extent in NOD mice.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, CD20/administration & dosage , Apoptosis/drug effects , Diabetes Mellitus, Type 1/drug therapy , Interleukin-10/administration & dosage , Pancreatitis/drug therapy , Adenoviridae/genetics , Animals , Antibodies, Monoclonal/genetics , Antigens, CD20/genetics , Apoptosis/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Drug Therapy, Combination , Female , Interleukin-10/genetics , Male , Mice , Mice, Inbred NOD , Pancreatitis/genetics , Pancreatitis/pathology
12.
Front Immunol ; 11: 10, 2020.
Article in English | MEDLINE | ID: mdl-32117219

ABSTRACT

The targeted delivery of therapies to diseased tissues offers a safe opportunity to achieve optimal efficacy while limiting systemic exposure. These considerations apply to many disease indications but are especially relevant for rheumatoid arthritis (RA), as RA is a systemic autoimmune disease which affects multiple joints. We have identified an antibody that is specific to damaged arthritic cartilage (anti-ROS-CII) that can be used to deliver treatments specifically to arthritic joints, yielding augmented efficacy in experimental arthritis. In the current study, we demonstrate that scaffolds enriched with bioactive payloads can be delivered precisely to an inflamed joint and achieve superior efficacy outcomes consistent with the pharmacological properties of these payloads. As a scaffold, we have used extracellular vesicles (EVs) prepared from human neutrophils (PMNs), which possess intrinsic anti-inflammatory properties and the ability to penetrate inflamed arthritic cartilage. EV fortified with anti-ROS-CII (EV/anti-ROS-CII) retained anti-ROS-CII specificity and bound exclusively to the damaged cartilage. Following systemic administration, EV/anti-ROS-CII (a) exhibited the ability to localize specifically in the arthritic joint in vivo and (b) was able to specifically target single (viral IL-10 or anti-TNF) or combined (viral IL-10 and anti-TNF) anti-inflammatory treatments to the arthritic joint, which accelerated attenuation of clinical and synovial inflammation. Overall, this study demonstrates the attainability of targeting a pro-resolving biological scaffold to the arthritic joint. The potential of targeting scaffolds such as EV, nanoparticles, or a combination thereof alongside combined therapeutics is paramount for designing systemically administered broad-spectrum of anti-inflammatory treatments.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Cartilage/immunology , Cartilage/pathology , Drug Delivery Systems/methods , Extracellular Vesicles , Animals , Female , Healthy Volunteers , Humans , Interleukin-10/administration & dosage , Knee Joint/drug effects , Leukocytes/cytology , Mice , Mice, Inbred C57BL , Treatment Outcome , Tumor Necrosis Factor-alpha/immunology , Viral Proteins/administration & dosage
13.
Cancer Prev Res (Phila) ; 13(1): 25-40, 2020 01.
Article in English | MEDLINE | ID: mdl-31771941

ABSTRACT

Tightly regulated immune responses must occur in the intestine to avoid unwanted inflammation, which may cause chronic sequela leading to diseases such as colorectal cancer. Toll-like receptors play an important role in preventing aberrant immune responses in the intestine by sensing endogenous commensal microbiota and delivering important regulatory signals to the tissue. However, the role that specific innate receptors may play in the development of chronic inflammation and their impact on the composition of the colonic microbiota is not well understood. Using a model of inflammation-induced colorectal cancer, we found that Lactobacillus species are lost more quickly in wild-type (WT) mice than TLR6-deficient mice resulting in overall differences in bacterial composition. Despite the longer retention of Lactobacillus, the TLR6-deficient mice presented with more tumors and a worse overall outcome. Restoration of the lost Lactobacillus species suppressed inflammation, reduced tumor number, and prevented change in the abundance of Proteobacteria only when given to WT mice, indicating the effect of these Lactobacillus are TLR6 dependent. We found that the TLR6-dependent effects of Lactobacillus could be dissociated from one another via the involvement of IL10, which was necessary to dampen the inflammatory microenvironment, but had no effect on bacterial composition. Altogether, these data suggest that innate immune signals can shape the composition of the microbiota under chronic inflammatory conditions, bias the cytokine milieu of the tissue microenvironment, and influence the response to microbiota-associated therapies.


