101
|
Nie Y, Zhang D, Qian F, Wu Y. Baccatin III ameliorates bleomycin-induced pulmonary fibrosis via suppression of TGF-β1 production and TGF-β1-induced fibroblast differentiation. Int Immunopharmacol 2019; 74:105696. [PMID: 31229901 DOI: 10.1016/j.intimp.2019.105696] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 12/25/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and generally lethal lung disease with a high mortality rate. Current therapeutic drugs exhibit limited efficacy but severe adverse effects. Paclitaxel has been identified to exert both anti-inflammatory and anti-fibrosis activity. Baccatin III (BAC), an important precursor of paclitaxel, has been identified as exhibiting immunomodulatory activity with decisively lower toxicity. However, its effects on pulmonary fibrosis remain unknown. In this study, the role of BAC in bleomycin (BLM)-induced pulmonary fibrosis and inflammation in mice was investigated in addition to elucidation of its mechanism of action. Our results demonstrated that administration of BAC in a dose-dependent manner reduced inflammatory infiltration, secretion of the pro-fibrotic mediator TGF-β1 and deposition of collagen and other components of the extracellular matrix (ECM), including alpha smooth muscle actin (α-SMA) and fibronectin. Administration of BAC to treat isolated macrophages stimulated with IL-13, known to activate macrophages, the principal source of TGF-β1, resulted in markedly reduced TGF-β1 expression from macrophages. The AKT/STAT6 signaling pathway was shown to be involved in this process. In addition, we have provided in vitro evidence that BAC inhibits TGF-β1-induced fibroblast differentiation via the Smad2/3 signaling pathway. Furthermore, intratracheal injection of rTGF-β1 significantly exacerbated the degree of fibrosis which was down-regulated by treatment with BAC. Taken together, our data suggest that BAC exerts a protective effect against lung fibrosis and may serve as a potential therapeutic strategy for IPF.
Collapse
Affiliation(s)
- Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, PR China
| | - Dan Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Feng Qian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Yaxian Wu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, PR China.
| |
Collapse
|
102
|
STAT Family Protein Expression and Phosphorylation State during moDC Development Is Altered by Platinum-Based Chemotherapeutics. J Immunol Res 2019; 2019:7458238. [PMID: 31309123 PMCID: PMC6594321 DOI: 10.1155/2019/7458238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/19/2019] [Accepted: 04/04/2019] [Indexed: 01/05/2023] Open
Abstract
The STAT signaling pathway is important in dendritic cell (DC) development and function. Tumor cells can induce STAT signaling, thereby inhibiting DC maturation and immunostimulatory functions, leading to hampered efficacy of DC-based immunotherapies. Platinum-based chemotherapeutics can inhibit STAT signaling, thereby making them an interesting tool to improve DC development and function. In this study, we provide a comprehensive overview of STAT expression and phosphorylation during DC differentiation and maturation and investigate the effects of platinum drugs on STAT signaling during these processes. Monocytes were differentiated into monocyte-derived DCs (moDCs) with IL-4 and GM-CSF and matured with cytokines or TLR ligands. STAT expression and phosphorylation were analyzed by western blotting, and moDC viability and phenotype were analyzed by flow cytometry. Platinum drugs were added at day 3 of differentiation or at the start of maturation to investigate regulation of the STAT signaling pathway. All STAT proteins were expressed during moDC differentiation and STAT1, STAT5, and STAT6 were phosphorylated. No significant changes occurred in the expression and phosphorylation state of the STAT proteins during differentiation. After maturation with TLR ligands, the expression of STAT1 increased, but other STAT proteins were not affected. Phosphorylation of STAT1 and STAT3 increased during maturation, where TLR ligands induced significantly higher levels of phosphorylation than cytokines. Platinum drugs cisplatin and oxaliplatin significantly inhibited phosphorylation of STAT6 during differentiation and maturation. Treatment did not affect the phenotype or viability of the cells. As STAT6 is an important regulator of DC function, these findings suggest a role for platinum-based chemotherapeutics to enhance DC function via inhibition of STAT signaling, thereby potentially enhancing efficacy of DC-based immunotherapies.
Collapse
|
103
|
Wheeler JC, Vanoni S, Zeng C, Waggoner L, Yang Y, Wu D, Uddin J, Karns R, Kottyan L, Mukkada V, Rothenberg ME, Hogan SP. 17β-Estradiol protects the esophageal epithelium from IL-13-induced barrier dysfunction and remodeling. J Allergy Clin Immunol 2019; 143:2131-2146. [PMID: 30578870 PMCID: PMC6556402 DOI: 10.1016/j.jaci.2018.10.070] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/30/2018] [Accepted: 10/25/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The incidence of eosinophilic esophagitis (EoE) is greater in male than female subjects, and the underlying molecular basis for this sex bias remains unclear. OBJECTIVE We sought to delineate the contribution of the sex hormone estrogen to the EoE phenotype and esophageal epithelial barrier function and remodeling. METHODS We performed demographic and incidence analyses of EoE in male and female subjects from a single-center pediatric cohort. Estrogen-responsive gene expression analyses and estrogen receptor (ESR) immunofluorescence staining of esophageal biopsy specimens from patients with EoE and control subjects were performed. The effect of 17β-estradiol (E2) on IL-13-induced signaling pathways, gene expression, and esophageal epithelial architecture and barrier function in a primary human esophageal keratinocyte cell (EPC2) culture system (EPC2-air-liquid interface) was examined. RESULTS We observed a male predominance in patients with EoE. Analyses of RNA sequencing data sets revealed a significant dysregulation of the estrogen-responsive gene network and expression of ESR1 and ESR2 in esophageal biopsy specimens from patients with EoE compared with control subjects. IL-13 stimulation of EPC2-air-liquid interface cells led to altered cellular architecture with induced dilation of intercellular spaces and barrier dysfunction. Pretreatment of EPC2s with E2 prior to IL-13 exposure abrogated IL-13-induced architectural changes and esophageal barrier dysfunction. Mechanistically, E2-protective effects were dependent on ESR2 and associated with diminishing of IL-13-induced tyrosine kinase 2 and signal transducer and activator of transcription 6 phosphorylation and EoE-dysregulated gene expression. CONCLUSIONS Estrogen-responsive genes are modified in patients with EoE compared with control subjects. E2 attenuated IL-13-induced architectural changes and esophageal epithelial barrier dysfunction through inhibition of the IL-13/tyrosine kinase 2/signal transducer and activator of transcription 6 pathway via ESR2-dependent process. Estrogen hormone signaling may protect against development of EoE in female subjects.
Collapse
Affiliation(s)
- Justin C Wheeler
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Simone Vanoni
- Division of Allergy and Immunology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Chang Zeng
- Division of Allergy and Immunology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lisa Waggoner
- Division of Allergy and Immunology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Yanfen Yang
- Division of Allergy and Immunology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - David Wu
- Division of Allergy and Immunology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jazib Uddin
- Center for Autoimmune Genomics and Etiology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Rebekah Karns
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Leah Kottyan
- Center for Autoimmune Genomics and Etiology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Vincent Mukkada
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Simon P Hogan
- Division of Allergy and Immunology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Mary H Weiser Food Allergy Center, Department of Pathology, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
104
|
Choi JY, Yun J, Hwang CJ, Lee HP, Kim HD, Chun H, Park PH, Choi DY, Han SB, Hong JT. (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) Phenol Ameliorates MPTP-Induced Dopaminergic Neurodegeneration by Inhibiting the STAT3 Pathway. Int J Mol Sci 2019; 20:ijms20112632. [PMID: 31146332 PMCID: PMC6600543 DOI: 10.3390/ijms20112632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is implicated in dopaminergic neurodegeneration. We have previously demonstrated that (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol (MMPP), a selective signal transducer and activator of transcription 3 (STAT3) inhibitor, has anti-inflammatory properties in several inflammatory disease models. We investigated whether MMPP could protect against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic cell loss and behavioral impairment. Imprinting control region (ICR) mice (8 weeks old, n = 10 per group) were administered MMPP (5 mg/kg) in drinking water for 1 month, and injected with MPTP (15 mg/kg, four times with 2 h intervals) during the last 7 days of treatment. MMPP decreased MPTP-induced behavioral impairments in rotarod, pole, and gait tests. We also showed that MMPP ameliorated dopamine depletion in the striatum and inflammatory marker elevation in primary cultured neurons by high-performance liquid chromatography and immunohistochemical analysis. Increased activation of STAT3, p38, and monoamine oxidase B (MAO-B) were observed in the substantia nigra and striatum after MPTP injection, effects that were attenuated by MMPP treatment. Furthermore, MMPP inhibited STAT3 activity and expression of neuroinflammatory proteins, including ionized calcium binding adaptor molecule 1 (Iba1), inducible nitric oxide synthase (iNOS), and glial fibrillary acidic protein (GFAP) in 1-methyl-4-phenylpyridinium (MPP+; 0.5 mM)-treated primary cultured cells. However, mitogen-activated protein kinase (MAPK) inhibitors augmented the activity of MMPP. Collectively, our results suggest that MMPP may be an anti-inflammatory agent that attenuates dopaminergic neurodegeneration and neuroinflammation through MAO-B and MAPK pathway-dependent inhibition of STAT3 activation.
Collapse
Affiliation(s)
- Ji Yeon Choi
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Hae Deun Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Hyungok Chun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Korea.
| | - Dong Young Choi
- College of Pharmacy, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Korea.
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| |
Collapse
|
105
|
Marbach-Breitrück E, Kalledat A, Heydeck D, Stehling S, Fluhr JW, Zuberbier T, Kuhn H. Atopic Patients Show Increased Interleukin 4 Plasma Levels but the Degree of Elevation Is Not Sufficient to Upregulate Interleukin-4-Sensitive Genes. Skin Pharmacol Physiol 2019; 32:192-200. [PMID: 31096247 DOI: 10.1159/000499431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 03/05/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Atopic diseases constitute a major health challenge for industrialized countries, and elevated levels of interleukin 4 (IL-4) frequently characterize these disorders. Previous in vitroanalyses have indicated that IL-4 strongly upregulates the expression of IL-4-sensitive genes in human monocytes. OBJECTIVE To explore whether similar expression alterations may contribute to the pathomechanisms of atopic diseases in vivo we carried out a small-scale case-control clinical study (n = 43), in which we quantified the plasma levels of IgE and IL-4 as well as the expression of selected IL-4-sensitive genes in blood leukocytes. METHODS 34 allergic patients suffering from allergic rhinitis (n = 11), atopic eczema (n = 11) and allergic asthma (n = 12) as well as 9 healthy control individuals were recruited. IgE and IL-4 plasma levels were determined by ELISA, and the expression of selected IL-4-sensitive gene products in blood leukocytes was quantified by qRT-PCR. In addition, the fatty acid oxygenase activity of isolated monocytes was measured by RP-HPLC analysis of the arachidonic acid oxygenation products (ex vivo activity assays). RESULTS We found that plasma levels of IgE and IL-4 were significantly elevated in atopic patients but the degree of elevation was not sufficient to upregulate the expression of the selected IL-4-sensitive genes in circulating leukocytes. Moreover, the arachidonic acid oxygenase activity of blood monocytes was not significantly altered in atopic patients. CONCLUSION Our data suggest that the IL-4 plasma levels of atopic patients are not high enough to impact the expression of IL-4-sensitive genes.
Collapse
Affiliation(s)
- Eugenia Marbach-Breitrück
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Andrea Kalledat
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Dagmar Heydeck
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Sabine Stehling
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Joachim W Fluhr
- Department of Dermatology and Allergology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Torsten Zuberbier
- Department of Dermatology and Allergology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Hartmut Kuhn
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany,
| |
Collapse
|
106
|
Inhibition of IL-13 and IL-13Rα2 Expression by IL-32θ in Human Monocytic Cells Requires PKCδ and STAT3 Association. Int J Mol Sci 2019; 20:ijms20081949. [PMID: 31010051 PMCID: PMC6514684 DOI: 10.3390/ijms20081949] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/06/2019] [Accepted: 04/19/2019] [Indexed: 12/27/2022] Open
Abstract
Interleukin (IL)-32θ, a newly identified IL-32 isoform, has been reported to exert pro-inflammatory effects through the association with protein kinase C delta (PKCδ). In this study, we further examined the effects of IL-32θ on IL-13 and IL-13Rα2 expression and the related mechanism in THP-1 cells. Upon stimulating IL-32θ-expressing and non-expressing cells with phorbol 12-myristate 13-acetate (PMA), the previous microarray analysis showed that IL-13Rα2 and IL-13 mRNA expression were significantly decreased by IL-32θ. The protein expression of these factors was also confirmed to be down-regulated. The nuclear translocation of transcription factors STAT3 and STAT6, which are necessary for IL-13Rα2 and IL-13 promoter activities, was suppressed by IL-32θ. Additionally, a direct association was found between IL-32θ, PKCδ, and signal transducer and activator of transcription 3 (STAT3), but not STAT6, revealing that IL-32θ might act mainly through STAT3 and indirectly affect STAT6. Moreover, the interaction of IL-32θ with STAT3 requires PKCδ, since blocking PKCδ activity eliminated the interaction and consequently limited the inhibitory effect of IL-32θ on STAT3 activity. Interfering with STAT3 or STAT6 binding by decoy oligodeoxynucleotides (ODNs) identified that IL-32θ had additive effects with the STAT3 decoy ODN to suppress IL-13 and IL-13Rα2 mRNA expression. Taken together, our data demonstrate the intracellular interaction of IL-32θ, PKCδ, and STAT3 to regulate IL-13 and IL-13Rα2 synthesis, supporting the role of IL-32θ as an inflammatory modulator.
Collapse
|
107
|
Monoamine Oxidase-Related Vascular Oxidative Stress in Diseases Associated with Inflammatory Burden. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8954201. [PMID: 31178977 PMCID: PMC6501417 DOI: 10.1155/2019/8954201] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/14/2019] [Indexed: 12/16/2022]
Abstract
Monoamine oxidases (MAO) with 2 isoforms, A and B, located at the outer mitochondrial membrane are flavoenzyme membranes with a major role in the metabolism of monoaminergic neurotransmitters and biogenic amines in the central nervous system and peripheral tissues, respectively. In the process of oxidative deamination, aldehydes, hydrogen peroxide, and ammonia are constantly generated as potential deleterious by-products. While being systematically studied for decades as sources of reactive oxygen species in brain diseases, compelling evidence nowadays supports the role of MAO-related oxidative stress in cardiovascular and metabolic pathologies. Indeed, oxidative stress and chronic inflammation are the most common pathomechanisms of the main noncommunicable diseases of our century. MAO inhibition with the new generation of reversible and selective drugs has recently emerged as a pharmacological strategy aimed at mitigating both processes. The aim of this minireview is to summarize available information regarding the contribution of MAO to the vascular oxidative stress and endothelial dysfunction in hypertension, metabolic disorders, and chronic kidney disease, all conditions associated with increased inflammatory burden.
Collapse
|
108
|
Kolivand S, Amini P, Saffar H, Rezapoor S, Motevaseli E, Najafi M, Nouruzi F, Shabeeb D, Musa AE. Evaluating the Radioprotective Effect of Curcumin on Rat’s Heart Tissues. Curr Radiopharm 2019; 12:23-28. [DOI: 10.2174/1874471011666180831101459] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/08/2018] [Accepted: 08/17/2018] [Indexed: 01/01/2023]
Abstract
Background: Heart injury is one of the most important concerns after exposure to a high
dose of radiation in chest cancer radiotherapy or whole body exposure to a radiation disaster. Studies
have proposed that increased level of inflammatory and pro-fibrotic cytokines following radiotherapy
or radiation events play a key role in the development of several side effects such as cardiovascular
disorders. In the current study, we aimed to evaluate the expression of IL-4 and IL-13 cytokines as well
as signaling pathways such as IL4Ra1, IL13Ra2, Duox1 and Duox2. In addition, we detected the
possible protective effect of curcumin on the expression of these factors and infiltration of inflammatory
cells.
Materials and Methods:
Twenty rats were divided into 4 groups including control; curcumin treated;
radiation; and radiation plus curcumin. After 10 weeks, rats were sacrificed for evaluation of mentioned
parameters.
Results:
Results showed an increase in the level of IL-4 and all evaluated genes, as well as increased
infiltration of lymphocytes and macrophages. Treatment with curcumin could attenuate these changes.
Conclusion:
Curcumin could reduce radiation-induced heart injury markers in rats.
Collapse
Affiliation(s)
- Sedighe Kolivand
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Hana Saffar
- Clinical and Anatomical Pathologist at Tehran University of Medical Science, Imam Khomeini Hospital Complex, Tehran, Iran
| | - Saeed Rezapoor
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farzad Nouruzi
- Department of Medical Radiation Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| |
Collapse
|
109
|
Liang Y, Yi P, Yuan DMK, Jie Z, Kwota Z, Soong L, Cong Y, Sun J. IL-33 induces immunosuppressive neutrophils via a type 2 innate lymphoid cell/IL-13/STAT6 axis and protects the liver against injury in LCMV infection-induced viral hepatitis. Cell Mol Immunol 2019; 16:126-137. [PMID: 29400707 PMCID: PMC6355846 DOI: 10.1038/cmi.2017.147] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/11/2017] [Accepted: 11/11/2017] [Indexed: 12/21/2022] Open
Abstract
Viral hepatitis is still a public health problem affecting several million people around the world. Neutrophils are polymorphonuclear cells that have a critical role in antibacterial infection. However, the role of neutrophils in viral infection is not fully understood. By using a mouse model of lymphocytic choriomeningitis virus infection-induced viral hepatitis, we observed increased neutrophil recruitment in the liver accompanied by enhanced CD8+ T-cell responses. Liver neutrophils expressed high levels of immunomodulatory cytokines, such as C-X-C chemokine ligand 2, arginase-1, inducible nitric oxide synthase and interleukin (IL)-10, demonstrating immunosuppressive properties. Depletion of neutrophils in vivo by a neutralizing antibody resulted in the exacerbation of liver injury and the promotion of T-cell responses at the immune contraction stage. IL-33 significantly induced neutrophil recruitment in the liver and attenuated liver injury by limiting effector T-cell accumulation. Mechanistically, we found that IL-33 promoted the expression of arginase-1 in neutrophils through the type 2 innate lymphoid cell (ILC2)-derived IL-13. Additionally, IL-13 increased the inhibitory effect of neutrophils on CD8+ T-cell proliferation in vitro, partially through arginase-1. Finally, we found that IL-13 induced arginase-1 expression, depending on signal transducer and activator of transcription factor 6 (STAT6) signaling. Therefore, IL-33 induced immunosuppressive neutrophils via an ILC2/IL-13/STAT6 axis. Collectively, our findings shed new light on the mechanisms associated with IL-33-triggered neutrophils in the liver and suggest potential targets for therapeutic investigation in viral hepatitis.
Collapse
Affiliation(s)
- Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX.
| | - Panpan Yi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Denley Ming Kee Yuan
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX
| | - Zuliang Jie
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX
| | - Zakari Kwota
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX
- Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX
- Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX.
- Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, 77555-1070, USA, TX.
| |
Collapse
|
110
|
Yang F, Liu Y, Ren H, Zhou G, Yuan X, Shi X. ER-stress regulates macrophage polarization through pancreatic EIF-2alpha kinase. Cell Immunol 2019; 336:40-47. [PMID: 30594305 DOI: 10.1016/j.cellimm.2018.12.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/04/2018] [Accepted: 12/20/2018] [Indexed: 02/08/2023]
Abstract
During the process of NAFLD progression, ER-stress is activated in macrophages and induces the pro-inflammatory polarization of macrophage. As one of the three ER membrane resident proteins, pancreatic eIF-2alpha kinase (PERK) plays an important role in ER stress, but its participation in macrophage polarization is largely unknown. In this study, we found that the PA mediated ER-stress activation could induce M1-type polarization in macrophages, and this phenotype polarization could be inhibited by ER-stress inhibitor 4-PBA as well as GSK2656157, an inhibitor of PERK. Moreover, the knockdown of PERK altered the STAT1 and STAT6 pathways in macrophages, which then led to the M1-to-M2 phenotypic shift. In summary, we found that PERK could regulate the phenotypic polarization of macrophages. This finding may provide new insight into the suppression of pathological progression of fatty liver or liver ischemia reperfusion injury induced by M1-type macrophages.
Collapse
Affiliation(s)
- Faji Yang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Yang Liu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Guang Zhou
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Xianwen Yuan
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Xiaolei Shi
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China.
| |
Collapse
|
111
|
Ajayi AAL. Itching, chloroquine, and malaria: a review of recent molecular and neuroscience advances and their contribution to mechanistic understanding and therapeutics of chronic non-histaminergic pruritus. Int J Dermatol 2018; 58:880-891. [PMID: 30362504 DOI: 10.1111/ijd.14252] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/23/2018] [Accepted: 08/31/2018] [Indexed: 12/20/2022]
Abstract
Chloroquine (CQ) is an antimalarial drug that elicits severe pruritus in black Africans with malaria fever. This acute itching (2-7 days duration) exhibits age dependency and a racial and genetic predilection. CQ itch is non-histaminergic, which makes it both a good model and a tool to probe the mechanisms of chronic itch. This review focuses on recently discovered mechanisms, neuroscience, mediators, and receptors that are implicated in molecular studies of CQ pruritus. CQ pruritus mechanisms are also compared to that of itching following other systemic diseases, such as chronic kidney disease, chronic liver disease, skin disorders, and burns. There are striking similarities between CQ itching pathways and other chronic itch secondary to systemic disease with or without skin lesions, which have not been previously highlighted. Prominent among these are the shared roles of skin, neural and spinal μ opiate receptors, kappa opiate receptor, nitric oxide, serotonin via 5HT1B/D receptors, cytokines, especially interleukins, and tumor necrosis factor. There is elaborate "cross talk" among the diverse mediators and receptors involved in CQ-induced pruritus. CQ also binds to the mas-related G protein coupled receptors MrgprA3/MrgprX1 present in a small proportion (4-5%) of dorsal root ganglion neurons and skin. The mrgprA3 CQ receptors are coupled to PLC-β3 and a chloride channel to initiate skin itch action potentials in C nerve fibers. Mrgpra3/X1 couples to TRPA1 for calcium influx into neuronal cells at noncutaneous sites. Central CQ itch occurs via gastrin-related peptide (GRP) and its receptor (GRPR) in the dorsal spinothalamic tracts, as well as glutamic mediated GRP projection to parabrachial nucleus. The possibility of chronic itch therapy based on personalized medicine, genetics, and transcriptomics or the use of itch "polypill/polycream" are discussed.
Collapse
Affiliation(s)
- Adesuyi A L Ajayi
- Department of Medicine, Division of Clinical Pharmacology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
112
|
Wodsedalek DJ, Paddock SJ, Wan TC, Auchampach JA, Kenarsary A, Tsaih SW, Flister MJ, O'Meara CC. IL-13 promotes in vivo neonatal cardiomyocyte cell cycle activity and heart regeneration. Am J Physiol Heart Circ Physiol 2018; 316:H24-H34. [PMID: 30339498 DOI: 10.1152/ajpheart.00521.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
There is great interest in identifying signaling mechanisms by which cardiomyocytes (CMs) can enter the cell cycle and promote endogenous cardiac repair. We have previously demonstrated that IL-13 stimulated cell cycle activity of neonatal CMs in vitro. However, the signaling events that occur downstream of IL-13 in CMs and the role of IL-13 in CM proliferation and regeneration in vivo have not been explored. Here, we tested the role of IL-13 in promoting neonatal CM cell cycle activity and heart regeneration in vivo and investigated the signaling pathway(s) downstream of IL-13 specifically in CMs. Compared with control, CMs from neonatal IL-13 knockout (IL-13-/-) mice showed decreased proliferative markers and coincident upregulation of the hypertrophic marker brain natriuretic peptide ( Nppb) and increased CM nuclear size. After apical resection in anesthetized newborn mice, heart regeneration was significantly impaired in IL-13-/- mice compared with wild-type mice. Administration of recombinant IL-13 reversed these phenotypes by increasing CM proliferation markers and decreasing Nppb expression. RNA sequencing on primary neonatal CMs treated with IL-13 revealed activation of gene networks regulated by ERK1/2 and Akt. Western blot confirmed strong phosphorylation of ERK1/2 and Akt in both neonatal and adult cultured CMs in response to IL-13. Our data demonstrated a role for endogenous IL-13 in neonatal CM cell cycle and heart regeneration. ERK1/2 and Akt signaling are important pathways known to promote CM proliferation and protect against apoptosis, respectively; thus, targeting IL-13 transmembrane receptor signaling or administering recombinant IL-13 may be therapeutic approaches for activating proregenerative and survival pathways in the heart. NEW & NOTEWORTHY Here, we demonstrate, for the first time, that IL-13 is involved in neonatal cardiomyocyte cell cycle activity and heart regeneration in vivo. Prior work has shown that IL-13 promotes cardiomyocyte cell cycle activity in vitro; however, the signaling pathways were unknown. We used RNA sequencing to identify the signaling pathways activated downstream of IL-13 in cardiomyocytes and found that ERK1/2 and Akt signaling was activated in response to IL-13.
Collapse
Affiliation(s)
- Dylan J Wodsedalek
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Samantha J Paddock
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Tina C Wan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - John A Auchampach
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Aria Kenarsary
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Shirng-Wern Tsaih
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Michael J Flister
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Caitlin C O'Meara
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
113
|
Barik S, Cattin-Roy AN, Miller MM, Ukah TK, Zaghouani H. IL-4 and IL-13 Guide Early Thymic Progenitors To Mature toward Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:2947-2958. [PMID: 30291166 DOI: 10.4049/jimmunol.1701186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/04/2018] [Indexed: 12/15/2022]
Abstract
Recently we reported that IL-4 and IL-13 signaling in murine early thymic progenitors (ETPs) expressing the heteroreceptor (HR) comprising IL-4 receptor α (IL-4Rα) and IL-13 receptor α 1 (IL-13Rα1) activate STAT6 and inhibit ETP maturation potential toward T cells. In this study, we asked whether IL-4 and IL-13 signaling through the HR mobilizes other STAT molecules to shape ETP fate decision. The findings indicate that HR+ ETPs undergoing cytokine signaling display increased STAT1, but not STAT3, phosphorylation in addition to STAT6 activation. In parallel, the ETPs had a STAT1-dependent heightened expression of IRF-8, a transcription factor essential for development of CD8α+ dendritic cells (DCs). Interestingly, STAT1 phosphorylation and IRF-8 upregulation, which are independent of STAT6 activation, guided ETP maturation toward myeloid cells with a CD8α+ DC phenotype. Furthermore, these CD8α+ DCs display a thymic resident phenotype, as they did not express SIRPα, a molecule presumed to be involved in cell migration. These findings suggest that IL-4 and IL-13 cytokine-induced HR signaling provides a double-edged sword that simultaneously blocks T cell lineage potential but advances myeloid maturation that could impact T cell selection and central tolerance.
Collapse
Affiliation(s)
- Subhasis Barik
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Alexis N Cattin-Roy
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Mindy M Miller
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Tobechukwu K Ukah
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Habib Zaghouani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212; .,Department of Neurology, University of Missouri School of Medicine, Columbia, MO 65212; and.,Department of Child Health, University of Missouri School of Medicine, Columbia, MO 65212
| |
Collapse
|
114
|
Tarique AA, Sly PD, Cardenas DG, Luo L, Stow JL, Bell SC, Wainwright CE, Fantino E. Differential expression of genes and receptors in monocytes from patients with cystic fibrosis. J Cyst Fibros 2018; 18:342-348. [PMID: 30177416 DOI: 10.1016/j.jcf.2018.07.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/29/2018] [Accepted: 07/30/2018] [Indexed: 11/30/2022]
Abstract
INTRODUCTION We previously reported defective alternative polarization (M2) of macrophages and early expression of classically polarized (M1) macrophage markers in unpolarized monocyte-derived macrophages (MDMs) in patients with cystic fibrosis (CF). The present study assessed whether the mechanism(s) underlying defective macrophage polarization resided in circulating monocytes. METHODS Monocyte subsets (classical, intermediate and non-classical), markers for monocyte activation (CD163) and recruitment (CD195), receptors/genes associated with macrophage differentiation and polarization were analyzed in CF and compared with healthy individuals. RESULTS No differences were observed in the monocyte subsets or in the expression of CD163 or CD195. Expression of the M-CSF receptor, TLR4, γC, IL-4Rα, IL-13Rα1, TIMP-1 and Cox-2 were higher in CF monocytes, albeit at low levels, whereas, LRP1, MMP9, MMP28 were downregulated compared to mooncytes from healthy individuals. CONCLUSIONS Our data suggest that differences in CF monocytes may contribute to the reported CFTR-dependent defect in macrophage differentiation, polarization and function.
Collapse
Affiliation(s)
- Abdullah A Tarique
- Child Health Research Centre (CHRC), The University of Queensland, Australia
| | - Peter D Sly
- Child Health Research Centre (CHRC), The University of Queensland, Australia; Department of Respiratory and Sleep Medicine, Children's Health Queensland, Brisbane, Australia.
| | - Diana G Cardenas
- Child Health Research Centre (CHRC), The University of Queensland, Australia
| | - Lin Luo
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, Australia
| | - Jennifer L Stow
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, Australia
| | - Scott C Bell
- QIMR Berghofer Medical Research Institute, Brisbane, Australia; Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| | - Claire E Wainwright
- Child Health Research Centre (CHRC), The University of Queensland, Australia; Department of Respiratory and Sleep Medicine, Children's Health Queensland, Brisbane, Australia
| | - Emmanuelle Fantino
- Child Health Research Centre (CHRC), The University of Queensland, Australia
| |
Collapse
|
115
|
Mao K, Chen W, Mu Y, Ao J, Chen X. Identification of two IL-4/13 homologues in large yellow croaker (Larimichthys crocea) revealed their similar roles in inducing alternative activation of monocytes/macrophages. FISH & SHELLFISH IMMUNOLOGY 2018; 80:180-190. [PMID: 29870826 DOI: 10.1016/j.fsi.2018.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/01/2018] [Indexed: 05/02/2023]
Abstract
Mammalian interleukin-4 (IL-4) and -13 (IL-13), two anti-inflammatory T helper cell type 2 (Th2) cytokines, play the central roles in mediating the alternative activation of monocytes/macrophages (MO/Mφs). However, exact functions in MO/Mφs polarization of IL-4/13 homologues in teleost fish remain largely unknown. In this study, we identified two IL-4/13 homologues from large yellow croaker Larimichthys crocea, LcIL-4/13A and LcIL-4/13B, which share low amino acid sequence identities to the known fish IL-4/13 molecules. Phylogenetic analysis showed that LcIL-4/13A is evolutionarily closely related to Dicentrarchus labrax IL-4/13A, and LcIL-4/13B to Takifugu rubripes IL-4/13B. The two LcIL-4/13 genes were constitutively expressed in all examined tissues, but with different expression levels. Both LcIL-4/13A and LcIL-4/13B were up-regulated by inactivated trivalent bacterial vaccine in the head kidney, and LcIL-4/13B appeared more responsive to bacterial vaccine than LcIL-4/13A. Recombinant LcIL-4/13A and LcIL-4/13B proteins (rLcIL-4/13A and rLcIL-4/13B) produced in Escherichia coli could significantly decrease production of reactive oxygen species (ROS) and nitrogen oxide (NO) in the head kidney MO/Mφs from large yellow croaker. Furthermore, rLcIL-4/13A and rLcIL-4/13B obviously down-regulated expression of pro-inflammatory cytokine (IL-1β and TNF-α) and inducible NO synthase (iNOS) genes in MO/Mφs, while they increased mRNA expression of anti-inflammatory cytokines (TGF-β and VEGF) and arginase-2. Additionally, the phagocytic activity of MO/Mφs was also inhibited by rLcIL-4/13A or rLcIL-4/13B. All these results therefore indicated that both LcIL-4/13A and LcIL-4/13B, although exhibiting a lower degree of sequence identity of 15.6% and differential expression pattern, have the similar roles in promoting alternative activation of head kidney MO/Mφs.
Collapse
Affiliation(s)
- Kaiqiong Mao
- Institute of Oceanology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China; Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, State Oceanic Administration, Xiamen, 361005, PR China; School of Life Sciences, Xiamen University, Xiamen, 361005, PR China
| | - Wei Chen
- School of Life Sciences, Xiamen University, Xiamen, 361005, PR China
| | - Yinnan Mu
- Institute of Oceanology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China.
| | - Jingqun Ao
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, State Oceanic Administration, Xiamen, 361005, PR China
| | - Xinhua Chen
- Institute of Oceanology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
116
|
Yang F, Wang S, Liu Y, Zhou Y, Shang L, Feng M, Yuan X, Zhu W, Shi X. IRE1α aggravates ischemia reperfusion injury of fatty liver by regulating phenotypic transformation of kupffer cells. Free Radic Biol Med 2018; 124:395-407. [PMID: 29969718 DOI: 10.1016/j.freeradbiomed.2018.06.043] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/29/2018] [Accepted: 06/30/2018] [Indexed: 12/18/2022]
Abstract
Fatty liver is one of the widely accepted marginal donor for liver transplantation, but is also more sensitive to ischemia and reperfusion injury (IRI) and produces more reactive oxygen species (ROS). Moreover, so far, no effective method has been developed to alleviate it. Endoplasmic reticulum stress (ER-stress) of hepatocyte is associated with the occurrence of fatty liver disease, but ER-stress of kupffer cells (KCs) in fatty liver is not clear at all. This study evaluates whether ER-stress of KCs is activated in fatty liver and accelerate IRI of fatty livers. ER-stress of KCs was activated in fatty liver, especially the IRE1α signal pathway. KCs with activated ER-stress secreted more proinflammatory cytokine to induce its M1-phenotypic shift in fatty liver, resulting in more severe IRI. Also, activated ER-stress of BMDMs in vitro by tunicamycin can induce its pro-inflammatory shift and can be reduced by 4-PBA, an ER-stress inhibitor. Knockdown of IRE1α could regulate the STAT1 and STAT6 pathway of macrophage to inhibit the M1-type polarization and promote M2-phenotypic shift. Furthermore, transfusion of IRE1α-knockdown KCs significantly reduced the liver IRI as well as the ROS of HFD feeding mice. Altogether, these data demonstrated that IRE1α of KCs may be a potential target to reduce the fatty liver associated IRI in liver transplantation.
Collapse
Affiliation(s)
- Faji Yang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Shuai Wang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Yang Liu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Yuan Zhou
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Longcheng Shang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Min Feng
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Xianwen Yuan
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China
| | - Wei Zhu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China.
| | - Xiaolei Shi
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321, Zhongshan Road, 210008 Nanjing, Jiangsu Province, China.
| |
Collapse
|
117
|
Soendergaard C, Bergenheim FH, Bjerrum JT, Nielsen OH. Targeting JAK-STAT signal transduction in IBD. Pharmacol Ther 2018; 192:100-111. [PMID: 30048708 DOI: 10.1016/j.pharmthera.2018.07.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An unmet medical need exists for novel targeted therapies for inflammatory bowel disease (IBD) as many patients experience inadequate responses to antibody-based biologics. An oral drug formulation with reduced production costs and redundancy for healthcare staff to administer therapy ideally should result in diminished healthcare expenses and improved patient compliance. A new drug class of small molecules, the Janus kinase (JAK) inhibitors (jakinibs), fulfills these criteria and has recently shown efficacy in IBD. Here we provide an overview of the mode of action of jakinibs and provide a comprehensive overview of existing clinical studies. Convincing clinical data show that a complex cytokine-driven inflammation can efficiently be modulated by therapeutic inhibition of the JAK proteins.
