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










Publication year range
1.
Aging (Albany NY) ; 12(1): 193-203, 2020 01 04.
Article in English | MEDLINE | ID: mdl-31901899

ABSTRACT

Our previous studies have demonstrated that interleukin-12p35 knockout (IL-12p35 KO) regulates the progression of various cardiovascular diseases, such as acute cardiac injury and hypertension. The aims of this study were to investigate whether IL-12p35 KO affects aging-related cardiac remodeling and to explore the possible mechanisms. First, the effects of IL-12p35 KO on heart structure and function were detected, and the results showed that IL-12p35 KO exacerbated cardiac remodeling and increased cardiac senescence-related protein levels in aged mice. In addition, whether IL-12p35 KO regulates cardiac senescence-related protein expression, cardiac mitochondrial dysfunction and cardiomyocyte apoptosis was also investigated. IL-12p35 KO increased mitochondrial calcium fluorescence intensity and ROS fluorescence intensity, while it reduced mitochondrial membrane potential. Furthermore, reduced mitochondrial complex (I-IV) activity and ATP levels and increased apoptosis-inducing factor (AIF)-related cardiomyocyte apoptosis were observed in aged IL-12p35 KO mice compared with wild-type mice. Our results demonstrate that aging is aggravated by IL-12p35 KO and that the mechanism may be related to exacerbation of mitochondrial dysfunction and AIF-related cardiomyocyte apoptosis.


Subject(s)
Aging/genetics , Interleukin-12 Subunit p35/deficiency , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Ventricular Remodeling/genetics , Age Factors , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cellular Senescence/genetics , Mice , Mice, Knockout , Mitochondria , Myocytes, Cardiac/metabolism
2.
Mediators Inflamm ; 2019: 3152040, 2019.
Article in English | MEDLINE | ID: mdl-31093011

ABSTRACT

Interleukin- (IL-) 35, a novel functional cytokine of regulatory T cells (Treg) comprised of the IL-12p35 subunit and the other subunit Epstein-Barr virus-induced gene 3 (EBI3), regulates the activity of CD4+ T cells and macrophages, thereby playing a critical role in inflammatory and autoimmune diseases. Previous studies demonstrated that both recombinant mice and human IL-35 attenuated atherosclerosis in ApoE-/- mice. Additionally, EBI3 deficiency enhanced the activation of macrophages and increased atherosclerotic lesions in LDLR-/- mice. This study generated double-deficient mice for ApoE and IL-12p35 (ApoE-/- IL-12p35-/- mice) and investigated the effect of IL-12p35 deficiency on atherosclerosis. IL-12p35 deficiency alleviated Th1/Th2 imbalance, aggravated Th17/Treg imbalance, and attenuated atherosclerotic plaque formation in ApoE-/- mice. Additionally, exogenous rIL-35 treatment reversed the imbalance of Th17/Treg and attenuated atherosclerosis in ApoE-/- mice. These findings suggest that IL-12p35 deficiency ameliorates atherosclerosis in ApoE-/- mice, partially, via attenuating the Th1/Th2 imbalance, although IL-12p35 deficiency aggravates the Th17/Treg imbalance.


Subject(s)
Interleukin-12 Subunit p35/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism , Animals , Apolipoproteins E , Atherosclerosis/immunology , Atherosclerosis/metabolism , Immunohistochemistry , Interleukin-10/metabolism , Interleukin-12 Subunit p35/deficiency , Interleukin-4/metabolism , Male , Mice , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism
3.
Cardiovasc Res ; 115(6): 1102-1113, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30395167

ABSTRACT

AIMS: Numerous studies have demonstrated that inflammation is involved in the progression of hypertension. Inflammatory cytokines interleukin (IL)-12 and IL-35 belong to the IL-12 cytokine family and share the same IL-12p35 subunit. Accumulating evidence has demonstrated that IL-12p35 knockout (IL-12p35 KO) leads to cardiovascular disease by regulating the inflammatory response. This study aimed to investigate whether IL-12p35 KO elevates blood pressure in a hypertension mouse model. METHODS AND RESULTS: Mice with angiotensin (Ang) II infusion showed marked aortic IL-12p35 expression; thus, aortic macrophages may be the main source of IL-12p35. Wild-type and IL-12p35 KO mice were infused with Ang II or saline. IL-12p35 KO promoted M1 macrophage differentiation, amplified the inflammatory response, aggravated vascular dysfunction, and elevated blood pressure in Ang II-treated mice. Then, some Ang II-infused mice were given phosphate buffer saline, mouse recombinant IL-12 (rIL-12), or rIL-35, and the results showed that rIL-12 but not rIL-35 treatment had an antihypertensive effect on Ang II-infused mice. In addition, detection of human plasma IL-12 levels in hypertensive patients and control subjects showed that IL-12 was significantly increased in hypertensive patients when compared with control subjects. In hypertensive patients, IL-12 levels were positively correlated with blood pressure. CONCLUSION: IL-12p35 KO amplifies the inflammatory response and promotes blood pressure elevation in Ang II-treated mice. In addition, IL-12, but not IL-35, plays a protective role in the Ang II-induced hypertension model. Thus, IL-12 may be a novel therapeutic agent for the prevention and treatment of clinical hypertension.