Subject(s)
Colitis/immunology , Colorectal Neoplasms/immunology , Gastrointestinal Microbiome/immunology , Neoplasms, Experimental/immunology , Toll-Like Receptor 6/deficiency , Animals , Azoxymethane/toxicity , Colitis/chemically induced , Colitis/genetics , Colitis/microbiology , Colon/immunology , Colon/microbiology , Colon/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/microbiology , Dextran Sulfate/toxicity , Female , Humans , Immunity, Innate , Interleukin-10/administration & dosage , Interleukin-10/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lactobacillus/immunology , Mice , Mice, Knockout , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/microbiology , Probiotics/administration & dosage , Proteobacteria/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 6/genetics
14.
Lancet Oncol ; 20(11): 1544-1555, 2019 11.
Article in English | MEDLINE | ID: mdl-31563517

ABSTRACT

BACKGROUND: IL-10 has anti-inflammatory and CD8+ T-cell stimulating activities. Pegilodecakin (pegylated IL-10) is a first-in-class, long-acting IL-10 receptor agonist that induces oligoclonal T-cell expansion and has single-agent activity in advanced solid tumours. We assessed the safety and activity of pegilodecakin with anti-PD-1 monoclonal antibody inhibitors in patients with advanced solid tumours. METHODS: We did a multicentre, multicohort, open-label, phase 1b trial (IVY) at 12 cancer research centres in the USA. Patients were assigned sequentially into cohorts. Here, we report on all enrolled patients from two cohorts treated with pegilodecakin combined with anti-PD-1 inhibitors. Eligible patients were aged at least 18 years with histologically or cytologically confirmed advanced malignant solid tumours refractory to previous therapies, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients with uncontrolled infectious diseases were excluded. Pegilodecakin was provided in single-use 3 mL vials and was self-administered subcutaneously by injection at home at 10 µg/kg or 20 µg/kg once per day in combination with pembrolizumab (2 mg/kg every 3 weeks or 200 mg every 3 weeks) or nivolumab (3 mg/kg every 2 weeks or 240 mg every 2 weeks or 480 mg every 4 weeks at the approved dosing), both of which were given intravenously at the study site. Patients received pembrolizumab or nivolumab with pegilodecakin until disease progression, toxicity necessitating treatment discontinuation, patient withdrawal of consent, or study end. The primary endpoints were safety and tolerability, assessed in all patients enrolled in the study who received any amount of study medication including at least one dose of pegilodecakin, and pharmacokinetics (previously published). Secondary endpoints included objective response by immune-related response criteria in all patients who were treated and had evaluable measurements. The study is active but no longer recruiting, and is registered with ClinicalTrials.gov, NCT02009449. FINDINGS: Between Feb 13, 2015, and Sept 12, 2017, 111 patients were enrolled in the two cohorts. 53 received pegilodecakin plus pembrolizumab, and 58 received pegilodecakin plus nivolumab. 34 (31%) of 111 patients had non-small-cell lung cancer, 37 (33%) had melanoma, and 38 (34%) had renal cell carcinoma; one (<1%) patient had triple-negative breast cancer and one (<1%) had bladder cancer. Data cutoff was July 1, 2018. Median follow-up was 26·9 months (IQR 22·3-31·5) for patients with non-small-cell lung cancer, 33·0 months (29·2-35·1) for those with melanoma, and 22·7 months (20·9-27·0) for those with renal cell carcinoma. At least one treatment-related adverse event occurred in 103 (93%) of 111 patients. Grade 3 or 4 events occurred in 73 (66%) of 111 patients (35 [66%] of 53 in the pembrolizumab group and 38 [66%] of 58 in the nivolumab group), the most common of which were anaemia (12 [23%] in the pembrolizumab group and 16 [28%] in the nivolumab group), thrombocytopenia (14 [26%] in the pembrolizumab group and 12 [21%] in the nivolumab group), fatigue (11 [21%] in the pembrolizumab group and 6 [10%] in the nivolumab group) and hypertriglyceridaemia (three [6%] in the pembrolizumab group and eight [14%] in the nivolumab group). There were no fatal adverse events determined to be related to the study treatments. Of the patients evaluable for response, objective responses were 12 (43%) of 28 (non-small-cell lung cancer), three (10%) of 31 (melanoma), and 14 (40%) of 35 (renal cell carcinoma). INTERPRETATION: In this patient population, pegilodecakin with anti-PD-1 monoclonal antibodies had a manageable toxicity profile and preliminary antitumour activity. Pegilodecakin with pembrolizumab or nivolumab could provide a new therapeutic opportunity for previously treated patients with renal cell carcinoma and non-small-cell carcinoma. FUNDING: ARMO BioSciences, a wholly owned subsidiary of Eli Lilly and Company.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Interleukin-10/administration & dosage , Neoplasms/drug therapy , Nivolumab/administration & dosage , Polyethylene Glycols/administration & dosage , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Female , Humans , Interleukin-10/adverse effects , Interleukin-10/pharmacokinetics , Male , Middle Aged , Neoplasms/immunology , Neoplasms/pathology , Nivolumab/adverse effects , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacokinetics , Programmed Cell Death 1 Receptor/immunology , United States
15.
Proc Natl Acad Sci U S A ; 116(26): 13036-13041, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31189602