Collapse
Affiliation(s)
| | | | | | - Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Denmark.
| |
Collapse
|
118
|
Dhabal S, Das P, Biswas P, Kumari P, Yakubenko VP, Kundu S, Cathcart MK, Kundu M, Biswas K, Bhattacharjee A. Regulation of monoamine oxidase A (MAO-A) expression, activity, and function in IL-13-stimulated monocytes and A549 lung carcinoma cells. J Biol Chem 2018; 293:14040-14064. [PMID: 30021838 DOI: 10.1074/jbc.ra118.002321] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 07/06/2018] [Indexed: 11/06/2022] Open
Abstract
Monoamine oxidase A (MAO-A) is a mitochondrial flavoenzyme implicated in the pathogenesis of atherosclerosis and inflammation and also in many neurological disorders. MAO-A also has been reported as a potential therapeutic target in prostate cancer. However, the regulatory mechanisms controlling cytokine-induced MAO-A expression in immune or cancer cells remain to be identified. Here, we show that MAO-A expression is co-induced with 15-lipoxygenase (15-LO) in interleukin 13 (IL-13)-activated primary human monocytes and A549 non-small cell lung carcinoma cells. We present evidence that MAO-A gene expression and activity are regulated by signal transducer and activator of transcription 1, 3, and 6 (STAT1, STAT3, and STAT6), early growth response 1 (EGR1), and cAMP-responsive element-binding protein (CREB), the same transcription factors that control IL-13-dependent 15-LO expression. We further established that in both primary monocytes and in A549 cells, IL-13-stimulated MAO-A expression, activity, and function are directly governed by 15-LO. In contrast, IL-13-driven expression and activity of MAO-A was 15-LO-independent in U937 promonocytic cells. Furthermore, we demonstrate that the 15-LO-dependent transcriptional regulation of MAO-A in response to IL-13 stimulation in monocytes and in A549 cells is mediated by peroxisome proliferator-activated receptor γ (PPARγ) and that signal transducer and activator of transcription 6 (STAT6) plays a crucial role in facilitating the transcriptional activity of PPARγ. We further report that the IL-13-STAT6-15-LO-PPARγ axis is critical for MAO-A expression, activity, and function, including migration and reactive oxygen species generation. Altogether, these results have major implications for the resolution of inflammation and indicate that MAO-A may promote metastatic potential in lung cancer cells.
Collapse
Affiliation(s)
- Sukhamoy Dhabal
- From the Department of Biotechnology, National Institute of Technology-Durgapur, Mahatma Gandhi Avenue, Durgapur-713209, Burdwan, West Bengal, India
| | - Pradip Das
- From the Department of Biotechnology, National Institute of Technology-Durgapur, Mahatma Gandhi Avenue, Durgapur-713209, Burdwan, West Bengal, India
| | - Pritam Biswas
- From the Department of Biotechnology, National Institute of Technology-Durgapur, Mahatma Gandhi Avenue, Durgapur-713209, Burdwan, West Bengal, India
| | - Priyanka Kumari
- From the Department of Biotechnology, National Institute of Technology-Durgapur, Mahatma Gandhi Avenue, Durgapur-713209, Burdwan, West Bengal, India
| | - Valentin P Yakubenko
- the Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, and Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio 44195, and
| | - Suman Kundu
- the Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, and Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio 44195, and
| | - Martha K Cathcart
- the Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, and Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio 44195, and
| | - Manjari Kundu
- the Division of Molecular Medicine, Bose Institute, Kolkata 700054, West Bengal, India
| | - Kaushik Biswas
- the Division of Molecular Medicine, Bose Institute, Kolkata 700054, West Bengal, India
| | - Ashish Bhattacharjee
- From the Department of Biotechnology, National Institute of Technology-Durgapur, Mahatma Gandhi Avenue, Durgapur-713209, Burdwan, West Bengal, India,
| |
Collapse
|
119
|
Park J, Decker JT, Margul DJ, Smith DR, Cummings BJ, Anderson AJ, Shea LD. Local Immunomodulation with Anti-inflammatory Cytokine-Encoding Lentivirus Enhances Functional Recovery after Spinal Cord Injury. Mol Ther 2018; 26:1756-1770. [PMID: 29778523 PMCID: PMC6037204 DOI: 10.1016/j.ymthe.2018.04.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 12/18/2022] Open
Abstract
Trauma to the spinal cord and associated secondary inflammation can lead to permanent loss of sensory and motor function below the injury level, with the resulting environment serving as a barrier that limits regeneration. In this study, we investigate the localized expression of anti-inflammatory cytokines IL-10 and IL-4 via lentiviral transduction in multichannel bridges. Porous multichannel bridges provide physical guidance for axonal outgrowth with the cytokines hypothesized to modulate the neuroinflammatory microenvironment and enhance axonal regeneration. Gene expression analyses indicated that induced IL-10 and IL-4 expression decreased expression of pro-inflammatory genes and increased pro-regenerative genes relative to control. Moreover, these factors led to increased numbers of axons and myelination, with approximately 45% of axons myelinated and the number of oligodendrocyte myelinated axons significantly increased by 3- to 4-fold. Furthermore, the combination of a bridge with IL-10 and IL-4 expression improved locomotor function after injury to an average score of 6 relative to an average score of 3 for injury alone. Collectively, these studies highlight the potential for localized immunomodulation to decrease secondary inflammation and enhance regeneration that may have numerous applications.
Collapse
Affiliation(s)
- Jonghyuck Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Joseph T Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Daniel J Margul
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Brian J Cummings
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Aileen J Anderson
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
120
|
Knight JM, Mandal P, Morlacchi P, Mak G, Li E, Madison M, Landers C, Saxton B, Felix E, Gilbert B, Sederstrom J, Varadhachary A, Singh MM, Chatterjee D, Corry DB, McMurray JS. Small molecule targeting of the STAT5/6 Src homology 2 (SH2) domains to inhibit allergic airway disease. J Biol Chem 2018; 293:10026-10040. [PMID: 29739850 PMCID: PMC6028980 DOI: 10.1074/jbc.ra117.000567] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 04/02/2018] [Indexed: 11/06/2022] Open
Abstract
Asthma is a chronic inflammatory disease of the lungs and airways and one of the most burdensome of all chronic maladies. Previous studies have established that expression of experimental and human asthma requires the IL-4/IL-13/IL-4 receptor α (IL-4Rα) signaling pathway, which activates the transcription factor STAT6. However, no small molecules targeting this important pathway are currently in clinical development. To this end, using a preclinical asthma model, we sought to develop and test a small-molecule inhibitor of the Src homology 2 domains in mouse and human STAT6. We previously developed multiple peptidomimetic compounds on the basis of blocking the docking site of STAT6 to IL-4Rα and phosphorylation of Tyr641 in STAT6. Here, we expanded the scope of our initial in vitro structure-activity relationship studies to include central and C-terminal analogs of these peptides to develop a lead compound, PM-43I. Conducting initial dose range, toxicity, and pharmacokinetic experiments with PM-43I, we found that it potently inhibits both STAT5- and STAT6-dependent allergic airway disease in mice. Moreover, PM-43I reversed preexisting allergic airway disease in mice with a minimum ED50 of 0.25 μg/kg. Of note, PM-43I was efficiently cleared through the kidneys with no long-term toxicity. We conclude that PM-43I represents the first of a class of small molecules that may be suitable for further clinical development against asthma.
Collapse
Affiliation(s)
- J Morgan Knight
- From the Departments of Medicine,
- Pathology and Immunology, and
| | - Pijus Mandal
- the Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| | - Pietro Morlacchi
- the Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| | | | - Evan Li
- From the Departments of Medicine
- the Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas 77030
| | - Matthew Madison
- the Translational Biology and Molecular Medicine Program, and
| | - Cameron Landers
- the Translational Biology and Molecular Medicine Program, and
| | | | - Ed Felix
- the Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| | | | - Joel Sederstrom
- the Flow Cytometry Core Facility, Baylor College of Medicine, Houston, Texas
| | - Atul Varadhachary
- Fannin Innovation Studio and Atrapos Therapeutics, LLC, Houston, Texas 77027
| | - Melissa M Singh
- Fannin Innovation Studio and Atrapos Therapeutics, LLC, Houston, Texas 77027
| | - Dev Chatterjee
- Fannin Innovation Studio and Atrapos Therapeutics, LLC, Houston, Texas 77027
| | - David B Corry
- From the Departments of Medicine,
- Pathology and Immunology, and
- the Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas 77030
- the Translational Biology and Molecular Medicine Program, and
- the Michael E. Debakey Veterans Affairs Center for Translational Research in Inflammatory Diseases, Houston, Texas 77030, and
| | - John S McMurray
- the Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
121
|
Arndt L, Dokas J, Gericke M, Kutzner CE, Müller S, Jeromin F, Thiery J, Burkhardt R. Tribbles homolog 1 deficiency modulates function and polarization of murine bone marrow-derived macrophages. J Biol Chem 2018; 293:11527-11536. [PMID: 29899113 DOI: 10.1074/jbc.ra117.000703] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/23/2018] [Indexed: 01/12/2023] Open
Abstract
Macrophages are essential for innate immunity and inflammatory responses and differentiate into various functional phenotypes. Tribbles homolog 1 (Trib1), a member of the mammalian Tribbles homolog pseudokinase family, has been implicated in regulation of cell differentiation, proliferation, and metabolism, but its role in macrophage biology has not been fully elucidated. Here, we investigated the consequences of Trib1 deficiency on macrophage functions and M1/M2 polarization. Bone marrow-derived macrophages (BMDMs) from Trib1-deficient (Trib1-/-) mice exhibited elevated phagocytic capacity, correlating with up-regulation of several scavenger receptors. Concomitantly, uptake of modified low-density lipoprotein was increased in Trib1-/- BMDMs. Trib1-/- macrophages also exhibited diminished migration in the presence of the chemokine MCP-1, associated with reduced expression of the MCP-1 receptor Ccr2 Furthermore, Trib1 deficiency attenuated the response of BMDMs to both M1 and M2 stimuli; induction of the M1-marker genes Il6, Il1b, and Nos2 upon LPS/IFNγ stimulation and of the M2-marker genes Cd206, Fizz1, and Arg1 upon IL-4 stimulation was reduced. Functionally, Trib1 deficiency decreased secretion of proinflammatory cytokines (IL-6, TNFα, IL-1β, and CXCL1) and reduced nitric oxide and reactive oxygen species production in M1-polarized macrophages. Supporting the attenuated M2 phenotype, IL-4-stimulated Trib1-/- macrophages secreted less IL-10 and TGFβ. Mechanistically, Trib1-/- BMDMs displayed lower levels of Janus kinase 1 (JAK1), resulting in reduced activation of LPS/IFNγ-mediated STAT1 signaling. Likewise, decreased levels of JAK1 along with lower activation of STAT6 and STAT3 were observed in M2-polarized Trib1-/- BMDMs. Our findings suggest that Trib1 extensively controls macrophage M1/M2 polarization via the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Lilli Arndt
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Janine Dokas
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Martin Gericke
- Institute of Anatomy, University of Leipzig, 04103 Leipzig, Germany
| | - Carl Elias Kutzner
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Silvana Müller
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Franziska Jeromin
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, 04103 Leipzig, Germany
| | - Ralph Burkhardt
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
122
|
Dong C, Fu T, Ji J, Li Z, Gu Z. The role of interleukin-4 in rheumatic diseases. Clin Exp Pharmacol Physiol 2018; 45:747-754. [PMID: 29655253 DOI: 10.1111/1440-1681.12946] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 03/31/2018] [Accepted: 04/04/2018] [Indexed: 01/10/2023]
Abstract
Rheumatism is a group of diseases, most of which are autoimmune diseases, that violate joints, bones, muscles, blood vessels and related soft tissue. As is well known, cytokines play a role in the pathogenesis of several rheumatic diseases, such as rheumatoid arthritis, spondyloarthritides, and systemic lupus erythematosus. Recently, the role of interleukin-4 (IL-4), which may participate in the mechanism of rheumatism, have been discovered. It is reported that IL-4 takes part in the regulation of T cell activation, differentiation, proliferation, and survival of different T cell types. IL-4 also has an immunomodulatory effect on B cells, mast cells, macrophages, and many cell types. A review of the literature on functions of IL-4 in rheumatic diseases is presented.
Collapse
Affiliation(s)
- Chen Dong
- School of Nursing, Nantong University, Nantong, Jiangsu Province, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Ting Fu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Juan Ji
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Zhenyu Li
- School of Nursing, Nantong University, Nantong, Jiangsu Province, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Zhifeng Gu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
123
|
Keegan AD, Zamorano J, Keselman A, Heller NM. IL-4 and IL-13 Receptor Signaling From 4PS to Insulin Receptor Substrate 2: There and Back Again, a Historical View. Front Immunol 2018; 9:1037. [PMID: 29868002 PMCID: PMC5962649 DOI: 10.3389/fimmu.2018.01037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022] Open
Abstract
In this historical perspective, written in honor of Dr. William E. Paul, we describe the initial discovery of one of the dominant substrates for tyrosine phosphorylation stimulated by IL-4. We further describe how this “IL-4-induced phosphorylated substrate” (4PS) was characterized as a member of the insulin receptor substrate (IRS) family of large adaptor proteins that link IL-4 and insulin receptors to activation of the phosphatidyl-inositol 3′ kinase pathway as well as other downstream signaling pathways. The relative contribution of the 4PS/IRS pathway to the early models of IL-4-induced proliferation and suppression of apoptosis are compared to our more recent understanding of the complex interplay between positive and negative regulatory pathways emanating from members of the IRS family that impact allergic responses.
Collapse
Affiliation(s)
- Achsah D Keegan
- Department of Microbiology and Immunology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States.,Baltimore VA Medical Center, Baltimore, MD, United States
| | - Jose Zamorano
- Unidad Investigacion, Complejo Hospitalario Universitario, Caceres, Spain
| | - Aleksander Keselman
- Department of Anesthesiology and Critical Care Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
124
|
Lee JH, Choi CS, Bae IH, Choi JK, Park YH, Park M. A novel, topical, nonsteroidal, TRPV1 antagonist, PAC-14028 cream improves skin barrier function and exerts anti-inflammatory action through modulating epidermal differentiation markers and suppressing Th2 cytokines in atopic dermatitis. J Dermatol Sci 2018; 91:S0923-1811(18)30204-4. [PMID: 29752146 DOI: 10.1016/j.jdermsci.2018.04.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND Although it is established that epidermal barrier disturbance and immune dysfunction resulting in IgE sensitization are critical factors in the development of cutaneous inflammation, the pathogenesis and targeted therapy of atopic dermatitis (AD)-specific pathways have still been unknown. OBJECTIVE Taking into account the fact that Th2 cytokines in AD have both unique and overlapping functions including increased epidermal thickening, inflammation, and decreased expressing of the barrier proteins keratinocyte differentiation, we sought to clarify our hypothesis that TRPV1 antagonist plays a critical role in skin barrier function and can be a therapeutic target for AD. METHODS AD-like dermatitis was induced in hairless mice by repeated oxazolone (Ox) challenges to hairless mice. The functional studies concerning skin barrier function, anti-inflammatory action, and molecular mechanism by TRPV1 antagonism were conducted by histopathological assays, ELISA, qPCR, western blotting, and skin blood flow measurement. RESULTS Topically administered TRPV1 antagonist, PAC-14028 (Asivatrep: C21H22F5N3O3S), improved AD-like dermatitis and skin barrier functions, and restored the expression of epidermal differentiation markers. In addition, the PAC-14028 cream significantly inhibited cutaneous inflammation by decreasing the expression of serum IgE, and the epidermal expression of IL-4, and IL-13 in Ox-AD mice. These results may provide a novel insight into the molecular mechanism of PAC-14028 cream involved in anti-inflammatory effects and skin barrier functions by suppressing the multiple signaling pathways including IL-4/-13-mediated activation of JAK/STAT, TRPV1, and neuropeptides. CONCLUSION PAC-14028 cream can be a potential therapeutic tool for the treatment of chronic inflammation and disrupted barrier function in patients with AD.
Collapse
Affiliation(s)
- Ji-Hae Lee
- Vital Beautie Research Institute, Amorepacific Corporation R&D Center, Yongin, Republic of Korea
| | - Chang Soon Choi
- Vital Beautie Research Institute, Amorepacific Corporation R&D Center, Yongin, Republic of Korea
| | - Il-Hong Bae
- Vital Beautie Research Institute, Amorepacific Corporation R&D Center, Yongin, Republic of Korea
| | - Jin Kyu Choi
- Medical Beauty QA Team, Aestura Corporation, Anseong, Republic of Korea
| | - Young-Ho Park
- Vital Beautie Research Institute, Amorepacific Corporation R&D Center, Yongin, Republic of Korea
| | - Miyoung Park
- Vital Beautie Research Institute, Amorepacific Corporation R&D Center, Yongin, Republic of Korea.
| |
Collapse
|
125
|
Maher P, Conti B. Deciphering the pathways that protect from IL-13-mediated potentiation of oxidative stress-induced dopaminergic nerve cell death. Cytokine 2018; 103:114-120. [PMID: 28969943 PMCID: PMC5808859 DOI: 10.1016/j.cyto.2017.09.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/31/2017] [Accepted: 09/18/2017] [Indexed: 01/27/2023]
Abstract
The majority of Parkinson's disease (PD) cases are sporadic with only about 10% of PD patients having a family history of the disease suggesting that this neurodegenerative disorder is the result of both environmental and genetic factors. Both oxidative stress and neuroinflammation are thought to contribute to PD. Previously, we showed that the activation of interleukin 13 receptor alpha 1 (IL-13Rα1) increases the sensitivity of dopaminergic neurons to oxidative damage both in cultured cells and in animals. In this study, we investigated the pathways involved in the IL-13-mediated potentiation of oxidative stress-induced dopaminergic cell death using a combination of cell survival assays and Western blotting with appropriate antibodies. In addition, siRNA was used to examine the role of 4E-BP1 in this cell toxicity paradigm. We show that activation of both the Jak-Stat and PI3 kinase-mTOR pathways play key roles in the promotion of cell death by IL-13 in the presence of mild oxidative stress. The Jak 1/2 inhibitor ruxolitinib, the mTOR inhibitor rapamycin and the PI3 kinase inhibitor LY294002 all prevented the potentiation of cell death by IL-13. Moreover, 4E-BP1, a target of mTOR, appeared to mediate the protective effects of rapamycin. Together, these results indicate that multiple signaling pathways downstream of IL-13Rα1 activation play a role in the toxic effects of IL-13 in dopaminergic neurons in the presence of mild oxidative stress and suggest that any of these pathways might provide potential targets for the treatment of PD.
Collapse
Affiliation(s)
- Pamela Maher
- The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, United States.
| | - Bruno Conti
- The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, United States
| |
Collapse
|
126
|
Weng SY, Wang X, Vijayan S, Tang Y, Kim YO, Padberg K, Regen T, Molokanova O, Chen T, Bopp T, Schild H, Brombacher F, Crosby JR, McCaleb ML, Waisman A, Bockamp E, Schuppan D. IL-4 Receptor Alpha Signaling through Macrophages Differentially Regulates Liver Fibrosis Progression and Reversal. EBioMedicine 2018; 29:92-103. [PMID: 29463471 PMCID: PMC5925448 DOI: 10.1016/j.ebiom.2018.01.028] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic hepatitis leads to liver fibrosis and cirrhosis. Cirrhosis is a major cause of worldwide morbidity and mortality. Macrophages play a key role in fibrosis progression and reversal. However, the signals that determine fibrogenic vs fibrolytic macrophage function remain ill defined. We studied the role of interleukin-4 receptor α (IL-4Rα), a potential central switch of macrophage polarization, in liver fibrosis progression and reversal. We demonstrate that inflammatory monocyte infiltration and liver fibrogenesis were suppressed in general IL-4Rα−/− as well as in macrophage-specific IL-4Rα−/− (IL-4RαΔLysM) mice. However, with deletion of IL-4RαΔLysM spontaneous fibrosis reversal was retarded. Results were replicated by pharmacological intervention using IL-4Rα-specific antisense oligonucleotides. Retarded resolution was linked to the loss of M2-type resident macrophages, which secreted MMP-12 through IL-4 and IL-13-mediated phospho-STAT6 activation. We conclude that IL-4Rα signaling regulates macrophage functional polarization in a context-dependent manner. Pharmacological targeting of macrophage polarization therefore requires disease stage-specific treatment strategies. Research in Context Alternative (M2-type) macrophage activation through IL-4Rα promotes liver inflammation and fibrosis progression but speeds up fibrosis reversal. This demonstrates context dependent, opposing roles of M2-type macrophages. During reversal IL-4Rα induces fibrolytic MMPs, especially MMP-12, through STAT6. Liver-specific antisense oligonucleotides efficiently block IL-4Rα expression and attenuate fibrosis progression. IL-4Rα is considered a central switch for alternative macrophage polarization. IL-4Rα on macrophages is shown to differentially regulate liver fibrosis progression vs reversal. Therapeutic IL-4Rα antisense oligonucleotides replicate these findings. MMP-12 induced via macrophage IL-4Rα is a key promoter of fibrosis reversal.
Collapse
Affiliation(s)
- Shih-Yen Weng
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Xiaoyu Wang
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Santosh Vijayan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Yilang Tang
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Yong Ook Kim
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Kornelius Padberg
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Tommy Regen
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Olena Molokanova
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Tao Chen
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Tobias Bopp
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Hansjörg Schild
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Frank Brombacher
- International Center for Genetic Engineering and Biotechnology, Institute of Infectious Disease and Molecular Medicine, South African Medical Research Council, Cape Town, South Africa
| | | | | | - Ari Waisman
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Ernesto Bockamp
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
127
|
Wojdasiewicz P, Poniatowski ŁA, Nauman P, Mandat T, Paradowska-Gorycka A, Romanowska-Próchnicka K, Szukiewicz D, Kotela A, Kubaszewski Ł, Kotela I, Kurkowska-Jastrzębska I, Gasik R. Cytokines in the pathogenesis of hemophilic arthropathy. Cytokine Growth Factor Rev 2018; 39:71-91. [DOI: 10.1016/j.cytogfr.2017.11.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 11/09/2017] [Indexed: 01/26/2023]
|
128
|
Das A, Yang CS, Arifuzzaman S, Kim S, Kim SY, Jung KH, Lee YS, Chai YG. High-Resolution Mapping and Dynamics of the Transcriptome, Transcription Factors, and Transcription Co-Factor Networks in Classically and Alternatively Activated Macrophages. Front Immunol 2018; 9:22. [PMID: 29403501 PMCID: PMC5778122 DOI: 10.3389/fimmu.2018.00022] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/04/2018] [Indexed: 12/16/2022] Open
Abstract
Macrophages are the prime innate immune cells of the inflammatory response, and the combination of multiple signaling inputs derived from the recognition of host factors [e.g., interferon-g (IFN-γ)] and invading pathogen products (e.g., toll-like receptors (TLRs) agonists) are required to maintain essential macrophage function. The profound effects on biological outcomes of inflammation associated with IFN-γ pretreatment (“priming”) and TLR4 ligand bacterial lipopolysaccharide (LPS)-induced macrophage activation (M1 or classical activation) have long been recognized, but the underlying mechanisms are not well defined. Therefore, we analyzed gene expression profiles of macrophages and identified genes, transcription factors (TFs), and transcription co-factors (TcoFs) that are uniquely or highly expressed in IFN-γ-mediated TLR4 ligand LPS-inducible versus only TLR4 ligand LPS-inducible primary macrophages. This macrophage gene expression has not been observed in macrophage cell lines. We also showed that interleukin (IL)-4 and IL-13 (M2 or alternative activation) elicited the induction of a distinct subset of genes related to M2 macrophage polarization. Importantly, this macrophage gene expression was also associated with promoter conservation. In particular, our approach revealed novel roles for the TFs and TcoFs in response to inflammation. We believe that the systematic approach presented herein is an important framework to better understand the transcriptional machinery of different macrophage subtypes.
Collapse
Affiliation(s)
- Amitabh Das
- Institute of Natural Science and Technology, Hanyang University, Ansan, South Korea
| | - Chul-Su Yang
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea.,Department of Bionanotechnology, Hanyang University, Seoul, South Korea
| | | | - Sojin Kim
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea
| | - Sun Young Kim
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea.,Department of Bionanotechnology, Hanyang University, Seoul, South Korea
| | - Kyoung Hwa Jung
- Institute of Natural Science and Technology, Hanyang University, Ansan, South Korea
| | - Young Seek Lee
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea
| | - Young Gyu Chai
- Department of Molecular and Life Sciences, Hanyang University, Ansan, South Korea.,Department of Bionanotechnology, Hanyang University, Seoul, South Korea
| |
Collapse
|
129
|
Pradel LP, Franke A, Ries CH. Effects of IL-10 and Th
2 cytokines on human Mφ phenotype and response to CSF1R inhibitor. J Leukoc Biol 2018; 103:545-558. [DOI: 10.1002/jlb.5ma0717-282r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/30/2017] [Accepted: 10/22/2017] [Indexed: 12/19/2022] Open
Affiliation(s)
- Leon P. Pradel
- Discovery Oncology; Roche Innovation Center Munich; Penzberg Germany
- Natural Sciences Faculty; Paris Lodron University Salzburg; Salzburg Austria
| | - Andreas Franke
- Large Molecule Research; Roche Innovation Center Munich; Penzberg Germany
| | - Carola H. Ries
- Discovery Oncology; Roche Innovation Center Munich; Penzberg Germany
| |
Collapse
|
130
|
Wang T, Johansson P, Abós B, Holt A, Tafalla C, Jiang Y, Wang A, Xu Q, Qi Z, Huang W, Costa MM, Diaz-Rosales P, Holland JW, Secombes CJ. First in-depth analysis of the novel Th2-type cytokines in salmonid fish reveals distinct patterns of expression and modulation but overlapping bioactivities. Oncotarget 2017; 7:10917-46. [PMID: 26870894 PMCID: PMC4905449 DOI: 10.18632/oncotarget.7295] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/24/2016] [Indexed: 12/12/2022] Open
Abstract
IL-4 and IL-13 are closely related canonical type-2 cytokines in mammals and have overlapping bioactivities via shared receptors. They are frequently activated together as part of the same immune response and are the signature cytokines produced by T-helper (Th)2 cells and type-2 innate lymphoid cells (ILC2), mediating immunity against extracellular pathogens. Little is known about the origin of type-2 responses, and whether they were an essential component of the early adaptive immune system that gave a fitness advantage by limiting collateral damage caused by metazoan parasites. Two evolutionary related type-2 cytokines, IL-4/13A and IL-4/13B, have been identified recently in several teleost fish that likely arose by duplication of an ancestral IL-4/13 gene as a consequence of a whole genome duplication event that occurred at the base of this lineage. However, studies of their comparative expression levels are largely missing and bioactivity analysis has been limited to IL-4/13A in zebrafish. Through interrogation of the recently released salmonid genomes, species in which an additional whole genome duplication event has occurred, four genomic IL-4/13 loci have been identified leading to the cloning of three active genes, IL-4/13A, IL-4/13B1 and IL-4/13B2, in both rainbow trout and Atlantic salmon. Comparative expression analysis by real-time PCR in rainbow trout revealed that the IL-4/13A expression is broad and high constitutively but less responsive to pathogen-associated molecular patterns (PAMPs) and pathogen challenge. In contrast, the expression of IL-4/13B1 and IL-4/13B2 is low constitutively but is highly induced by viral haemorrhagic septicaemia virus (VHSH) infection and during proliferative kidney disease (PKD) in vivo, and by formalin-killed bacteria, PAMPs, the T cell mitogen PHA, and the T-cell cytokines IL-2 and IL-21 in vitro. Moreover, bioactive recombinant cytokines of both IL-4/13A and B were produced and found to have shared but also distinct bioactivities. Both cytokines rapidly induce the gene expression of antimicrobial peptides and acute phase proteins, providing an effector mechanism of fish type-2 cytokines in immunity. They are anti-inflammatory via up-regulation of IL-10 and down-regulation of IL-1β and IFN-γ. They modulate the expression of cellular markers of T cells, macrophages and B cells, the receptors of IFN-γ, the IL-6 cytokine family and their own potential receptors, suggesting multiple target cells and important roles of fish type-2 cytokines in the piscine cytokine network. Furthermore both cytokines increased the number of IgM secreting B cells but had no effects on the proliferation of IgM+ B cells in vitro. Taken as a whole, fish IL-4/13A may provide a basal level of type-2 immunity whilst IL-4/13B, when activated, provides an enhanced type-2 immunity, which may have an important role in specific cell-mediated immunity. To our knowledge this is the first in-depth analysis of the expression, modulation and bioactivities of type-2 cytokines in the same fish species, and in any early vertebrate. It contributes to a broader understanding of the evolution of type-2 immunity in vertebrates, and establishes a framework for further studies and manipulation of type-2 cytokines in fish.
Collapse
Affiliation(s)
- Tiehui Wang
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Petronella Johansson
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Beatriz Abós
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos (Madrid), Spain
| | - Amy Holt
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Carolina Tafalla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos (Madrid), Spain
| | - Youshen Jiang
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK.,College of Fishery and Life Science, Shanghai Ocean University, Shanghai, China
| | - Alex Wang
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Qiaoqing Xu
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK.,School of Animal Science, Yangtze University, Jingzhou, Hubei Province, China
| | - Zhitao Qi
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK.,Central Laboratory of Biology, Chemical and Biological Engineering College, Yancheng Institute of Technology, Yancheng, Jiangsu Province, China
| | - Wenshu Huang
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK.,Fisheries College, Jimei University, Xiamen, Fujian Province, China
| | - Maria M Costa
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK.,Instituto de Investigaciones Marinas, Consejo Superior de Investigaciones Científicas (CSIC), Vigo, Spain
| | - Patricia Diaz-Rosales
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Jason W Holland
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Christopher J Secombes
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
131
|
Lee SE, Kang SG, Choi MJ, Jung SB, Ryu MJ, Chung HK, Chang JY, Kim YK, Lee JH, Kim KS, Kim HJ, Lee HK, Yi HS, Shong M. Growth Differentiation Factor 15 Mediates Systemic Glucose Regulatory Action of T-Helper Type 2 Cytokines. Diabetes 2017; 66:2774-2788. [PMID: 28874416 DOI: 10.2337/db17-0333] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/24/2017] [Indexed: 11/13/2022]
Abstract
T-helper type 2 (Th2) cytokines, including interleukin (IL)-13 and IL-4, produced in adipose tissue, are critical regulators of intra-adipose and systemic lipid and glucose metabolism. Furthermore, IL-13 is a potential therapy for insulin resistance in obese mouse models. Here, we examined mediators produced by adipocytes that are responsible for regulating systemic glucose homeostasis in response to Th2 cytokines. We used RNA sequencing data analysis of cultured adipocytes to screen factors secreted in response to recombinant IL-13. Recombinant IL-13 induced expression of growth differentiation factor 15 (GDF15) via the Janus kinase-activated STAT6 pathway. In vivo administration of α-galactosylceramide or IL-33 increased IL-4 and IL-13 production, thereby increasing GDF15 levels in adipose tissue and in plasma of mice; however, these responses were abrogated in STAT6 knockout mice. Moreover, administration of recombinant IL-13 to wild-type mice fed a high-fat diet (HFD) improved glucose intolerance; this was not the case for GDF15 knockout mice fed the HFD. Taken together, these data suggest that GDF15 is required for IL-13-induced improvement of glucose intolerance in mice fed an HFD. Thus, beneficial effects of Th2 cytokines on systemic glucose metabolism and insulin sensitivity are mediated by GDF15. These findings open up a potential pharmacological route for reversing insulin resistance associated with obesity.
Collapse
Affiliation(s)
- Seong Eun Lee
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Seul Gi Kang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Min Jeong Choi
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Saet-Byel Jung
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyo Kyun Chung
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
| | - Joon Young Chang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Yong Kyung Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
| | - Ju Hee Lee
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
| | - Koon Soon Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyun Jin Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Hyon-Seung Yi
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
132
|
Hassanbhai AM, Lau CS, Wen F, Jayaraman P, Goh BT, Yu N, Teoh SH. In Vivo Immune Responses of Cross-Linked Electrospun Tilapia Collagen Membrane. Tissue Eng Part A 2017; 23:1110-1119. [DOI: 10.1089/ten.tea.2016.0504] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Ammar Mansoor Hassanbhai
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Chau Sang Lau
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Feng Wen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Praveena Jayaraman
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Bee Tin Goh
- National Dental Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School Singapore, Singapore, Singapore
| | - Na Yu
- National Dental Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School Singapore, Singapore, Singapore
| | - Swee-Hin Teoh
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
133
|
Nakai K, He YY, Nishiyama F, Naruse F, Haba R, Kushida Y, Katsuki N, Moriue T, Yoneda K, Kubota Y. IL-17A induces heterogeneous macrophages, and it does not alter the effects of lipopolysaccharides on macrophage activation in the skin of mice. Sci Rep 2017; 7:12473. [PMID: 28963556 PMCID: PMC5622065 DOI: 10.1038/s41598-017-12756-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/14/2017] [Indexed: 01/09/2023] Open
Abstract
Macrophages are central to inflammatory response and become polarized towards the M1 or M2 states upon activation by immunostimulants. In this study, we investigated the effects of lipopolysaccharides (LPS) and interleukin (IL)-17A on the activation of macrophages in in vivo mouse skin. We examined whether macrophages are activated in the skin of imiquimod (IMQ)-treated mice, a model for IL-17A-induced psoriasis-like skin inflammation, and flaky-tail (Flgft) mice, a model for IL-17A-induced chronic atopic dermatitis-like skin inflammation. LPS and IL-17A independently increased the expression levels of iNOS, CX3CR1, CD206, phospho-STAT1 and phospho-STAT3 proteins in the skin of B6 mice, and the effects of LPS was not altered by IL-17A. The expression levels of these proteins were increased in the skin of IMQ-treated and Flgft mice. IL-17A neutralization increased the expressions of iNOS and phospho-STAT1 in the IMQ-treated skin, but it decreased the expressions of CD206 and phospho-STAT3 proteins in the skin of Flgft mice, suggesting that macrophages to change from the M2 to the M1 state in the skin of these mice. These results suggest that IL-17A is involved in the activation of macrophages that are in the process of adopting the heterogeneous profiles of both the M1 and M2 states.
Collapse
Affiliation(s)
- Kozo Nakai
- Department of Dermatology, Kagawa University, Kagawa, Japan.
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, USA
| | | | - Fukiko Naruse
- Department of Dermatology, Kagawa University, Kagawa, Japan
| | - Reiji Haba
- Department of Diagnostic Pathology, Kagawa University, Kagawa, Japan
| | - Yoshio Kushida
- Department of Diagnostic Pathology, Kagawa University, Kagawa, Japan
| | - Naomi Katsuki
- Department of Diagnostic Pathology, Kagawa University, Kagawa, Japan
| | - Tetsuya Moriue
- Department of Dermatology, Kagawa University, Kagawa, Japan
| | - Kozo Yoneda
- Department of Pharmacology, Osaka Ohtani University, Osaka, Japan
| | - Yasuo Kubota
- Department of Dermatology, Kagawa University, Kagawa, Japan
| |
Collapse
|
134
|
SOCS molecules: the growing players in macrophage polarization and function. Oncotarget 2017; 8:60710-60722. [PMID: 28948005 PMCID: PMC5601173 DOI: 10.18632/oncotarget.19940] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023] Open
Abstract
The concept of macrophage polarization is defined in terms of macrophage phenotypic heterogeneity and functional diversity. Cytokines signals are thought to be required for the polarization of macrophage populations toward different phenotypes at different stages in development, homeostasis and disease. The suppressors of cytokine signaling family of proteins contribute to the magnitude and duration of cytokines signaling, which ultimately control the subtle adjustment of the balance between divergent macrophage phenotypes. This review highlights the specific roles and mechanisms of various cytokines family and their negative regulators link to the macrophage polarization programs. Eventually, breakthrough in the identification of these molecules will provide the novel therapeutic approaches for a host of diseases by targeting macrophage phenotypic shift.
Collapse
|
135
|
Evolution of Th2 responses: characterization of IL-4/13 in sea bass (Dicentrarchus labrax L.) and studies of expression and biological activity. Sci Rep 2017; 7:2240. [PMID: 28533556 PMCID: PMC5440397 DOI: 10.1038/s41598-017-02472-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/11/2017] [Indexed: 02/07/2023] Open
Abstract
Th2 immunity is a primary host defence against metazoan pathogens and two of the important cytokines involved in this immune response in mammals are IL-4 and IL-13. Recently the origin and evolution of Th2 immune responses have been investigated in fish where a molecule with relatedness to both IL-4 and IL-13 is present, termed IL-4/13. Different IL-4/13 paralogues (IL-4/13 A and IL-4/13B) exist in teleost fish. In this paper, we have focused on the IL-4/13 isoforms found in the European sea bass (Dicentrarchus labrax L.). Two tandem duplicated but divergent IL-4/13 A isoforms and one IL-4/13B are present, a unique situation compared to other teleosts. These genes were studied in terms of their in vitro and in vivo transcript levels after different treatments and their biological activities after production of the recombinant isoforms. The results show that the presence of these three paralogues is associated with different activities, both in terms of their expression profiles and the ability of the proteins to modulate the expression of immune genes in head kidney leukocytes. It is clear that the initiation and control of type-2 responses in seabass is complex due to the presence of multiple IL-4/13 isoforms with overlapping but distinct activities.
Collapse
|
136
|
Ramani M, Mudge MC, Morris RT, Zhang Y, Warcholek SA, Hurst MN, Riviere JE, DeLong RK. Zinc Oxide Nanoparticle-Poly I:C RNA Complexes: Implication as Therapeutics against Experimental Melanoma. Mol Pharm 2017; 14:614-625. [PMID: 28135100 DOI: 10.1021/acs.molpharmaceut.6b00795] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
There is current interest in harnessing the combined anticancer and immunological effect of nanoparticles (NPs) and RNA. Here, we evaluate the bioactivity of poly I:C (pIC) RNA, bound to anticancer zinc oxide NP (ZnO-NP) against melanoma. Direct RNA association to unfunctionalized ZnO-NP is shown by observing change in size, zeta potential, and absorption/fluorescence spectra upon complexation. RNA corona was visualized by transmission electron microscopy (TEM) for the first time. Binding constant (Kb = 1.6-2.8 g-1 L) was determined by modified Stern-Volmer, absorption, and biological surface activity index analysis. The pIC-ZnO-NP complex increased cell death for both human (A375) and mouse (B16F10) cell lines and suppressed tumor cell growth in BALB/C-B16F10 mouse melanoma model. Ex vivo tumor analysis indicated significant molecular activity such as changes in the level of phosphoproteins JNK, Akt, and inflammation markers IL-6 and IFN-γ. High throughput proteomics analysis revealed zinc oxide and poly I:C-specific and combinational patterns that suggested possible utility as an anticancer and immunotherapeutic strategy against melanoma.
Collapse
Affiliation(s)
| | - Miranda C Mudge
- Department of Biomedical Science, Missouri State University , Springfield, Missouri 65897, United States
| | - R Tyler Morris
- Department of Biomedical Science, Missouri State University , Springfield, Missouri 65897, United States
| | | | | | - Miranda N Hurst
- Department of Biomedical Science, Missouri State University , Springfield, Missouri 65897, United States
| | | | - Robert K DeLong
- Department of Biomedical Science, Missouri State University , Springfield, Missouri 65897, United States
| |
Collapse
|
137
|
Kalish S, Lyamina S, Manukhina E, Malyshev Y, Raetskaya A, Malyshev I. M3 Macrophages Stop Division of Tumor Cells In Vitro and Extend Survival of Mice with Ehrlich Ascites Carcinoma. Med Sci Monit Basic Res 2017; 23:8-19. [PMID: 28123171 PMCID: PMC5291087 DOI: 10.12659/msmbr.902285] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background M1 macrophages target tumor cells. However, many tumors produce anti-inflammatory cytokines, which reprogram the anti-tumor M1 macrophages into the pro-tumor M2 macrophages. We have hypothesized that the problem of pro-tumor macrophage reprogramming could be solved by using a special M3 switch phenotype. The M3 macrophages, in contrast to the M1 macrophages, should respond to anti-inflammatory cytokines by increasing production of pro-inflammatory cytokines to retain its anti-tumor properties. Objectives of the study were to form an M3 switch phenotype in vitro and to evaluate the effect of M3 macrophages on growth of Ehrlich ascites carcinoma (EAC) in vitro and in vivo. Material/Methods Tumor growth was initiated by an intraperitoneal injection of EAC cells into C57BL/6J mice. Results 1) The M3 switch phenotype can be programed by activation of M1-reprogramming pathways with simultaneous inhibition of the M2 phenotype transcription factors, STAT3, STAT6, and/or SMAD3. 2) M3 macrophages exerted an anti-tumor effect both in vitro and in vivo, which was superior to anti-tumor effects of cisplatin or M1 macrophages. 3) The anti-tumor effect of M3 macrophages was due to their anti-proliferative effect. Conclusions Development of new biotechnologies for restriction of tumor growth using in vitro reprogrammed M3 macrophages is very promising.
Collapse
Affiliation(s)
- Sergey Kalish
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry n.a. A.I. Evdokimov, Moscow, Russian Federation
| | - Svetlana Lyamina
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry n.a. A.I. Evdokimov, Moscow, Russian Federation
| | - Eugenia Manukhina
- Department of Stress and Adaptation, Institute of General Pathology and Pathophysiology, Moscow, Russian Federation.,University of North Texas Health Science Center, Fort Worth, TX, USA.,South Ural State University Biomedical School, Chelyabinsk, Russian Federation
| | | | - Anastasiya Raetskaya
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry n.a. A.I. Evdokimov, Moscow, Russian Federation
| | - Igor Malyshev
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry n.a. A.I. Evdokimov, Moscow, Russian Federation.,Department of Stress and Adaptation, Institute of General Pathology and Pathophysiology, Moscow, Russian Federation.,University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
138
|
The role of PTEN in regulation of hepatic macrophages activation and function in progression and reversal of liver fibrosis. Toxicol Appl Pharmacol 2017; 317:51-62. [PMID: 28095306 DOI: 10.1016/j.taap.2017.01.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/02/2016] [Accepted: 01/12/2017] [Indexed: 12/25/2022]
Abstract
Activation of Kupffer cells (KCs) plays a pivotal role in the pathogenesis of liver fibrosis. The progression and reversal of CCl4-induced mouse liver fibrosis showed a mixed induction of hepatic classical (M1) and alternative (M2) macrophage markers. Although the role of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in modulating myeloid cell activation has recently been identified, its function in macrophage activation during hepatic fibrosis remains to be fully appreciated. In our study, PTEN expression of KCs was remarkably decreased in CCl4-induced mice but increased to a near-normal level in reversed mice. Moreover, PTEN was significantly decreased in IL4-induced RAW 264.7 cells in vitro and lower expression of PTEN was observed in M2 macrophages in vivo. In addition, loss- and gain-of-function studies suggested that PTEN regulates M2 macrophages polarization via activation of PI3K/Akt/STAT6 signaling, but had a limited effect on M1 macrophages polarization in vitro. Additionally, Ly294002, a chemical inhibitor of PI3K/Akt, could dramatically down-regulate the hallmarks of M2 macrophages. In conclusion, PTEN mediates macrophages activation by PI3K/Akt/STAT6 signaling pathway, which provides novel compelling evidences on the potential of PTEN in liver injury and opens new cellular target for the pharmacological therapy of liver fibrosis.
Collapse
|
139
|
Sharma J, Bhar S, Devi CS. A review on interleukins: The key manipulators in rheumatoid arthritis. Mod Rheumatol 2017; 27:723-746. [DOI: 10.1080/14397595.2016.1266071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jatin Sharma
- School of Biosciences and Technology, VIT University, Vellore, India
| | - Sutonuka Bhar
- School of Biosciences and Technology, VIT University, Vellore, India
| | - C. Subathra Devi
- School of Biosciences and Technology, VIT University, Vellore, India
| |
Collapse
|
140
|
Ayala-Lopez N, Watts SW. New actions of an old friend: perivascular adipose tissue's adrenergic mechanisms. Br J Pharmacol 2016; 174:3454-3465. [PMID: 27813085 DOI: 10.1111/bph.13663] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/03/2016] [Accepted: 10/21/2016] [Indexed: 12/17/2022] Open
Abstract
The revolutionary discovery in 1991 by Soltis and Cassis that perivascular adipose tissue (PVAT) has an anti-contractile effect changed how we think about the vasculature. Most experiments on vascular pharmacology begin by removing the fat surrounding vessels. Thus, PVAT was thought to have a minor role in vascular function and its presence was just for structural support. The need to rethink PVAT's role was precipitated by observations that obesity carries a high cardiovascular risk and PVAT dysfunction is associated with obesity. PVAT is a vascular-adipose organ that has intimate connections with the nervous and immune system. A complex world of physiology resides in PVAT, including the presence of an 'adrenergic system' that is able to release, take up and metabolize noradrenaline. Adipocytes, stromal vascular cells and nerves within PVAT contain components that make up this adrenergic system. Some of the great strides in PVAT research came from studying adipose tissue as a whole. Adipose tissue has many roles and participates in regulating energy balance, energy stores, inflammation and thermoregulation. However, PVAT is dissimilar from non-PVAT adipose tissues. PVAT is intimately connected with the vasculature, which is what makes its role in body homeostasis unique. The adrenergic system within PVAT may be an integral link connecting the effects of obesity with the vascular dysfunction observed in obesity-associated hypertension, a condition in which the sympathetic nervous system has a significant role. This review will explore what is known about the adrenergic system in adipose tissue and PVAT, plus the translational importance of these findings. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- Nadia Ayala-Lopez
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
141
|
Signal transducer and activator of transcription proteins: regulators of myeloid-derived suppressor cell-mediated immunosuppression in cancer. Arch Pharm Res 2016; 39:1597-1608. [DOI: 10.1007/s12272-016-0822-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/18/2016] [Indexed: 12/31/2022]
|
142
|
McCormick SM, Gowda N, Fang JX, Heller NM. Suppressor of Cytokine Signaling (SOCS)1 Regulates Interleukin-4 (IL-4)-activated Insulin Receptor Substrate (IRS)-2 Tyrosine Phosphorylation in Monocytes and Macrophages via the Proteasome. J Biol Chem 2016; 291:20574-87. [PMID: 27507812 DOI: 10.1074/jbc.m116.746164] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Indexed: 11/06/2022] Open
Abstract
Allergic asthma is a chronic lung disease initiated and driven by Th2 cytokines IL-4/-13. In macrophages, IL-4/-13 bind IL-4 receptors, which signal through insulin receptor substrate (IRS)-2, inducing M2 macrophage differentiation. M2 macrophages correlate with disease severity and poor lung function, although the mechanisms that regulate M2 polarization are not understood. Following IL-4 exposure, suppressor of cytokine signaling (SOCS)1 is highly induced in human monocytes. We found that siRNA knockdown of SOCS1 prolonged IRS-2 tyrosine phosphorylation and enhanced M2 differentiation, although siRNA knockdown of SOCS3 did not affect either. By co-immunoprecipitation, we found that SOCS1 complexes with IRS-2 at baseline, and this association increased after IL-4 stimulation. Because SOCS1 is an E3 ubiquitin ligase, we examined the effect of proteasome inhibitors on IL-4-induced IRS-2 phosphorylation. Proteasomal inhibition prolonged IRS-2 tyrosine phosphorylation, increased ubiquitination of IRS-2, and enhanced M2 gene expression. siRNA knockdown of SOCS1 inhibited ubiquitin accumulation on IRS-2, although siRNA knockdown of SOCS3 had no effect on ubiquitination of IRS-2. Monocytes from healthy and allergic individuals revealed that SOCS1 is induced by IL-4 in healthy monocytes but not allergic cells, whereas SOCS3 is highly induced in allergic monocytes. Healthy monocytes displayed greater ubiquitination of IRS-2 and lower M2 polarization than allergic monocytes in response to IL-4 stimulation. Here, we identify SOCS1 as a key negative regulator of IL-4-induced IRS-2 signaling and M2 differentiation. Our findings provide novel insight into how dysregulated expression of SOCS increases IL-4 responses in allergic monocytes, and this may represent a new therapeutic avenue for managing allergic disease.
Collapse
Affiliation(s)
- Sarah M McCormick
- From the Department of Anesthesiology and Critical Care Medicine and
| | - Nagaraj Gowda
- From the Department of Anesthesiology and Critical Care Medicine and
| | - Jessie X Fang
- From the Department of Anesthesiology and Critical Care Medicine and
| | - Nicola M Heller
- From the Department of Anesthesiology and Critical Care Medicine and Division of Allergy and Clinical Immunology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
143
|
Ebenezer AJ, Arunachalam P, Elden BT. H4R activation utilizes distinct signaling pathways for the production of RANTES and IL-13 in human mast cells. J Recept Signal Transduct Res 2016; 37:133-140. [DOI: 10.1080/10799893.2016.1203938] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Angel Jemima Ebenezer
- Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur, Tamil Nadu, India
| | - Prema Arunachalam
- Department of Pediatrics, SRM Medical College Hospital and Research Centre, Kattankulathur, Tamil Nadu, India
| | - Berla Thangam Elden
- Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
144
|
Neuroimmunology of the Interleukins 13 and 4. Brain Sci 2016; 6:brainsci6020018. [PMID: 27304970 PMCID: PMC4931495 DOI: 10.3390/brainsci6020018] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 12/20/2022] Open
Abstract
The cytokines interleukin 13 and 4 share a common heterodimeric receptor and are important modulators of peripheral allergic reactions. Produced primarily by T-helper type 2 lymphocytes, they are typically considered as anti-inflammatory cytokines because they can downregulate the synthesis of T-helper type 1 pro-inflammatory cytokines. Their presence and role in the brain is only beginning to be investigated and the data collected so far shows that these molecules can be produced by microglial cells and possibly by neurons. Attention has so far been given to the possible role of these molecules in neurodegeneration. Both neuroprotective or neurotoxic effects have been proposed based on evidence that interleukin 13 and 4 can reduce inflammation by promoting the M2 microglia phenotype and contributing to the death of microglia M1 phenotype, or by potentiating the effects of oxidative stress on neurons during neuro-inflammation. Remarkably, the heterodimeric subunit IL-13Rα1 of their common receptor was recently demonstrated in dopaminergic neurons of the ventral tegmental area and the substantia nigra pars compacta, suggesting the possibility that both cytokines may affect the activity of these neurons regulating reward, mood, and motor coordination. In mice and man, the gene encoding for IL-13Rα1 is expressed on the X chromosome within the PARK12 region of susceptibility to Parkinson’s disease (PD). This, together with finding that IL-13Rα1 contributes to loss of dopaminergic neurons during inflammation, indicates the possibility that these cytokines may contribute to the etiology or the progression of PD.
Collapse
|
145
|
Targeting the JAK-STAT pathway in the treatment of 'Th2-high' severe asthma. Future Med Chem 2016; 8:405-19. [PMID: 26934038 DOI: 10.4155/fmc.16.4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Severe asthma is a heterogeneous disease characterized by reversible airway obstruction, chronic inflammation and airway remodeling. Phenotyping and/or endotyping can lead to a more personalized treatment strategy, improving the efficacy of novel drugs. Atopic asthma is associated with high levels of Th2 cells, implicated in a number of inflammatory responses. Differentiation of these cells from naive T cells occurs primarily via the JAK-STAT signaling pathway. Targeting this pathway through inhibition of activating cytokines (IL-4 and IL-13) and their receptors, the JAKs or the STATs, has been shown to have a therapeutic effect on asthma pathology. There are a number of novel drugs currently in development, which target various pathway components; these include both biologics and small molecules at various stages of development.
Collapse
|
146
|
Leung G, Petri B, Reyes JL, Wang A, Iannuzzi J, McKay DM. Cryopreserved Interleukin-4-Treated Macrophages Attenuate Murine Colitis in an Integrin β7 - Dependent Manner. Mol Med 2015; 21:924-936. [PMID: 26701314 DOI: 10.2119/molmed.2015.00193] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/08/2015] [Indexed: 01/15/2023] Open
Abstract
The adoptive transfer of alternatively activated macrophages (AAMs) has proven to attenuate inflammation in multiple mouse models of colitis; however, the effect of cryopreservation on AAMs, the ability of previously frozen AAMs to block dinitrobenzene sulfonic acid (DNBS) (Th1) and oxazolone (Th2) colitis and their migration postinjection remains unknown. Here we have found that while cryopreservation reduced mRNA expression of canonical markers of interleukin (IL)-4-treated macrophages [M(IL-4)], this step did not translate to reduced protein or activity, and the cells retained their capacity to drive the suppression of colitis. The anticolitic effect of M(IL-4) adoptive transfer required neither T or B cell nor peritoneal macrophages in the recipient. After injection into the peritoneal cavity, M(IL-4)s migrated to the spleen, mesenteric lymph nodes and colon of DNBS-treated mice. The chemokines CCL2, CCL4 and CX3CL1 were expressed in the colon during the course of DNBS-induced colitis. The expression of integrin β7 on transferred M(IL-4)s was required for their anticolitic effect, whereas the presence of the chemokine receptors CCR2 and CX3CR1 were dispensable in this model. Collectively, the data show that M(IL-4)s can be cryopreserved M(IL-4)s and subsequently used to suppress colitis in an integrin β7-dependent manner, and we suggest that these proof-of-concept studies may lead to new cellular therapies for human inflammatory bowel disease.
Collapse
Affiliation(s)
- Gabriella Leung
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Björn Petri
- Mouse Phenomics Resource Laboratory, Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - José Luis Reyes
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Arthur Wang
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jordan Iannuzzi
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Derek M McKay
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
147
|
Leitner NR, Witalisz-Siepracka A, Strobl B, Müller M. Tyrosine kinase 2 - Surveillant of tumours and bona fide oncogene. Cytokine 2015; 89:209-218. [PMID: 26631911 DOI: 10.1016/j.cyto.2015.10.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 10/29/2015] [Indexed: 12/16/2022]
Abstract
Tyrosine kinase 2 (TYK2) is a member of the Janus kinase (JAK) family, which transduces cytokine and growth factor signalling. Analysis of TYK2 loss-of-function revealed its important role in immunity to infection, (auto-) immunity and (auto-) inflammation. TYK2-deficient patients unravelled high similarity between mice and men with respect to cellular signalling functions and basic immunology. Genome-wide association studies link TYK2 to several autoimmune and inflammatory diseases as well as carcinogenesis. Due to its cytokine signalling functions TYK2 was found to be essential in tumour surveillance. Lately TYK2 activating mutants and fusion proteins were detected in patients diagnosed with leukaemic diseases suggesting that TYK2 is a potent oncogene. Here we review the cell intrinsic and extrinsic functions of TYK2 in the characteristics preventing and enabling carcinogenesis. In addition we describe an unexpected function of kinase-inactive TYK2 in tumour rejection.
Collapse
Affiliation(s)
- Nicole R Leitner
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Agnieszka Witalisz-Siepracka
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria.
| |
Collapse
|
148
|
Ivanov I, Kuhn H, Heydeck D. Structural and functional biology of arachidonic acid 15-lipoxygenase-1 (ALOX15). Gene 2015; 573:1-32. [PMID: 26216303 PMCID: PMC6728142 DOI: 10.1016/j.gene.2015.07.073] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/26/2015] [Accepted: 07/21/2015] [Indexed: 12/14/2022]
Abstract
Lipoxygenases (LOX) form a family of lipid peroxidizing enzymes, which have been implicated in a number of physiological processes and in the pathogenesis of inflammatory, hyperproliferative and neurodegenerative diseases. They occur in two of the three domains of terrestrial life (bacteria, eucarya) and the human genome involves six functional LOX genes, which encode for six different LOX isoforms. One of these isoforms is ALOX15, which has first been described in rabbits in 1974 as enzyme capable of oxidizing membrane phospholipids during the maturational breakdown of mitochondria in immature red blood cells. During the following decades ALOX15 has extensively been characterized and its biological functions have been studied in a number of cellular in vitro systems as well as in various whole animal disease models. This review is aimed at summarizing the current knowledge on the protein-chemical, molecular biological and enzymatic properties of ALOX15 in various species (human, mouse, rabbit, rat) as well as its implication in cellular physiology and in the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Igor Ivanov
- Institute of Biochemistry, Charité - University Medicine Berlin, Charitéplatz 1, CCO-Building, Virchowweg 6, D-10117 Berlin, Germany
| | - Hartmut Kuhn
- Institute of Biochemistry, Charité - University Medicine Berlin, Charitéplatz 1, CCO-Building, Virchowweg 6, D-10117 Berlin, Germany.
| | - Dagmar Heydeck
- Institute of Biochemistry, Charité - University Medicine Berlin, Charitéplatz 1, CCO-Building, Virchowweg 6, D-10117 Berlin, Germany
| |
Collapse
|
149
|
Current Concept and Update of the Macrophage Plasticity Concept: Intracellular Mechanisms of Reprogramming and M3 Macrophage "Switch" Phenotype. BIOMED RESEARCH INTERNATIONAL 2015; 2015:341308. [PMID: 26366410 PMCID: PMC4561113 DOI: 10.1155/2015/341308] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/18/2015] [Accepted: 07/22/2015] [Indexed: 12/17/2022]
Abstract
Macrophages play a key role in immunity. In this review, we consider the traditional notion of macrophage plasticity, data that do not fit into existing concepts, and a hypothesis for existence of a new switch macrophage phenotype. Depending on the microenvironment, macrophages can reprogram their phenotype toward the proinflammatory M1 phenotype or toward the anti-inflammatory M2 phenotype. Macrophage reprogramming involves well-coordinated changes in activities of signalling and posttranslational mechanisms. Macrophage reprogramming is provided by JNK-, PI3K/Akt-, Notch-, JAK/STAT-, TGF-β-, TLR/NF-κB-, and hypoxia-dependent pathways. Posttranscriptional regulation is based on micro-mRNA. We have hypothesized that, in addition to the M1 and M2 phenotypes, an M3 switch phenotype exists. This switch phenotype responds to proinflammatory stimuli with reprogramming towards the anti-inflammatory M2 phenotype or, contrarily, it responds to anti-inflammatory stimuli with reprogramming towards the proinflammatory M1 phenotype. We have found signs of such a switch phenotype in lung diseases. Understanding the mechanisms of macrophage reprogramming will assist in the selection of new therapeutic targets for correction of impaired immunity.
Collapse
|
150
|
Namgaladze D, Snodgrass RG, Angioni C, Grossmann N, Dehne N, Geisslinger G, Brüne B. AMP-activated protein kinase suppresses arachidonate 15-lipoxygenase expression in interleukin 4-polarized human macrophages. J Biol Chem 2015; 290:24484-94. [PMID: 26276392 DOI: 10.1074/jbc.m115.678243] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Indexed: 01/20/2023] Open
Abstract
Macrophages respond to the Th2 cytokine IL-4 with elevated expression of arachidonate 15-lipoxygenase (ALOX15). Although IL-4 signaling elicits anti-inflammatory responses, 15-lipoxygenase may either support or inhibit inflammatory processes in a context-dependent manner. AMP-activated protein kinase (AMPK) is a metabolic sensor/regulator that supports an anti-inflammatory macrophage phenotype. How AMPK activation is linked to IL-4-elicited gene signatures remains unexplored. Using primary human macrophages stimulated with IL-4, we observed elevated ALOX15 mRNA and protein expression, which was attenuated by AMPK activation. AMPK activators, e.g. phenformin and aminoimidazole-4-carboxamide 1-β-d-ribofuranoside inhibited IL-4-evoked activation of STAT3 while leaving activation of STAT6 and induction of typical IL-4-responsive genes intact. In addition, phenformin prevented IL-4-induced association of STAT6 and Lys-9 acetylation of histone H3 at the ALOX15 promoter. Activating AMPK abolished cellular production of 15-lipoxygenase arachidonic acid metabolites in IL-4-stimulated macrophages, which was mimicked by ALOX15 knockdown. Finally, pretreatment of macrophages with IL-4 for 48 h increased the mRNA expression of the proinflammatory cytokines IL-6, IL-12, CXCL9, and CXCL10 induced by subsequent stimulation with lipopolysaccharide. This response was attenuated by inhibition of ALOX15 or activation of AMPK during incubation with IL-4. In conclusion, limiting ALOX15 expression by AMPK may promote an anti-inflammatory phenotype of IL-4-stimulated human macrophages.
Collapse
Affiliation(s)
| | | | - Carlo Angioni
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Nina Grossmann
- From the Institute of Biochemistry I, Faculty of Medicine and
| | - Nathalie Dehne
- From the Institute of Biochemistry I, Faculty of Medicine and
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Bernhard Brüne
- From the Institute of Biochemistry I, Faculty of Medicine and
| |
Collapse
|