Subject(s)
Angiotensin II , Aorta/metabolism , Blood Pressure , Cell Differentiation , Hypertension/metabolism , Interleukin-12 Subunit p35/deficiency , Macrophages/metabolism , Adult , Aged , Animals , Antihypertensive Agents/administration & dosage , Aorta/pathology , Aorta/physiopathology , Blood Pressure/drug effects , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Female , Humans , Hypertension/chemically induced , Hypertension/drug therapy , Hypertension/physiopathology , Interleukin-12/administration & dosage , Interleukin-12 Subunit p35/blood , Interleukin-12 Subunit p35/genetics , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phenotype , Signal Transduction , Vasoconstriction , Vasodilation
4.
EBioMedicine ; 35: 29-39, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30228093

ABSTRACT

BACKGROUND: Recent evidence has demonstrated that interleukin 12p35 knockout (IL-12p35 KO) is involved in cardiac diseases by regulating the inflammatory response. The involvement of inflammatory cells has also been observed in doxorubicin (DOX)-induced cardiac injury. This study aimed to investigate whether IL-12p35 KO affects DOX-induced cardiac injury and the underlying mechanisms. METHODS: First, the effect of DOX treatment on cardiac IL-12p35 expression was assessed. In addition, to investigate the effect of IL-12p35 KO on DOX-induced cardiac injury, IL-12p35 KO mice were treated with DOX. Because IL-12p35 is the mutual subunit of IL-12 and IL-35, to determine the cytokine that mediates the effect of IL-12p35 KO on DOX-induced cardiac injury, mice were given phosphate-buffered saline (PBS), mouse recombinant IL-12 (rIL-12) or rIL-35 before treatment with DOX. RESULTS: DOX treatment significantly increased the level of cardiac IL-12p35 expression. In addition, IL-12p35 KO mice exhibited higher serum and heart lactate dehydrogenase levels, higher serum and heart creatine kinase myocardial bound levels, and greater cardiac dysfunction than DOX-treated mice. Furthermore, IL-12p35 KO further increased M1 macrophage and decreased M2 macrophage differentiation, aggravated the imbalance of oxidants and antioxidants, and further activated the mitochondrial apoptotic pathway and endoplasmic reticulum stress autophagy pathway. Both rIL-12 and rIL-35 protected against DOX-induced cardiac injury by alleviating the inflammatory response, oxidative stress, apoptosis and autophagy. CONCLUSIONS: IL-12p35 KO aggravated DOX-induced cardiac injury by amplifying the levels of inflammation, oxidative stress, apoptosis and autophagy. (234 words).


Subject(s)
Apoptosis , Autophagy , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/physiopathology , Doxorubicin/adverse effects , Inflammation/pathology , Interleukin-12 Subunit p35/deficiency , Oxidative Stress , Animals , Apoptosis/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy/drug effects , Biomarkers/metabolism , Cardiotonic Agents/metabolism , Cardiovascular Diseases/diagnostic imaging , Cardiovascular Diseases/pathology , Endoplasmic Reticulum Stress/drug effects , Hemodynamics/drug effects , Interleukin-12 Subunit p35/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Myocardium/ultrastructure , Oxidative Stress/drug effects , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism
5.
Cardiovasc Res ; 109(2): 249-59, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26614777

ABSTRACT

AIMS: IL-12p35 is a pro-inflammatory cytokine that participates in a variety of inflammatory diseases. This study aimed to determine whether IL-12 regulates cardiac injury and repair following acute myocardial infarction (AMI) and investigate the underlying mechanisms. METHODS AND RESULTS: Mice with AMI showed a marked increase in IL-12p35 expression of ischaemic cardiac tissues. IL-12 was mainly produced by CD11b(+) monocytes. Cardiac functions were significantly improved in IL-12p35 knockout (p35-KO) mice compared with wild-type (WT) littermates in response to AMI. IL-12p35 deficiency attenuated the infarct scar and hypertrophy compared with WT mice. RNA transcriptome sequencing and quantitative RT-PCR analysis of CD11b(+) monocytes isolated from WT and p35-KO ischaemic hearts revealed a distinct transcriptional profile in p35-KO CD11b(+) monocytes, displaying pro-angiogenesis and anti-inflammation properties. Angiogenesis was enhanced in p35-KO mice with AMI and hindlimb ischaemia. Moreover, tube formation assay and Matrigel plug analysis demonstrated that IL-12 inhibition of angiogenesis was dependent on monocytes. IL-12p35 deficiency inhibited inflammation by reducing chemokine production and monocyte infiltration into the heart. Finally, administration of an IL-12p35-neutralizing antibody limited AMI-induced inflammatory cell infiltration into the heart and improved angiogenesis and cardiac function. CONCLUSIONS: Deficiency of IL-12p35 limited AMI-induced cardiac injury by promoting pro-angiogenesis and anti-inflammatory functions of monocytes.