ABSTRACT

Human cytomegalovirus (HCMV) causes severe disease in infants and immunocompromised people. There is no approved HCMV vaccine, and vaccine development strategies are complicated by evidence of both persistent infection and reinfection of people with prior immunity. The greatest emphasis has been placed on reducing transmission to seronegative pregnant women to prevent vertical transmission and its potentially severe sequelae. Increasing evidence suggests that the earliest host-HCMV interactions establish conditions for viral persistence, including evasion of host immune responses to the virus. Using a nonhuman primate model of HCMV infection, we show that rhesus macaques immunized against viral interleukin-10 (IL-10) manifest delayed rhesus cytomegalovirus (RhCMV) acquisition and altered immune responses to the infection when it does occur. Among animals with the greatest antiviral IL-10-neutralizing activity, the timing of RhCMV seroconversion was delayed by an average of 12 weeks. After acquisition, such animals displayed an antibody response to the new infection, which peaked as expected after 2 weeks but then declined rapidly. In contrast, surprisingly, vaccination with glycoprotein B (gB) protein had no discernible impact on these outcomes. Our results demonstrate that viral IL-10 is a key regulator of successful host immune responses to RhCMV. Viral IL-10 is, therefore, an important target for vaccine strategies against cytomegalovirus (CMV). Furthermore, given the immunoregulatory function of viral IL-10, targeting this protein may prove synergistic with other vaccine therapies and targets. Our study also provides additional evidence that the earliest host-CMV interactions can have a significant impact on the nature of persistent infection.


Subject(s)
Antigens, Viral/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/pharmacology , Cytomegalovirus/immunology , Infectious Disease Transmission, Vertical/prevention & control , Interleukin-10/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, Viral/administration & dosage , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/transmission , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/immunology , Cytomegalovirus Vaccines/therapeutic use , Disease Models, Animal , Female , Host-Pathogen Interactions/immunology , Humans , Immunity, Mucosal , Immunogenicity, Vaccine , Interleukin-10/administration & dosage , Macaca mulatta , Pregnancy , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Viral Envelope Proteins/administration & dosage , Virus Shedding/immunology
16.
J Neuroinflammation ; 16(1): 93, 2019 Apr 30.
Article in English | MEDLINE | ID: mdl-31039819

ABSTRACT

BACKGROUND: The anti-inflammatory cytokine interleukin-10 (IL-10) has been explored previously as a treatment method for spinal cord injury (SCI) due to its ability to attenuate pro-inflammatory cytokines and reduce apoptosis. Primary limitations when using systemic injections of IL-10 are that it is rapidly cleared from the injury site and that it does not cross the blood-spinal cord barrier. OBJECTIVE: Here, mineral-coated microparticles (MCMs) were used to obtain a local sustained delivery of IL-10 directly into the injury site after SCI. METHODS: Female Sprague-Dawley rats were contused at T10 and treated with either an intraperitoneal injection of IL-10, an intramedullary injection of IL-10, or MCMs bound with IL-10 (MCMs+IL-10). After treatment, cytokine levels were measured in the spinal cord, functional testing and electrophysiology were performed, axon tracers were injected into the brainstem and motor cortex, macrophage levels were counted using flow cytometry and immunohistochemistry, and lesion size was measured. RESULTS: When treated with MCMs+IL-10, IL-10 was significantly elevated in the injury site and inflammatory cytokines were significantly suppressed, prompting significantly less cells expressing antigens characteristic of inflammatory macrophages and significantly more cells expressing antigens characteristic of earlier stage anti-inflammatory macrophages. Significantly more axons were preserved within the rubrospinal and reticulospinal tracts through the injury site when treated with MCMs+IL-10; however, there was no significant difference in corticospinal tract axons preserved, regardless of treatment group. The rats treated with MCMs+IL-10 were the only group with a significantly higher functional score compared to injured controls 28 days post-contusion. CONCLUSION: These data demonstrate that MCMs can effectively deliver biologically active IL-10 for an extended period of time altering macrophage phenotype and aiding in functional recovery after SCI.