Subject(s)
Inflammation/metabolism , Interleukin-12 Subunit p35/metabolism , Monocytes/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Neovascularization, Pathologic/metabolism , Animals , Disease Models, Animal , Interleukin-12 Subunit p35/deficiency , Mice, Knockout , Neovascularization, Pathologic/genetics
6.
PLoS One ; 9(5): e96766, 2014.
Article in English | MEDLINE | ID: mdl-24801628

ABSTRACT

Infections with Leishmania parasites of the Leishmania Viannia subgenus give rise to both localized cutaneous (CL), and metastatic leishmaniasis. Metastasizing disease forms including disseminated (DCL) and mutocutaneous (MCL) leishmaniasis result from parasitic dissemination and lesion formation at sites distal to infection and have increased inflammatory responses. The presence of Leishmania RNA virus (LRV) in L. guyanensis parasites contributes to the exacerbation of disease and impacts inflammatory responses via activation of TLR3 by the viral dsRNA. In this study we investigated other innate immune response adaptor protein modulators and demonstrated that both MyD88 and TLR9 played a crucial role in the development of Th1-dependent healing responses against L. guyanensis parasites regardless of their LRV status. The absence of MyD88- or TLR9-dependent signaling pathways resulted in increased Th2 associated cytokines (IL-4 and IL-13), which was correlated with low transcript levels of IL-12p40. The reliance of IL-12 was further confirmed in IL12AB-/- mice, which were completely susceptible to infection. Protection to L. guyanensis infection driven by MyD88- and TLR9-dependent immune responses arises independently to those induced due to high LRV burden within the parasites.


Subject(s)
Immunity, Innate , Leishmania guyanensis/virology , Myeloid Differentiation Factor 88/metabolism , RNA Viruses/physiology , Toll-Like Receptor 9/metabolism , Animals , Disease Susceptibility , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/metabolism , Interleukin-12 Subunit p40/deficiency , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Leishmania guyanensis/physiology , Leishmaniasis, Mucocutaneous/immunology , Leishmaniasis, Mucocutaneous/pathology , Leishmaniasis, Mucocutaneous/veterinary , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Signal Transduction , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics
7.
PLoS One ; 8(9): e74225, 2013.
Article in English | MEDLINE | ID: mdl-24040208

ABSTRACT

Primary biliary cirrhosis (PBC) is considered a model autoimmune disease, with the most highly directed and specific autoantibody in both murine and human autoimmunity, the anti-mitochondrial autoantibody (AMA). However, therapeutic advances in this disease have lagged behind. Herein we have taken advantage of our unique model of murine PBC in which mice immunized with 2-octynoic acid coupled to BSA (2OA-BSA), a compound identified by quantitative structure activity relationships (QSAR) of human AMA binding, develop an intense inflammatory cholangitis with striking similarities to humans with PBC. In particular, we have constructed several unique gene-deleted mice, including mice deleted of IL-12p40, IL-12p35, IFN-γ, IL-23p19, IL-17A, IL-17F and IL-22, immunized these animals with 2OA-BSA and followed the natural history of immunopathology to identify key pathways that might provide clues for successful therapy. Our data indicate that whereas both IL-12/Th1 and IL-23/Th17 are involved in cholangitis, it is the IL-12/Th1 signaling pathway that elicits pathology. In fact, deletion of IFN-γ prevents disease and suppresses autoantibodies. Importantly, deletion of the Th17 cytokines IL-17A and IL-22, but not IL-17F, reduces biliary damage; IL-17A-knockout mice have reduced levels of anti-mitochondrial antibody. We further demonstrate that the production of IFN-γ is significantly decreased in the liver of IL-23p19(-/-), IL-17A(-/-) and IL-22(-/-) mice compared with controls. However, the ability of T cells to produce IFN-γ was not affected in Th17 cytokine-deficient mice. Our data indicate that a deficient Th17 pathway suppresses the accumulation of IFN-γ producing cells in liver during the early phase of cholangitis. In conclusion, whereas IFN-γ has a pivotal role in the early events involved in the pathogenesis of autoimmune cholangitis induced by 2OA-BSA, the IL-23/Th17 pathway potentiates the effects of IL-12/IFN-γ-mediated immunopathology.


Subject(s)
Autoantibodies/biosynthesis , Cholangitis/genetics , Interleukin-12 Subunit p35/immunology , Liver Cirrhosis, Biliary/genetics , Liver/metabolism , Signal Transduction/immunology , Animals , Autoimmunity , Cholangitis/chemically induced , Cholangitis/immunology , Cholangitis/pathology , Disease Models, Animal , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/immunology , Gene Expression Regulation , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/immunology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-23 Subunit p19/deficiency , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/immunology , Interleukins/deficiency , Interleukins/genetics , Interleukins/immunology , Liver/immunology , Liver/pathology , Liver Cirrhosis, Biliary/chemically induced , Liver Cirrhosis, Biliary/immunology , Liver Cirrhosis, Biliary/pathology , Mice , Mice, Knockout , Mitochondria/immunology , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/immunology , Th1-Th2 Balance , Interleukin-22
8.
J Infect Dis ; 208(5): 749-60, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23532102

ABSTRACT

BACKGROUND: Murine Staphylococcus aureus-mediated brain abscess comprises 2 major phases, an initial phase of cerebritis, followed by a healing phase characterized by capsule formation. METHODS: C57BL/6 wild-type (WT) and IL-12p35(-/-) mice were intracerebrally infected with S. aureus to induce brain abscesses. Clinical disease activity and bacterial load were monitored. The cell populations that were involved, as well as their specific mediators, were analyzed by immunohistochemistry, quantitative real-time polymerase chain reaction, and flow cytometry. RESULTS: In the acute phase, IL-12p35(-/-) mice were protected from disease. This was associated with enhanced recruitment of granulocytes, accompanied by upregulated expression of Il17a, Csf2 (which encodes granulocyte-macrophage colony-stimulating factor), Cxcl1, and Cxcl5, as well as increased expression of proinflammatory mediators, including Nos2 (which encodes inducible nitric oxide synthase), Ptgs2 (which encodes cyclooxygenase 2), and Tnf, that were primarily produced by granulocytes and activated microglia/macrophages. Furthermore, mechanisms associated with beneficial wound healing, including an accelerated formation of a fibrous capsule, were demonstrated by prominent VEGF-A production and collagen deposition driven by an earlier onset of T-helper 2 immunity in the absence of interleukin 12 (IL-12). CONCLUSIONS: Brain abscess development is orchestrated by IL-12 at different stages of disease. Our data indicate that IL-12 has a nonprotective role in the acute phase and that IL-12 deficiency results in the accelerated formation of a protective capsule during the healing phase, which we consider crucial for early recovery from disease.


Subject(s)
Brain Abscess/immunology , Brain Abscess/pathology , Interleukin-12 Subunit p35/deficiency , Staphylococcal Infections/immunology , Staphylococcal Infections/pathology , Staphylococcus aureus/immunology , Animals , Bacterial Load , Brain Abscess/microbiology , Disease Models, Animal , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Staphylococcal Infections/microbiology
9.
Hepatology ; 57(2): 806-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22576253

ABSTRACT

Mice with a dominant-negative transforming growth factor ß receptor restricted to T cells (dnTGFßRII mice) develop an inflammatory biliary ductular disease that strongly resembles human primary biliary cirrhosis (PBC). Furthermore, deletion of the gene encoding interleukin (IL)-12p40 resulted in a strain (IL-12p40(-/-) dnTGFßRII) with dramatically reduced autoimmune cholangitis. To further investigate the role of the IL-12 cytokine family in dnTGFßRII autoimmune biliary disease, we deleted the gene encoding the IL-12p35 subunit from dnTGFßRII mice, resulting in an IL-12p35(-/-) dnTGFßRII strain which is deficient in two members of the IL-12 family, IL-12 and IL-35. In contrast to IL-12p40(-/-) mice, the IL-12p35(-/-) mice developed liver inflammation and bile duct damage with similar severity but delayed onset as the parental dnTGFßRII mice. The p35(-/-) mice also demonstrated a distinct cytokine profile characterized by a shift from a T-helper 1 (Th1) to a Th17 response. Strikingly, liver fibrosis was frequently observed in IL-12p35(-/-) mice. In conclusion, IL-12p35(-/-) dnTGFßRII mice, histologically and immunologically, reflect key features of PBC, providing a useful generic model to understand the immunopathology of human PBC.


Subject(s)
Interleukin-12 Subunit p35/genetics , Liver Cirrhosis/pathology , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Animals , Disease Models, Animal , Hepatitis, Animal/etiology , Hepatitis, Animal/pathology , Interleukin-12/deficiency , Interleukin-12/physiology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p40/genetics , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/pathology , Mice , Receptor, Transforming Growth Factor-beta Type II , Th1 Cells/physiology , Th17 Cells/physiology
10.
J Immunol ; 188(4): 1905-14, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22238458

ABSTRACT

A Th1 response is required for the development of Plasmodium berghei ANKA (PbA)-induced experimental cerebral malaria (ECM). The role of pro-Th1 IL-12 in malaria is complex and controversial. In this study, we addressed the role of IL-12Rß2 in ECM development. C57BL/6 mice deficient for IL-12Rß2, IL-12p40, or IL-12p35 were analyzed for ECM development after blood-stage PbA infection in terms of ischemia and blood flow by noninvasive magnetic resonance imaging and angiography, T cell recruitment, and gene expression. Without IL-12Rß2, no neurologic sign of ECM developed upon PbA infection. Although wild-type mice developed distinct brain microvascular pathology, ECM-resistant, IL-12Rß2-deficient mice showed unaltered cerebral microcirculation and the absence of ischemia after PbA infection. In contrast, mice deficient for IL-12p40 or IL-12p35 were sensitive to ECM development. The resistance of IL-12Rß2-deficient mice to ECM correlated with reduced recruitment of activated T cells and impaired overexpression of lymphotoxin-α, TNF-α, and IFN-γ in the brain after PbA infection. Therefore, IL-12Rß2 signaling is essential for ECM development but independent from IL-12p40 and IL-12p35. We document a novel link between IL-12Rß2 and lymphotoxin-α, TNF-α, and IFN-γ expression, key cytokines for ECM pathogenesis.


Subject(s)
Interleukin-12 Receptor beta 2 Subunit/metabolism , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Th1 Cells/immunology , Animals , Brain/metabolism , Brain/microbiology , Brain/pathology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/biosynthesis , Interleukin-12 Receptor beta 2 Subunit/deficiency , Interleukin-12 Receptor beta 2 Subunit/genetics , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/immunology , Interleukin-12 Subunit p40/deficiency , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Lymphocyte Activation/immunology , Lymphotoxin-alpha/biosynthesis , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasmodium berghei/growth & development , Plasmodium berghei/pathogenicity , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/biosynthesis
11.
Gastroenterology ; 141(5): 1875-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21820391

ABSTRACT

BACKGROUND & AIMS: The soluble hematopoietin receptor Epstein-Barr virus-induced protein (EBI)-3 is an immune regulator that has been associated with the pathogenesis of inflammatory bowel disease. However, the concept that EBI3 is part of an interleukin (IL)-27 heterodimer that mediates chronic inflammatory and autoimmune diseases has been challenged by the description of IL-35, a bioactive cytokine comprising EBI3 and IL-12 p35. We investigated the roles of IL-27 and IL-35 in chronic inflammation of the intestine. METHODS: We analyzed EBI3-deficient mice and IL-27p28-deficient mice with spontaneous or T-cell transfer-induced colitis and compared outcomes with wild-type mice (controls). We constructed vectors that express EBI3 covalently linked to the IL-12p35 chain (recombinant [r]IL-35). RESULTS: Intestines of EBI3-deficient mice had increased pathologic features of colitis, compared with IL-27p28-deficient or control mice; they also had shorter survival times, indicating that IL-35, rather than IL-27, protects the intestine from immune responses in mice. The mucosa of EBI3-deficient mice accumulated subsets of activated CD4+ T cells that produced T-helper (Th)1 and Th17 cytokines. Adoptive transfer of these T cells induced colitis in RAG-deficient mice. The rIL-35 significantly reduced the development of several forms of experimental colitis and reduced levels of markers of Th1 and Th17 cells. CONCLUSIONS: IL-35 controls the development of T-cell-dependent colitis in mice. It might be developed as a therapeutic target for patients with chronic intestinal inflammation.


Subject(s)
Colitis/immunology , Colitis/metabolism , Immunity, Mucosal/physiology , Interleukins/metabolism , Intestinal Mucosa/metabolism , T-Lymphocytes/physiology , Animals , Colitis/pathology , Disease Models, Animal , Female , Immunity, Mucosal/drug effects , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Interleukins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Phenotype , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , T-Lymphocytes/pathology , Th1 Cells/pathology , Th1 Cells/physiology , Th17 Cells/pathology , Th17 Cells/physiology , Up-Regulation
12.
Infect Immun ; 79(9): 3778-83, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21746859

ABSTRACT

CD40 ligand (CD40L) transduction of antigen-pulsed dendritic cells (DCs) can result in antigen-specific humoral immune responses even in CD4(+) T-cell-depleted settings. Here, we show that CD40L transduction of DCs results in the induction of interleukin-12p40 (IL-12p40), IL-12p70, and IL-23. Using DCs that were deficient in IL-12p40, IL-12p35, or IL-23p19, we show that these molecules are dispensable for primary IgG1 responses to Pneumocystis, but IgG2c was dependent on IL-12p40 and IL-23p19 but not IL-12p35. Antigen-specific recall responses in CD4-deficient mice were critically dependent on IL-12p40 and IL-23p19 expression in DCs and were not affected by the lack of IL-12p35. To confirm that this defect in recall was due to IL-23, transduction of IL-12p40(-/-) DCs with a recombinant adenovirus expressing functional IL-23 restored recall responses in DC-vaccinated CD4-deficient mice. These data show that DC-produced IL-23 is critical for vaccine-induced antigen-specific IgG2c and recall antibody responses in the setting of CD4(+) T-cell depletion.


Subject(s)
Dendritic Cells/immunology , Interleukin-23 Subunit p19/immunology , Interleukin-23/immunology , Pneumocystis/immunology , Receptors, IgG/immunology , Adenoviridae/genetics , Animals , CD4 Antigens/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/genetics , CD40 Ligand/immunology , CD40 Ligand/metabolism , Dendritic Cells/metabolism , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/immunology , Interleukin-12 Subunit p40/biosynthesis , Interleukin-12 Subunit p40/deficiency , Interleukin-12 Subunit p40/immunology , Interleukin-23/metabolism , Interleukin-23 Subunit p19/deficiency , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, IgG/biosynthesis , Receptors, IgG/metabolism , Transduction, Genetic , Vaccination
13.
J Immunol ; 185(3): 1476-84, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20581146

ABSTRACT

The functional role of IL-12 in rheumatoid arthritis is controversial. Moreover, whether IL-12 contributes to regulation of Ab-induced joint inflammation remains unclear. To address these issues, we explored the functional roles of IL-12 in Ab-induced arthritis using the K/BxN serum transfer model. IL-12p35(-/-) and IL-12Rbeta(2)(-/-) mice were resistant to the development of arthritis. Injection of K/BxN serum into IL-12p40-yellow fluorescence protein reporter (yet40) mice induced CD11b(+) cells, CD11c(+) cells, and Gr-1(+) granulocytes to produce IL-12p40 in the joints. The levels of IFN-gamma, IL-4, and IL-6 production were lower in joint tissues of IL-12p35(-/-) and IL-12Rbeta(2)(-/-) mice than in B6 mice, whereas levels of TGF-beta expression were higher. Administering IL-12p35(-/-) mice rIL-12 or IFN-gamma restored joint inflammation and suppressed TGF-beta production in joint tissues. Moreover, administering neutralizing anti-TGF-beta mAb enhanced joint inflammation. Among the immune cells that infiltrated joint tissues during Ab-induced arthritis, NKT cells expressed IL-12beta(2) receptors. Furthermore, the adoptive transfer of splenocytes from B6 or Gr-1(+) granulocyte-depleted mice restored joint inflammation in IL-12Rbeta(2)(-/-) mice as much as in B6 mice, whereas splenocytes from Jalpha18(-/-) mice did not. These findings indicate that signals via IL-12beta(2) receptors on NKT cells play a critical role in the development of Ab-induced arthritis. The IL-12p35/IFN-gamma axis promotes Ab-induced joint inflammation by activating NKT cells and suppressing TGF-beta, which may provide novel information for the development of new therapeutic strategies for the inhibition of rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Immune Sera/administration & dosage , Interleukin-12 Subunit p35/physiology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/administration & dosage , Arthritis, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Knock-In Techniques , Immune Complex Diseases/immunology , Immune Complex Diseases/metabolism , Immune Complex Diseases/pathology , Immune Sera/blood , Inflammation Mediators/administration & dosage , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Receptors, Interleukin-12/deficiency , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/physiology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/immunology
14.
J Immunol ; 181(12): 8576-84, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050277

ABSTRACT

IL-12 family members are an important link between innate and adaptive immunity. IL-12 drives Th1 responses by augmenting IFN-gamma production, which is key for clearance of intracellular pathogens. IL-23 promotes the development of IL-17-producing CD4(+) T cells that participate in the control of extracellular pathogens and the induction of autoimmunity. However, recent studies have shown that these cytokines can modulate lymphocyte migration and cellular interactions. Therefore, we sought to determine the individual roles of IL-12 and IL-23 in naive CD8(+) T cell activation by addressing their ability to influence IFN-gamma production and cellular interaction dynamics during priming by Listeria monocytogenes-infected dendritic cells (DC). We found that IL-12 was the major cytokine influencing the level of IFN-gamma production by CD8(+) T cells while IL-23 had little effect on this response. In addition, we observed that IL-12 promoted longer duration conjugation events between CD8(+) T cells and DC. This enhanced cognate interaction time correlated with increased production of the chemokines CCL1 and CCL17 by WT but not IL-12-deficient DC. Neutralization of both chemokines resulted in reduced interaction time and IFN-gamma production, demonstrating their importance in priming naive CD8(+) T cells. Our study demonstrates a novel mechanism through which IL-12 augments naive CD8(+) T cell activation by facilitating chemokine production, thus promoting more stable cognate interactions during priming.


Subject(s)
Adjuvants, Immunologic/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL17/biosynthesis , Chemokine CCL1/biosynthesis , Dendritic Cells/immunology , Interleukin-12 Subunit p35/biosynthesis , Interleukin-12 Subunit p40/biosynthesis , Lymphocyte Activation/immunology , Adjuvants, Immunologic/physiology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cell Communication/immunology , Cells, Cultured , Chemokine CCL1/physiology , Chemokine CCL17/physiology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Interferon-gamma/biosynthesis , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/physiology , Interleukin-12 Subunit p40/deficiency , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/physiology , Interleukin-23/physiology , Listeria monocytogenes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Up-Regulation/immunology
15.
J Exp Med ; 205(4): 799-810, 2008 Apr 14.
Article in English | MEDLINE | ID: mdl-18391061

ABSTRACT

Experimental autoimmune uveitis (EAU) represents autoimmune uveitis in humans. We examined the role of the interleukin (IL)-23-IL-17 and IL-12-T helper cell (Th)1 pathways in the pathogenesis of EAU. IL-23 but not IL-12 was necessary to elicit disease by immunization with the retinal antigen (Ag) interphotoreceptor retinoid-binding protein (IRBP) in complete Freund's adjuvant. IL-17 played a dominant role in this model; its neutralization prevented or reversed disease, and Th17 effector cells induced EAU in the absence of interferon (IFN)-gamma. In a transfer model, however, a polarized Th1 line could induce severe EAU independently of host IL-17. Furthermore, induction of EAU with IRBP-pulsed mature dendritic cells required generation of an IFN-gamma-producing effector response, and an IL-17 response by itself was insufficient to elicit pathology. Finally, genetic deficiency of IL-17 did not abrogate EAU susceptibility. Thus, autoimmune pathology can develop in the context of either a Th17 or a Th1 effector response depending on the model. The data suggest that the dominant effector phenotype may be determined at least in part by conditions present during initial exposure to Ag, including the quality/quantity of Toll-like receptor stimulation and/or type of Ag-presenting cells. These data also raise the possibility that the nonredundant requirement for IL-23 in EAU may extend beyond its role in promoting the Th17 effector response and help provide a balance in the current Th1 versus Th17 paradigm.


Subject(s)
Autoimmune Diseases/enzymology , Autoimmune Diseases/pathology , Autoimmunity/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Animals , Antigens/immunology , Autoimmune Diseases/prevention & control , Cell Line , Dendritic Cells/immunology , Disease Susceptibility , Eye/pathology , Humans , Inflammation Mediators/metabolism , Interferon-gamma/deficiency , Interferon-gamma/immunology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/immunology , Interleukin-17/immunology , Interleukin-23/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Neutralization Tests , Up-Regulation , Uveitis/immunology , Uveitis/prevention & control
16.
J Immunol ; 180(4): 2140-8, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18250420

ABSTRACT

Engagement of OX40 greatly improves CD4 T cell function and survival. Previously, we showed that both OX40 engagement and CTLA-4 blockade led to enhanced CD4 T cell expansion, but only OX40 signaling increased survival. To identify pathways associated with OX40-mediated survival, the gene expression of Ag-activated CD4 T cells isolated from mice treated with anti-OX40 and -CTLA-4 was compared. This comparison revealed a potential role for IL-12 through increased expression of the IL-12R-signaling subunit (IL-12Rbeta2) on T cells activated 3 days previously with Ag and anti-OX40. The temporal expression of IL-12Rbeta2 on OX40-stimulated CD4 T cells was tightly regulated and peaked approximately 4-6 days after initial activation/expansion, but before the beginning of T cell contraction. IL-12 signaling, during this window of IL-12Rbeta2 expression, was required for enhanced T cell survival and survival was associated with STAT4-specific signaling. The findings from these observations were exploited in several different mouse tumor models where we found that the combination of anti-OX40 and IL-12 showed synergistic therapeutic efficacy. These results may lead to the elucidation of the molecular pathways involved with CD4 T cell survival that contribute to improved memory, and understanding of these pathways could lead to greater efficacy of immune stimulatory Abs in tumor-bearing individuals.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Interleukin-12/physiology , Receptors, OX40/immunology , Animals , Antibodies, Blocking/administration & dosage , CD4-Positive T-Lymphocytes/metabolism , Cell Survival/immunology , Female , Interleukin-12/deficiency , Interleukin-12/genetics , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Prostatic Neoplasms/immunology , Receptors, Interleukin-12/biosynthesis , Receptors, Interleukin-12/deficiency , Receptors, Interleukin-12/genetics , Receptors, OX40/agonists , Receptors, OX40/metabolism , Sarcoma, Experimental/immunology , Signal Transduction/immunology
17.
Nature ; 450(7169): 566-9, 2007 Nov 22.
Article in English | MEDLINE | ID: mdl-18033300

ABSTRACT

Regulatory T (T(reg)) cells are a critical sub-population of CD4+ T cells that are essential for maintaining self tolerance and preventing autoimmunity, for limiting chronic inflammatory diseases, such as asthma and inflammatory bowel disease, and for regulating homeostatic lymphocyte expansion. However, they also suppress natural immune responses to parasites and viruses as well as anti-tumour immunity induced by therapeutic vaccines. Although the manipulation of T(reg) function is an important goal of immunotherapy, the molecules that mediate their suppressive activity remain largely unknown. Here we demonstrate that Epstein-Barr-virus-induced gene 3 (Ebi3, which encodes IL-27beta) and interleukin-12 alpha (Il12a, which encodes IL-12alpha/p35) are highly expressed by mouse Foxp3+ (forkhead box P3) T(reg) cells but not by resting or activated effector CD4+ T (T(eff)) cells, and that an Ebi3-IL-12alpha heterodimer is constitutively secreted by T(reg) but not T(eff) cells. Both Ebi3 and Il12a messenger RNA are markedly upregulated in T(reg) cells co-cultured with T(eff) cells, thereby boosting Ebi3 and IL-12alpha production in trans. T(reg)-cell restriction of this cytokine occurs because Ebi3 is a downstream target of Foxp3, a transcription factor that is required for T(reg)-cell development and function. Ebi3-/- and Il12a-/- T(reg) cells have significantly reduced regulatory activity in vitro and fail to control homeostatic proliferation and to cure inflammatory bowel disease in vivo. Because these phenotypic characteristics are distinct from those of other IL-12 family members, this novel Ebi3-IL-12alpha heterodimeric cytokine has been designated interleukin-35 (IL-35). Ectopic expression of IL-35 confers regulatory activity on naive T cells, whereas recombinant IL-35 suppresses T-cell proliferation. Taken together, these data identify IL-35 as a novel inhibitory cytokine that may be specifically produced by T(reg) cells and is required for maximal suppressive activity.