Subject(s)
Inflammation/pathology , Interleukin-10/administration & dosage , Neuroprotective Agents/administration & dosage , Spinal Cord Injuries/pathology , Animals , Dosage Forms , Female , Random Allocation , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects
17.
Plast Reconstr Surg ; 143(6): 1215e-1223e, 2019 06.
Article in English | MEDLINE | ID: mdl-31136482

ABSTRACT

BACKGROUND: Large calvarial defects represent a major reconstructive challenge, as they do not heal spontaneously. Infection causes inflammation and scarring, further reducing the healing capacity of the calvaria. Bone morphogenetic protein-2 (BMP2) has been shown to stimulate osteogenesis but has significant side effects in high doses. BMP2 has not been tested in combination with antiinflammatory cytokines such as interleukin-10. METHODS: Sixteen New Zealand White rabbits underwent 15 × 15-mm flap calvarectomies. The flap was incubated in Staphylococcus aureus and replaced, and infection and scarring were allowed to develop. The flap was subsequently removed and the wound débrided. A 15 × 15-mm square of acellular dermal matrix biopatterned with low-dose BMP2, interleukin-10, or a combination was implanted. Computed tomographic scans were taken over 42 days. Rabbits were then killed and histology was performed. RESULTS: Defects treated with BMP2 showed significantly (p < 0.05) greater osseous regeneration than untreated controls. Interleukin-10 did not significantly augment the healing achieved with BMP2, and interleukin-10 alone did not significantly increase healing compared with controls. Histology showed evidence of bone formation in defects treated with BMP2. Untreated controls and defects treated with interleukin-10 alone showed only fibrous tissue in the defect site. CONCLUSIONS: Low-dose BMP2 delivered directly to the scarred calvarial defect augments bony healing. Interleukin-10 at the dose applied did not significantly augment healing alone or in combination with BMP2. Healing had not finished at 42 days and analysis at later time points or the use of higher doses of BMP2 may yield greater healing.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Bone Regeneration/drug effects , Interleukin-10/pharmacology , Skull/physiology , Animals , Bone Morphogenetic Protein 2/administration & dosage , Cicatrix/drug therapy , Drug Combinations , Interleukin-10/administration & dosage , Male , Rabbits , Skull/drug effects , Skull/surgery , Staphylococcal Infections/physiopathology , Staphylococcus aureus , Surgical Flaps , Tomography, X-Ray Computed
18.
Biomaterials ; 203: 52-62, 2019 05.
Article in English | MEDLINE | ID: mdl-30852423

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a life-threatening progressive lung disorder with limited therapeutic options. While interleukin-10 (IL-10) is a potent anti-inflammatory and anti-fibrotic cytokine, its utility in treating lung fibrosis has been limited by its short half-life. We describe an innovative hydrogel-based approach to deliver recombinant IL-10 to the lung for the prevention and reversal of pulmonary fibrosis in a mouse model of bleomycin-induced lung injury. Our studies show that a hyaluronan and heparin-based hydrogel system locally delivers IL-10 by capitalizing on the ability of heparin to reversibly bind IL-10 without bleeding or other complications. This formulation is significantly more effective than soluble IL-10 for both preventing and reducing collagen deposition in the lung parenchyma after 7 days of intratracheal administration. The anti-fibrotic effect of IL-10 in this system is dependent on suppression of TGF-ß driven collagen production by lung fibroblasts and myofibroblasts. We conclude that hydrogel-based delivery of IL-10 to the lung is a promising therapy for fibrotic lung disorders.