Subject(s)
Interleukin-12 Subunit p35/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Line , Cell Proliferation , Disease Models, Animal , Flow Cytometry , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Inflammatory Bowel Diseases/metabolism , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/transplantation
18.
Infect Immun ; 75(11): 5338-45, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17785474

ABSTRACT

The production of gamma interferon (IFN-gamma) is a key step in the protective innate immune response to Francisella tularensis. Natural killer cells and T cells in the liver are important sources of this cytokine during primary F. tularensis infections, and interleukin-12 (IL-12) appears to be an essential coactivating cytokine for hepatic IFN-gamma expression. The present study was undertaken to determine whether or not macrophages (Mphi) or dendritic cells (DC) provide coactivating signals for the liver IFN-gamma response in vitro, whether IL-12 mediates these effects, and whether Toll-like receptor (TLR) signaling is essential to induce this costimulatory activity. Both bone marrow-derived Mphi and DC significantly augmented the IFN-gamma response of F. tularensis-challenged liver lymphocytes in vitro. While both cell types produced IL-12p40 in response to F. tularensis challenge, only DC secreted large quantities of IL-12p70. DC from both IL-12p35-deficient and TLR2-deficient mice failed to produce IL-12p70 and did not costimulate liver lymphocytes for IFN-gamma production in response to viable F. tularensis organisms. Conversely, liver lymphocytes from TLR2-deficient mice cocultured with wild-type accessory cells produced IFN-gamma at levels comparable to those for wild-type hepatic lymphocytes. These findings indicate that TLR2 controls hepatic lymphocyte IFN-gamma responses to F. tularensis by regulating DC IL-12 production. While Mphi also coinduced hepatic IFN-gamma production in response to F. tularensis, they did so in a fashion less dependent on TLR2.


Subject(s)
Francisella tularensis/immunology , Interferon-gamma/biosynthesis , Liver/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 2/immunology , Animals , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Female , Interleukin-12/biosynthesis , Interleukin-12/immunology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p40/biosynthesis , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 2/deficiency
19.
J Immunol ; 178(4): 2589-98, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277169

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE), a T cell-mediated inflammatory disease of the CNS, is a rodent model of human multiple sclerosis. IL-23 is one of the critical cytokines in EAE development and is currently believed to be involved in the maintenance of encephalitogenic responses during the tissue damage effector phase of the disease. In this study, we show that encephalitogenic T cells from myelin oligodendrocyte glycopeptide (MOG)-immunized wild-type (WT) mice caused indistinguishable disease when adoptively transferred to WT or IL-23-deficient (p19 knockout (KO)) recipient mice, demonstrating that once encephalitogenic cells have been generated, EAE can develop in the complete absence of IL-23. Furthermore, IL-12/23 double-deficient (p35/p19 double KO) recipient mice developed EAE that was indistinguishable from WT recipients, indicating that IL-12 did not compensate for IL-23 deficiency during the effector phase of EAE. In contrast, MOG-specific T cells from p19KO mice induced EAE with delayed onset and much lower severity when transferred to WT recipient mice as compared with the EAE that was induced by cells from WT controls. MOG-specific T cells from p19KO mice were highly deficient in the production of IFN-gamma, IL-17A, and TNF, indicating that IL-23 plays a critical role in development of encephalitogenic T cells and facilitates the development of T cells toward both Th1 and Th17 pathways.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-12 Subunit p35/immunology , Interleukin-23 Subunit p19/immunology , Multiple Sclerosis/immunology , Th1 Cells/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Interleukin-12 Subunit p35/deficiency , Interleukin-23 Subunit p19/deficiency , Male , Mice , Mice, Knockout , Multiple Sclerosis/pathology , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Associated Glycoprotein/toxicity , Myelin-Oligodendrocyte Glycoprotein , Th1 Cells/pathology , Th1 Cells/transplantation
20.
J Exp Med ; 203(11): 2485-94, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17030948

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract that is caused in part by a dysregulated immune response to the intestinal flora. The common interleukin (IL)-12/IL-23p40 subunit is thought to be critical for the pathogenesis of IBD. We have analyzed the role of IL-12 versus IL-23 in two models of Helicobacter hepaticus-triggered T cell-dependent colitis, one involving anti-IL-10R monoclonal antibody treatment of infected T cell-sufficient hosts, and the other involving CD4+ T cell transfer into infected Rag-/- recipients. Our data demonstrate that IL-23 and not IL-12 is essential for the development of maximal intestinal disease. Although IL-23 has been implicated in the differentiation of IL-17-producing CD4+ T cells that alone are sufficient to induce autoimmune tissue reactivity, our results instead support a model in which IL-23 drives both interferon gamma and IL-17 responses that together synergize to trigger severe intestinal inflammation.


Subject(s)
Colitis/immunology , Colitis/microbiology , Helicobacter Infections/immunology , Helicobacter hepaticus/immunology , Interleukin-23/physiology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Colitis/metabolism , Female , Helicobacter Infections/metabolism , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-10/antagonists & inhibitors , Receptors, Interleukin-10/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...