Subject(s)
Bleomycin/toxicity , Hydrogels/chemistry , Interleukin-10/administration & dosage , Interleukin-10/therapeutic use , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Hyaluronic Acid/chemistry , Immunohistochemistry , Mice
19.
Nat Microbiol ; 4(3): 429-437, 2019 03.
Article in English | MEDLINE | ID: mdl-30617349

ABSTRACT

Severe fever with thrombocytopenia syndrome phlebovirus (SFTSV), listed in the World Health Organization Prioritized Pathogens, is an emerging phlebovirus with a high fatality1-4. Owing to the lack of therapies and vaccines5,6, there is a pressing need to understand SFTSV pathogenesis. SFSTV non-structural protein (NSs) has been shown to block type I interferon induction7-11 and facilitate disease progression12,13. Here, we report that SFTSV-NSs targets the tumour progression locus 2 (TPL2)-A20-binding inhibitor of NF-κB activation 2 (ABIN2)-p105 complex to induce the expression of interleukin-10 (IL-10) for viral pathogenesis. Using a combination of reverse genetics, a TPL2 kinase inhibitor and Tpl2-/- mice showed that NSs interacted with ABIN2 and promoted TPL2 complex formation and signalling activity, resulting in the marked upregulation of Il10 expression. Whereas SFTSV infection of wild-type mice led to rapid weight loss and death, Tpl2-/- mice or Il10-/- mice survived an infection. Furthermore, SFTSV-NSs P102A and SFTSV-NSs K211R that lost the ability to induce TPL2 signalling and IL-10 production showed drastically reduced pathogenesis. Remarkably, the exogenous administration of recombinant IL-10 effectively rescued the attenuated pathogenic activity of SFTSV-NSs P102A, resulting in a lethal infection. Our study demonstrates that SFTSV-NSs targets the TPL2 signalling pathway to induce immune-suppressive IL-10 cytokine production as a means to dampen the host defence and promote viral pathogenesis.


Subject(s)
Host-Pathogen Interactions , MAP Kinase Kinase Kinases/metabolism , Phlebovirus/pathogenicity , Proto-Oncogene Proteins/metabolism , Signal Transduction , Viral Nonstructural Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Bunyaviridae Infections/immunology , Bunyaviridae Infections/pathology , Female , HEK293 Cells , HeLa Cells , Humans , Interleukin-10/administration & dosage , Interleukin-10/genetics , MAP Kinase Kinase Kinases/immunology , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phlebovirus/drug effects , Proto-Oncogene Proteins/immunology , RAW 264.7 Cells , Reverse Genetics
20.
Fundam Clin Pharmacol ; 33(1): 31-40, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30144156

ABSTRACT

The activation of extracellular signal-regulated kinase 1 and 2 (ERK 1/2) pathway promotes increased vascular contractility in angiotensin II (Ang II)-induced hypertensive mice. Interleukin-10 (IL-10) is an immune-regulatory cytokine with the ability to prevent vascular hypercontractility during hypertension. We hypothesized that IL-10 would downregulate vascular ERK 1/2 activation during Ang II-induced hypertension. Wild-type (WT) or IL-10 knockout (IL-10-/- ) mice received Ang II infusion (90 ηg.min) or vehicle (saline), via osmotic mini-pumps (0.25 µL/h for 14 days), whereas another WT group were infused with exogenous IL-10 (0.5 ηg/min, 14 days) simultaneously, or not, with Ang II. Aortic rings were mounted in a myograph, and concentration-response curves to phenylephrine were evaluated, in the presence or absence of ERK 1/2 inhibitor (PD98059, 10 µm, 40 min). Protein expression of vascular ERK 1/2 was determined by Western blot. Ang II infusion increased the maximal contractile response in both WT and IL-10-/- mice. Concomitant infusion of IL-10 and Ang II prevented hypercontractility in the vasculature. Exogenous IL-10 infusion prevented ERK 1/2 activation and hypercontractility, induced by Ang II. These findings suggest that IL-10 negatively modulates ERK 1/2 activation and prevents hypercontractility during Ang II-induced hypertension.


Subject(s)
Angiotensin II/administration & dosage , Aorta/metabolism , Hypertension/physiopathology , Interleukin-10/metabolism , Animals , Blotting, Western , Disease Models, Animal , Flavonoids/pharmacology , Interleukin-10/administration & dosage , Interleukin-10/genetics , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phenylephrine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL