1
|
Su W, Yin Y, Zhao J, Hu R, Zhang H, Hu J, Ren R, Zhang Y, Wang A, Lyu Z, Mu Y, Cheng Y. Exosomes derived from umbilical cord-derived mesenchymal stem cells exposed to diabetic microenvironment enhance M2 macrophage polarization and protect against diabetic nephropathy. FASEB J 2024; 38:e23798. [PMID: 38989582 DOI: 10.1096/fj.202400359r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/07/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024]
Abstract
The role of mesenchymal-stem-cell-derived exosomes (MSCs-Exo) in the regulation of macrophage polarization has been recognized in several diseases. There is emerging evidence that MSCs-Exo partially prevent the progression of diabetic nephropathy (DN). This study aimed to investigate whether exosomes secreted by MSCs pre-treated with a diabetic environment (Exo-pre) have a more pronounced protective effect against DN by regulating the balance of macrophages. Exo-pre and Exo-Con were isolated from the culture medium of UC-MSCs pre-treated with a diabetic mimic environment and natural UC-MSCs, respectively. Exo-pre and Exo-Con were injected into the tail veins of db/db mice three times a week for 6 weeks. Serum creatinine and serum urea nitrogen levels, the urinary protein/creatinine ratio, and histological staining were used to determine renal function and morphology. Macrophage phenotypes were analyzed by immunofluorescence, western blotting, and quantitative reverse transcription polymerase chain reaction. In vitro, lipopolysaccharide-induced M1 macrophages were incubated separately with Exo-Con and Exo-pre. We performed microRNA (miRNA) sequencing to identify candidate miRNAs and predict their target genes. An miRNA inhibitor was used to confirm the role of miRNAs in macrophage modulation. Exo-pre were more potent than Exo-Con at alleviating DN. Exo-pre administration significantly reduced the number of M1 macrophages and increased the number of M2 macrophages in the kidney compared to Exo-Con administration. Parallel outcomes were observed in the co-culture experiments. Moreover, miR-486-5p was distinctly expressed in Exo-Con and Exo-pre groups, and it played an important role in macrophage polarization by targeting PIK3R1 through the PI3K/Akt pathway. Reducing miR-486-5p levels in Exo-pre abolished macrophage polarization modulation. Exo-pre administration exhibited a superior effect on DN by remodeling the macrophage balance by shuttling miR-486-5p, which targets PIK3R1.
Collapse
Affiliation(s)
- Wanlu Su
- School of Medicine, Nankai University, Tianjin, China
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yaqi Yin
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jian Zhao
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ruofan Hu
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haixia Zhang
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jia Hu
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Rui Ren
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yue Zhang
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Anning Wang
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhaohui Lyu
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yiming Mu
- School of Medicine, Nankai University, Tianjin, China
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yu Cheng
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
2
|
Bruserud Ø, Mosevoll KA, Bruserud Ø, Reikvam H, Wendelbo Ø. The Regulation of Neutrophil Migration in Patients with Sepsis: The Complexity of the Molecular Mechanisms and Their Modulation in Sepsis and the Heterogeneity of Sepsis Patients. Cells 2023; 12:cells12071003. [PMID: 37048076 PMCID: PMC10093057 DOI: 10.3390/cells12071003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Common causes include gram-negative and gram-positive bacteria as well as fungi. Neutrophils are among the first cells to arrive at an infection site where they function as important effector cells of the innate immune system and as regulators of the host immune response. The regulation of neutrophil migration is therefore important both for the infection-directed host response and for the development of organ dysfunctions in sepsis. Downregulation of CXCR4/CXCL12 stimulates neutrophil migration from the bone marrow. This is followed by transmigration/extravasation across the endothelial cell barrier at the infection site; this process is directed by adhesion molecules and various chemotactic gradients created by chemotactic cytokines, lipid mediators, bacterial peptides, and peptides from damaged cells. These mechanisms of neutrophil migration are modulated by sepsis, leading to reduced neutrophil migration and even reversed migration that contributes to distant organ failure. The sepsis-induced modulation seems to differ between neutrophil subsets. Furthermore, sepsis patients should be regarded as heterogeneous because neutrophil migration will possibly be further modulated by the infecting microorganisms, antimicrobial treatment, patient age/frailty/sex, other diseases (e.g., hematological malignancies and stem cell transplantation), and the metabolic status. The present review describes molecular mechanisms involved in the regulation of neutrophil migration; how these mechanisms are altered during sepsis; and how bacteria/fungi, antimicrobial treatment, and aging/frailty/comorbidity influence the regulation of neutrophil migration.
Collapse
Affiliation(s)
- Øystein Bruserud
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence:
| | - Knut Anders Mosevoll
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Section for Infectious Diseases, Department of Clinical Research, University of Bergen, 5021 Bergen, Norway
| | - Øyvind Bruserud
- Department for Anesthesiology and Intensive Care, Haukeland University Hospital, 5021 Bergen, Norway
| | - Håkon Reikvam
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Øystein Wendelbo
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Faculty of Health, VID Specialized University, Ulriksdal 10, 5009 Bergen, Norway
| |
Collapse
|
3
|
Wang Y, Zhu CL, Li P, Liu Q, Li HR, Yu CM, Deng XM, Wang JF. The role of G protein-coupled receptor in neutrophil dysfunction during sepsis-induced acute respiratory distress syndrome. Front Immunol 2023; 14:1112196. [PMID: 36891309 PMCID: PMC9986442 DOI: 10.3389/fimmu.2023.1112196] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
Sepsis is defined as a life-threatening dysfunction due to a dysregulated host response to infection. It is a common and complex syndrome and is the leading cause of death in intensive care units. The lungs are most vulnerable to the challenge of sepsis, and the incidence of respiratory dysfunction has been reported to be up to 70%, in which neutrophils play a major role. Neutrophils are the first line of defense against infection, and they are regarded as the most responsive cells in sepsis. Normally, neutrophils recognize chemokines including the bacterial product N-formyl-methionyl-leucyl-phenylalanine (fMLP), complement 5a (C5a), and lipid molecules Leukotriene B4 (LTB4) and C-X-C motif chemokine ligand 8 (CXCL8), and enter the site of infection through mobilization, rolling, adhesion, migration, and chemotaxis. However, numerous studies have confirmed that despite the high levels of chemokines in septic patients and mice at the site of infection, the neutrophils cannot migrate to the proper target location, but instead they accumulate in the lungs, releasing histones, DNA, and proteases that mediate tissue damage and induce acute respiratory distress syndrome (ARDS). This is closely related to impaired neutrophil migration in sepsis, but the mechanism involved is still unclear. Many studies have shown that chemokine receptor dysregulation is an important cause of impaired neutrophil migration, and the vast majority of these chemokine receptors belong to the G protein-coupled receptors (GPCRs). In this review, we summarize the signaling pathways by which neutrophil GPCR regulates chemotaxis and the mechanisms by which abnormal GPCR function in sepsis leads to impaired neutrophil chemotaxis, which can further cause ARDS. Several potential targets for intervention are proposed to improve neutrophil chemotaxis, and we hope that this review may provide insights for clinical practitioners.
Collapse
Affiliation(s)
- Yi Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Cheng-long Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Peng Li
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qiang Liu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hui-ru Li
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Faculty of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Chang-meng Yu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiao-ming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Faculty of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Jia-feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
4
|
Zhou YY, Sun BW. Recent advances in neutrophil chemotaxis abnormalities during sepsis. Chin J Traumatol 2022; 25:317-324. [PMID: 35786510 PMCID: PMC9751578 DOI: 10.1016/j.cjtee.2022.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 05/15/2022] [Accepted: 06/03/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis remains one of the leading causes of death globally, in spite of advanced developments in intensive care and better understandings of pathophysiology related to sepsis. There is no special treatment or drug available for sepsis, currently. Under normal circumstances, neutrophil is a major player in acute infection control. However, during sepsis, the migration abilities and antimicrobial functions of neutrophils are impaired, resulting in a dysregulated immune response. Recent studies have indeed demonstrated that blocking or reversing neutrophil migration and impaired antibacterial function can improve the outcomes in septic animal models. This article systemically synthesized information regarding related factors and signaling involved in the functions of neutrophils in sepsis. This review also discussed the possibility that neutrophils be used as a marker for specific diagnosis and/or prediction of the outcomes of sepsis.
Collapse
|
5
|
Xiong W, Jia L, Liang J, Cai Y, Chen Y, Nie Y, Jin J, Zhu J. Investigation into the anti-airway inflammatory role of the PI3Kγ inhibitor JN-PK1: An in vitro and in vivo study. Int Immunopharmacol 2022; 111:109102. [PMID: 35964410 DOI: 10.1016/j.intimp.2022.109102] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/16/2022] [Accepted: 07/25/2022] [Indexed: 12/17/2022]
Abstract
Phosphatidylinositol 3-kinase gamma (PI3Kγ) has been proven to be a potential target for the treatment of inflammatory diseases of the airway; however, there are few reports of selective PI3Kγ inhibitors being used in the field of airway inflammation thus far. Herein, a study employing in vitro and in vivo methodologies was carried out to assess the anti-airway inflammatory effects of JN-PK1, a selective PI3Kγ inhibitor. In RAW264.7 macrophages, JN-PK1 inhibited PI3Kγ-dependent, cellular C5a-induced AKT Ser473 phosphorylation in a concentration- and time-dependent manner and had no significant effect on cell viability.Furthermore, JN-PK1 significantly suppressed LPS-induced, proinflammatory cytokine expression and nitric oxide production through inhibition of the PI3K signaling pathway in RAW264.7 cells. Then, a murine asthma model was established to evaluate the anti-airway inflammation effect of JN-PK1. BALB/c mice were sensitized and challenged with ovalbumin (OVA) to develop an inflammatory response, fibrosis formation, and other airway changes similar to the symptomatology of asthma in humans. Oral administration of JN-PK1 remarkably attenuated OVA-induced asthma in association with the inhibition of the PI3K signaling pathway. That is to say, the oral administration significantly inhibited increases in inflammatory cell counts and reduced T-helper type 2 cytokine production in bronchoalveolar lavage fluid. Pulmonary histological studies showed that oral administration of JN-PK1 not only reduced the infiltration of inflammatory cells but also retarded airway inflammation and fibration. Taken together, JN-PK1 could be developed as a promising candidate for inflammation therapy, and our findings support some potential for therapeutic inhibition of PI3Kγ to treat inflammatory airway diseases.
Collapse
Affiliation(s)
- Wendian Xiong
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Lei Jia
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Junjie Liang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Jingyu Zhu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
6
|
Pereira CMC, Júnior GJD, Lima JVDN, Alves Lemos SI, da Rocha Rodrigues L, Dos Santos Ferreira J, Araújo ASML, de Oliveira JC, Monteiro CE, Franco ÁX, Pereira Alves EH, Oliveira Silva FG, de Carvalho Filgueiras M, Soares PMG, Pereira Vasconcelos DF, de Oliveira JS, de Brito TV, Barbosa ALR. Phosphatidylinositol 3-kinase gamma participates in nimesulide-induced hepatic damage. J Pharm Pharmacol 2021; 73:496-504. [PMID: 33793830 DOI: 10.1093/jpp/rgaa049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To evaluate the participation of the phosphatidylinositol 3-kinase pathway in the liver damage caused by nimesulide. METHODS Liver damage been induced by nimesulide. Mice were treated with either 2% dimethyl sulfoxide or AS605240, a phosphatidylinositol 3-kinase gamma pathway antagonist. Blood samples were collected for function assays of liver. The liver was removed for analysis of liver weight/animal weight ratio, histopathological parameters, oxidative and nitrous stress, cytokine levels, and the immunostaining for cyclooxygenase 2 and nuclear factor kappa B. KEY FINDINGS Liver injured by nimesulide and treated with phosphatidylinositol 3-kinase gamma inhibitor significantly reversed (P < 0.05) the damage; it decreased the liver weight/animal weight ratio, histopathological scores, and neutrophil infiltration, consequently reducing oxidative stress. In addition, we show that phosphatidylinositol 3-kinase gamma is associated with hepatic damage induced by nimesulide, because it altered liver function and increased the protein immunostaining of cyclooxygenase 2 and nuclear factor kappa B in the liver tissue of nimesulide-treated animals. CONCLUSIONS The findings from the present study allows us to infer that nimesulide causes liver damage through the phosphatidylinositol 3-kinase gamma pathway.
Collapse
Affiliation(s)
- Cynthia Maria C Pereira
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Genilson José Dias Júnior
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - José Victor do N Lima
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Sarah Izabelly Alves Lemos
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Lauanda da Rocha Rodrigues
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Jayro Dos Santos Ferreira
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Anna Sofia Miranda Loiola Araújo
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Joveline Costa de Oliveira
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Carlos Eduardo Monteiro
- Laboratory of Physiopharmacology Study of Gastrointestinal Tract (LEFFAG), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Álvaro Xavier Franco
- Laboratory of Physiopharmacology Study of Gastrointestinal Tract (LEFFAG), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Even Herlany Pereira Alves
- Laboratory of Analysis and Histological Processing (LAPHIS), Department of Biomedicine, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Francisca Géssica Oliveira Silva
- Laboratory of Physiopharmacology Study of Gastrointestinal Tract (LEFFAG), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Marcelo de Carvalho Filgueiras
- Laboratory of Muscle Morphology and Physiology (LAMFIM), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Pedro M G Soares
- Laboratory of Physiopharmacology Study of Gastrointestinal Tract (LEFFAG), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Daniel Fernando Pereira Vasconcelos
- Laboratory of Analysis and Histological Processing (LAPHIS), Department of Biomedicine, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Jefferson Soares de Oliveira
- Biochemistry Laboratory of Laticifers Plants (LABPL), Department of Biomedicine, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Tarcisio Vieira de Brito
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - André Luiz Reis Barbosa
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| |
Collapse
|
7
|
Merecz-Sadowska A, Sitarek P, Śliwiński T, Zajdel R. Anti-Inflammatory Activity of Extracts and Pure Compounds Derived from Plants via Modulation of Signaling Pathways, Especially PI3K/AKT in Macrophages. Int J Mol Sci 2020; 21:ijms21249605. [PMID: 33339446 PMCID: PMC7766727 DOI: 10.3390/ijms21249605] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
The plant kingdom is a source of important therapeutic agents. Therefore, in this review, we focus on natural compounds that exhibit efficient anti-inflammatory activity via modulation signaling transduction pathways in macrophage cells. Both extracts and pure chemicals from different species and parts of plants such as leaves, roots, flowers, barks, rhizomes, and seeds rich in secondary metabolites from various groups such as terpenes or polyphenols were included. Selected extracts and phytochemicals control macrophages biology via modulation signaling molecules including NF-κB, MAPKs, AP-1, STAT1, STAT6, IRF-4, IRF-5, PPARγ, KLF4 and especially PI3K/AKT. Macrophages are important immune effector cells that take part in antigen presentation, phagocytosis, and immunomodulation. The M1 and M2 phenotypes are related to the production of pro- and anti-inflammatory agents, respectively. The successful resolution of inflammation mediated by M2, or failed resolution mediated by M1, may lead to tissue repair or chronic inflammation. Chronic inflammation is strictly related to several disorders. Thus, compounds of plant origin targeting inflammatory response may constitute promising therapeutic strategies.
Collapse
Affiliation(s)
- Anna Merecz-Sadowska
- Department of Computer Science in Economics, University of Lodz, 90-214 Lodz, Poland
- Correspondence: (A.M.-S.); (T.Ś.)
| | - Przemysław Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, 90-151 Lodz, Poland;
| | - Tomasz Śliwiński
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
- Correspondence: (A.M.-S.); (T.Ś.)
| | - Radosław Zajdel
- Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland;
| |
Collapse
|
8
|
Nishikoba N, Kumagai K, Kanmura S, Nakamura Y, Ono M, Eguchi H, Kamibayashiyama T, Oda K, Mawatari S, Tanoue S, Hashimoto S, Tsubouchi H, Ido A. HGF-MET Signaling Shifts M1 Macrophages Toward an M2-Like Phenotype Through PI3K-Mediated Induction of Arginase-1 Expression. Front Immunol 2020; 11:2135. [PMID: 32983173 PMCID: PMC7492554 DOI: 10.3389/fimmu.2020.02135] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/06/2020] [Indexed: 01/01/2023] Open
Abstract
Backgrounds and Aims: Hepatocyte Growth Factor (HGF)-MET signaling is known to promote biological functions such as cell survival, cell motility, and cell proliferation. However, it is unknown if HGF-MET alters the macrophage phenotype. In this study, we aimed to study the effects of HGF-MET signaling on the M1 macrophage phenotype. Methods and Materials: Bone marrow-derived macrophages (BMDMs) isolated from mice were either polarized to an M1 phenotype by IFN-γ and LPS treatment or to an M2 phenotype by IL-4 treatment. Changes in M1 or M2 markers induced by HGF-MET signaling were evaluated. Mechanisms responsible for alternations in the macrophage phenotype and intracellular metabolism were analyzed. Results: c-Met was expressed especially in M1 macrophages polarized by treatment with IFN-γ and LPS. In M1 macrophages, HGF-MET signaling induced the expression of Arg-1 mRNA and secretion of IL-10 and TGF-β1 and downregulated the mRNA expression of iNOS, TNF-α, and IL-6. In addition, activation of the PI3K pathway and inactivation of NFκB were also observed in M1 macrophages treated with HGF. The increased Arg-1 expression and IL-10 secretion were abrogated by PI3K inhibition, whereas, no changes were observed in TNF-α and IL-6 expression. The inactivation of NFκB was found to be independent of the PI3K pathway. HGF-MET signaling shifted the M1 macrophages to an M2-like phenotype, mainly through PI3K-mediated induction of Arg-1 expression. Finally, HGF-MET signaling also shifted the M1 macrophage intracellular metabolism toward an M2 phenotype, especially with respect to fatty acid metabolism. Conclusion: Our results suggested that HGF treatment not only promotes regeneration in epithelial cells, but also leads to tissue repair by altering M1 macrophages to an M2-like phenotype.
Collapse
Affiliation(s)
- Nao Nishikoba
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kotaro Kumagai
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shuji Kanmura
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuko Nakamura
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mayumi Ono
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hiromi Eguchi
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tomomi Kamibayashiyama
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kohei Oda
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Seiichi Mawatari
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shiroh Tanoue
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shinichi Hashimoto
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hirohito Tsubouchi
- Department of Gastroenterology and Hepatology, Kagoshima City Hospital, Kagoshima, Japan
| | - Akio Ido
- Digestive and Lifestyle Diseases, Department of Human and Environmental Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
9
|
Yolkin Isolated from Hen Egg Yolk as a Natural Immunoregulator, Activating Innate Immune Response in BMDM Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5731021. [PMID: 32509146 PMCID: PMC7245654 DOI: 10.1155/2020/5731021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023]
Abstract
One of the goals of biomedical sciences is to search and identify natural compounds that are safe, have no side effects, and possess immunostimulatory activity. It has been proven that medicines of natural origin can be effective agents, supporting the therapy of many diseases, not only in the weakened immune system of the body but also in the prevention of many diseases in healthy people. It has been shown that yolkin, a polypeptide complex isolated from hen egg yolk as a fraction accompanying immunoglobulin Y (IgY), possesses potential biological activity. However, the mechanism of its action has not been explained. The objective of this investigation was to examine the molecular mechanisms of innate immune response, activated in response to yolkin, in murine bone marrow-derived macrophages (BMDM). It was shown that yolkin induced phosphorylation of extracellular signal-kinases (ERK1/2) and c-Jun N-terminal kinase (JNK) and upregulated expression and production of type I interferons, TNF-α (tumor necrosis factor α), and nitric oxide (NO), in BMDM cells. Using pharmacological inhibitors of ERK 1/2 and JNK kinases, we revealed that the JNK signaling cascade is required for yolkin-induced inducible NOS expression and upregulation of NO production in mouse macrophages. Using the TLR4-deficient BMDM cell line, we established that yolkin can activate macrophages in a TLR4-dependent manner. It was also shown that NO, TNF-α, and type I IFNs (α/β) produced by BMDM cells in response to yolkin triggered antiviral activity. These data indicate that yolkin affects the regulation of the immune system and antiviral response; therefore, it can be used as an effective immunostimulator of the innate immunity or as a supplement of the conventional therapy of immunodeficiency.
Collapse
|
10
|
The Peptidylarginine Deiminase Inhibitor Cl-Amidine Suppresses Inducible Nitric Oxide Synthase Expression in Dendritic Cells. Int J Mol Sci 2017; 18:ijms18112258. [PMID: 29077055 PMCID: PMC5713228 DOI: 10.3390/ijms18112258] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 02/07/2023] Open
Abstract
The conversion of peptidylarginine into peptidylcitrulline by calcium-dependent peptidylarginine deiminases (PADs) has been implicated in the pathogenesis of a number of diseases, identifying PADs as therapeutic targets for various diseases. The PAD inhibitor Cl-amidine ameliorates the disease course, severity, and clinical manifestation in multiple disease models, and it also modulates dendritic cell (DC) functions such as cytokine production, antigen presentation, and T cell proliferation. The beneficial effects of Cl-amidine make it an attractive compound for PAD-targeting therapeutic strategies in inflammatory diseases. Here, we found that Cl-amidine inhibited nitric oxide (NO) generation in a time- and dose-dependent manner in maturing DCs activated by lipopolysaccharide (LPS). This suppression of NO generation was independent of changes in NO synthase (NOS) enzyme activity levels but was instead dependent on changes in inducible NO synthase (iNOS) transcription and expression levels. Several upstream signaling pathways for iNOS expression, including the mitogen-activated protein kinase, nuclear factor-κB p65 (NF-κB p65), and hypoxia-inducible factor 1 pathways, were not affected by Cl-amidine. By contrast, the LPS-induced signal transducer and the activator of transcription (STAT) phosphorylation and activator protein-1 (AP-1) transcriptional activities (c-Fos, JunD, and phosphorylated c-Jun) were decreased in Cl-amidine-treated DCs. Inhibition of Janus kinase/STAT signaling dramatically suppressed iNOS expression and NO production, whereas AP-1 inhibition had no effect. These results indicate that Cl-amidine-inhibited STAT activation may suppress iNOS expression. Additionally, we found mildly reduced cyclooxygenase-2 expression and prostaglandin E2 production in Cl-amidine-treated DCs. Our findings indicate that Cl-amidine acts as a novel suppressor of iNOS expression, suggesting that Cl-amidine has the potential to ameliorate the effects of excessive iNOS/NO-linked immune responses.
Collapse
|
11
|
Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, Tsatsanis C. Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization. THE JOURNAL OF IMMUNOLOGY 2017; 198:1006-1014. [PMID: 28115590 DOI: 10.4049/jimmunol.1601515] [Citation(s) in RCA: 694] [Impact Index Per Article: 99.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 09/26/2016] [Indexed: 01/05/2023]
Abstract
Macrophages become activated initiating innate immune responses. Depending on the signals, macrophages obtain an array of activation phenotypes, described by the broad terms of M1 or M2 phenotype. The PI3K/Akt/mTOR pathway mediates signals from multiple receptors including insulin receptors, pathogen-associated molecular pattern receptors, cytokine receptors, adipokine receptors, and hormones. As a result, the Akt pathway converges inflammatory and metabolic signals to regulate macrophage responses modulating their activation phenotype. Akt is a family of three serine-threonine kinases, Akt1, Akt2, and Akt3. Generation of mice lacking individual Akt, PI3K, or mTOR isoforms and utilization of RNA interference technology have revealed that Akt signaling pathway components have distinct and isoform-specific roles in macrophage biology and inflammatory disease regulation, by controlling inflammatory cytokines, miRNAs, and functions including phagocytosis, autophagy, and cell metabolism. Herein, we review the current knowledge on the role of the Akt signaling pathway in macrophages, focusing on M1/M2 polarization and highlighting Akt isoform-specific functions.
Collapse
Affiliation(s)
- Eleni Vergadi
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion 71003, Greece; and.,Laboratory of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - Eleftheria Ieronymaki
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion 71003, Greece; and
| | - Konstantina Lyroni
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion 71003, Greece; and
| | - Katerina Vaporidi
- Laboratory of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion 71003, Greece; and
| |
Collapse
|
12
|
Shen XF, Cao K, Jiang JP, Guan WX, Du JF. Neutrophil dysregulation during sepsis: an overview and update. J Cell Mol Med 2017; 21:1687-1697. [PMID: 28244690 PMCID: PMC5571534 DOI: 10.1111/jcmm.13112] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/05/2017] [Indexed: 12/15/2022] Open
Abstract
Sepsis remains a leading cause of death worldwide, despite advances in critical care, and understanding of the pathophysiology and treatment strategies. No specific therapy or drugs are available for sepsis. Neutrophils play a critical role in controlling infection under normal conditions, and it is suggested that their migration and antimicrobial activity are impaired during sepsis which contribute to the dysregulation of immune responses. Recent studies further demonstrated that interruption or reversal of the impaired migration and antimicrobial function of neutrophils improves the outcome of sepsis in animal models. In this review, we provide an overview of the associated mediators and signal pathways involved which govern the survival, migration and antimicrobial function of neutrophils in sepsis, and discuss the potential of neutrophils as a target to specifically diagnose and/or predict the outcome of sepsis.
Collapse
Affiliation(s)
- Xiao-Fei Shen
- Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Ke Cao
- Department of Intensive Care Unit, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jin-Peng Jiang
- Department of Rehabilitation Medicine, PLA Army General Hospital, Beijing, China
| | - Wen-Xian Guan
- Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jun-Feng Du
- Department of General Surgery, PLA Army General Hospital, Beijing, China
| |
Collapse
|
13
|
Sônego F, Castanheira FVES, Ferreira RG, Kanashiro A, Leite CAVG, Nascimento DC, Colón DF, Borges VDF, Alves-Filho JC, Cunha FQ. Paradoxical Roles of the Neutrophil in Sepsis: Protective and Deleterious. Front Immunol 2016; 7:155. [PMID: 27199981 PMCID: PMC4844928 DOI: 10.3389/fimmu.2016.00155] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/11/2016] [Indexed: 12/16/2022] Open
Abstract
Sepsis, an overwhelming inflammatory response syndrome secondary to infection, is one of the costliest and deadliest medical conditions worldwide. Neutrophils are classically considered to be essential players in the host defense against invading pathogens. However, several investigations have shown that impairment of neutrophil migration to the site of infection, also referred to as neutrophil paralysis, occurs during severe sepsis, resulting in an inability of the host to contain and eliminate the infection. On the other hand, the neutrophil antibacterial arsenal contributes to tissue damage and the development of organ dysfunction during sepsis. In this review, we provide an overview of the main events in which neutrophils play a beneficial or deleterious role in the outcome of sepsis.
Collapse
Affiliation(s)
- Fabiane Sônego
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo , Ribeirão Preto , Brazil
| | | | - Raphael Gomes Ferreira
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo , Ribeirão Preto , Brazil
| | - Alexandre Kanashiro
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo , Ribeirão Preto , Brazil
| | | | - Daniele Carvalho Nascimento
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo , Ribeirão Preto , Brazil
| | - David Fernando Colón
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo , Ribeirão Preto , Brazil
| | - Vanessa de Fátima Borges
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo , Ribeirão Preto , Brazil
| | - José Carlos Alves-Filho
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo , Ribeirão Preto , Brazil
| | - Fernando Queiróz Cunha
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo , Ribeirão Preto , Brazil
| |
Collapse
|
14
|
Toll-Like Receptor 4 Promotes NO Synthesis by Upregulating GCHI Expression under Oxidative Stress Conditions in Sheep Monocytes/Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:359315. [PMID: 26576220 PMCID: PMC4630417 DOI: 10.1155/2015/359315] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/26/2015] [Indexed: 12/14/2022]
Abstract
Many groups of Gram-negative bacteria cause diseases that are harmful to sheep. Toll-like receptor 4 (TLR4), which is critical for detecting Gram-negative bacteria by the innate immune system, is activated by lipopolysaccharide (LPS) to initiate inflammatory responses and oxidative stress. Oxidation intermediates are essential activators of oxidative stress, as low levels of free radicals form a stressful oxidative environment that can clear invading pathogens. NO is an oxidation intermediate and its generation is regulated by nitric oxide synthase (iNOS). Guanosine triphosphate cyclohydrolase (GCHI) is the rate-limiting enzyme for tetrahydrobiopterin (BH4) synthesis, which is essential for the production of inducible iNOS. Previously, we made vectors to overexpress the sheep TLR4 gene. Herein, first generation (G1) of transgenic sheep was stimulated with LPS in vivo and in vitro, and oxidative stress and GCHI expression were investigated. Oxidative injury caused by TLR4 overexpression was tightly regulated in tissues. However, the transgenic (Tg) group still secreted nitric oxide (NO) when an iNOS inhibitor was added. Furthermore, GCHI expression remained upregulated in both serum and monocytes/macrophages. Thus, overexpression of TLR4 in transgenic sheep might accelerate the clearance of invading microbes through NO generation following LPS stimulation. Additionally, TLR4 overexpression also enhances GCHI activation.
Collapse
|
15
|
Choi WS, Seo YB, Shin PG, Kim WY, Lee SY, Choi YJ, Kim GD. Veratric acid inhibits iNOS expression through the regulation of PI3K activation and histone acetylation in LPS-stimulated RAW264.7 cells. Int J Mol Med 2014; 35:202-10. [PMID: 25352364 DOI: 10.3892/ijmm.2014.1982] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 10/09/2014] [Indexed: 11/05/2022] Open
Abstract
In the present study, we investigated regulatory effects of veratric acid on the production of nitric oxide (NO) in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. NO production was significantly decreased by veratric acid in the LPS-stimulated RAW264.7 cells in a dose-dependent manner. The reduction in nitric oxide production was induced by the downregulation of inducible NO synthase (iNOS) expression. Veratric acid suppressed the LPS-induced effects on the regulatory and catalytic subunits of phosphoinositide 3-kinase (PI3K), comprised of p85, p110α, p110β and Akt. The acetylation of p300 and the phosphorylation of activating transcription factor 2 (ATF-2) induced by LPS were downregulated following treatment with veratric acid; similar effects were observed following treatment with LY294002, a specific inhibitor of PI3K/Akt. The LPS-induced expression of histone deacetylase (HDAC)3 decreased to basal levels following treatment with veratric acid, and its expression was also downregulated by LY294002. In the measurement of histone acetylation levels, the LPS-stimulated acetylation of histone H4 was significantly attenuated by veratric acid, and was also reduced following the inhibition of PI3K/Akt with LY294002. From our data, it can be concluded that veratric acid exerts a regulatory effect on LPS-induced iNOS expression. Our results suggest that veratric acid impedes the PI3K/Akt-mediated histone acetyl-transferase (HAT) activation and HDAC expression induced by LPS, thereby abrogating iNOS expression.
Collapse
Affiliation(s)
- Woo-Suk Choi
- Department of Microbiology, College of Natural Sciences, Pukyong National University, Busan 608-737, Republic of Korea
| | - Yong-Bae Seo
- Department of Microbiology, College of Natural Sciences, Pukyong National University, Busan 608-737, Republic of Korea
| | - Pyung-Gyun Shin
- Mushroom Research Division, National Institute of Horticultural and Herbal Science, Rural Development Administration, Suwon 441-707, Republic of Korea
| | - Woe-Yeon Kim
- Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Sang Yeol Lee
- Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Young-Ju Choi
- Department of Food and Nutrition, College of Medical Life, Silla University, Busan 617-736, Republic of Korea
| | - Gun-Do Kim
- Department of Microbiology, College of Natural Sciences, Pukyong National University, Busan 608-737, Republic of Korea
| |
Collapse
|
16
|
Verschoor CP, Johnstone J, Loeb M, Bramson JL, Bowdish DME. Anti-pneumococcal deficits of monocyte-derived macrophages from the advanced-age, frail elderly and related impairments in PI3K-AKT signaling. Hum Immunol 2014; 75:1192-6. [PMID: 25446401 DOI: 10.1016/j.humimm.2014.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/07/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
Abstract
The advanced-age, frail elderly are especially vulnerable to developing pneumococcal infection and disease. Macrophages are critical mediators in the defence against Streptococcus pneumoniae at the upper respiratory tract, however, little is known of their anti-pneumococcal capacity in the elderly. Herein we demonstrate that monocyte-derived macrophages (MDMs) from the advanced-age, frail elderly produce less TNF, IL-6, IL-1β and IL-8 in response to heat-killed S. pneumoniae, which does not appear to be related to mRNA stability or decay. Furthermore, despite maintaining the ability to bind and phagocytose bacteria, MDMs from these individuals have a reduced capacity to kill S. pneumoniae. These defects parallel reduced PI3K-AKT signaling, which can significantly abrogate bacterial killing, but does not affect cytokine responses. Since macrophages are critical in the defence against S. pneumoniae, this study adds valuable insight into the susceptibility of the elderly to pneumococcal disease and highlights the PI3K-AKT signaling pathway as a potential therapeutic target.
Collapse
Affiliation(s)
- Chris P Verschoor
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada.
| | - Jennie Johnstone
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada.
| | - Mark Loeb
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada.
| | - Jonathan L Bramson
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada.
| | - Dawn M E Bowdish
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
17
|
Eräsalo H, Laavola M, Hämäläinen M, Leppänen T, Nieminen R, Moilanen E. PI3K Inhibitors LY294002 and IC87114 Reduce Inflammation in Carrageenan-Induced Paw Oedema and Down-Regulate Inflammatory Gene Expression in Activated Macrophages. Basic Clin Pharmacol Toxicol 2014; 116:53-61. [DOI: 10.1111/bcpt.12284] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/28/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Heikki Eräsalo
- The Immunopharmacology Research Group; University of Tampere School of Medicine and Tampere University Hospital; Tampere Finland
| | - Mirka Laavola
- The Immunopharmacology Research Group; University of Tampere School of Medicine and Tampere University Hospital; Tampere Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group; University of Tampere School of Medicine and Tampere University Hospital; Tampere Finland
| | - Tiina Leppänen
- The Immunopharmacology Research Group; University of Tampere School of Medicine and Tampere University Hospital; Tampere Finland
| | - Riina Nieminen
- The Immunopharmacology Research Group; University of Tampere School of Medicine and Tampere University Hospital; Tampere Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group; University of Tampere School of Medicine and Tampere University Hospital; Tampere Finland
| |
Collapse
|
18
|
|
19
|
Gillette DD, Tridandapani S, Butchar JP. Monocyte/macrophage inflammatory response pathways to combat Francisella infection: possible therapeutic targets? Front Cell Infect Microbiol 2014; 4:18. [PMID: 24600590 PMCID: PMC3930869 DOI: 10.3389/fcimb.2014.00018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 02/02/2014] [Indexed: 01/05/2023] Open
Abstract
Francisella tularensis can bypass and suppress host immune responses, even to the point of manipulating immune cell phenotypes and intercellular inflammatory networks. Strengthening these responses such that immune cells more readily identify and destroy the bacteria is likely to become a viable (and perhaps necessary) strategy for combating infections with Francisella, especially given the likelihood of antibiotic resistance in the foreseeable future. Monocytes and macrophages offer a niche wherein Francisella can invade and replicate, resulting in substantially higher bacterial load that can overcome the host. As such, understanding their responses to Francisella may uncover potential avenues of therapy that could promote a lowering of bacterial burden and clearance of infection. These response pathways include Toll-like Receptor 2 (TLR2), the caspase-1 inflammasome, Interferons, NADPH oxidase, Phosphatidylinositide 3-kinase (PI3K), and the Ras pathway. In this review we summarize the literature pertaining to the roles of these pathways during Francisella infection, with an emphasis on monocyte/macrophage responses. The therapeutic targeting of one or more such pathways may ultimately become a valuable tool for the treatment of tularemia, and several possibilities are discussed.
Collapse
Affiliation(s)
- Devyn D Gillette
- Department of Internal Medicine, Wexner Medical Center, The Ohio State University Columbus, OH, USA
| | - Susheela Tridandapani
- Department of Internal Medicine, Wexner Medical Center, The Ohio State University Columbus, OH, USA
| | - Jonathan P Butchar
- Department of Internal Medicine, Wexner Medical Center, The Ohio State University Columbus, OH, USA
| |
Collapse
|
20
|
Shanmuganathan MV, Krishnan S, Fu X, Prasadarao NV. Escherichia coli K1 induces pterin production for enhanced expression of Fcγ receptor I to invade RAW 264.7 macrophages. Microbes Infect 2014; 16:134-41. [PMID: 24161960 PMCID: PMC3946618 DOI: 10.1016/j.micinf.2013.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 10/04/2013] [Accepted: 10/15/2013] [Indexed: 11/16/2022]
Abstract
Macrophages serve as permissive niches for Escherichia coli (E. coli) K1 to attain high grade bacteremia in the pathogenesis of meningitis in neonates. Although pterin levels are a diagnostic marker for immune activation, the role of macrophages in pterin production and in the establishment of meningitis is unknown. Here, we demonstrate that macrophages infected with E. coli K1 produce both neopterin and biopterin through increased expression of GTP-cyclohydrolase 1 (GCH1). Of note, increased production of biopterin enhances the expression of Fc-gamma receptor I (CD64), which in turn, aided the entry of E. coli K1 in macrophages while increased neopterin suppresses reactive oxygen species (ROS), thereby aiding bacterial survival. Inhibition of GCH1 by 2, 4-Diamino-6-hydroxypyrimidine (DAHP) prevented the E. coli K1 induced expression of CD64 in macrophages in vitro and the development of bacteremia in a newborn mouse model of meningitis. These studies suggest that targeting GCH1 could be therapeutic strategy for preventing neonatal meningitis by E. coli K1.
Collapse
Affiliation(s)
- Muthusamy V Shanmuganathan
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd., Los Angeles, CA 90027, USA
| | - Subramanian Krishnan
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd., Los Angeles, CA 90027, USA
| | - Xiaowei Fu
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd., Los Angeles, CA 90027, USA
| | - Nemani V Prasadarao
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd., Los Angeles, CA 90027, USA; Department of Surgery, Children's Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd., Los Angeles, CA 90027, USA.
| |
Collapse
|
21
|
Tsai CY, Chang AYW, Chan JYH, Chan SHH. Activation of PI3K/Akt signaling in rostral ventrolateral medulla impairs brain stem cardiovascular regulation that underpins circulatory depression during mevinphos intoxication. Biochem Pharmacol 2014; 88:75-85. [PMID: 24462917 DOI: 10.1016/j.bcp.2014.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/12/2014] [Accepted: 01/13/2014] [Indexed: 11/27/2022]
Abstract
As the most widely used pesticides in the globe, the organophosphate compounds are understandably linked with the highest incidence of suicidal poisoning. Whereas the elicited toxicity is often associated with circulatory depression, the underlying mechanisms require further delineation. Employing the pesticide mevinphos as our experimental tool, we evaluated the hypothesis that transcriptional upregulation of nitric oxide synthase II (NOS II) by NF-κB on activation of the PI3K/Akt cascade in the rostral ventrolateral medulla (RVLM), the brain stem site that maintains blood pressure and sympathetic vasomotor tone, underpins the circulatory depressive effects of organophosphate poisons. Microinjection of mevinphos (10 nmol) bilaterally into the RVLM of anesthetized Sprague-Dawley rats induced a progressive hypotension that was accompanied sequentially by an increase (Phase I) and a decrease (Phase II) of an experimental index for the baroreflex-mediated sympathetic vasomotor tone. There were also progressive augmentations in PI3K or Akt enzyme activity and phosphorylation of p85 or Akt(Thr308) subunit in the RVLM that were causally related to an increase in NF-κB transcription activity and elevation in NOS II or peroxynitrite expression. Loss-of-function manipulations of PI3K or Akt in the RVLM significantly antagonized the reduced baroreflex-mediated sympathetic vasomotor tone and hypotension during Phase II mevinphos intoxication, and blunted the increase in NF-κB/NOS II/peroxynitrite signaling. We conclude that activation of the PI3K/Akt cascade, leading to upregulation of NF-κB/NOS II/peroxynitrite signaling in the RVLM, elicits impairment of brain stem cardiovascular regulation that underpins circulatory depression during mevinphos intoxication.
Collapse
Affiliation(s)
- Ching-Yi Tsai
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan, ROC
| | - Alice Y W Chang
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan, ROC
| | - Julie Y H Chan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan, ROC
| | - Samuel H H Chan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan, ROC.
| |
Collapse
|
22
|
Ratajczak-Wrona W, Jablonska E, Garley M, Jablonski J, Radziwon P, Iwaniuk A, Grubczak K. PI3K-Akt/PKB signaling pathway in neutrophils and mononuclear cells exposed to N-nitrosodimethylamine. J Immunotoxicol 2013; 11:231-7. [PMID: 23971717 DOI: 10.3109/1547691x.2013.826307] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Neutrophils (PMN) play diverse regulatory and effector functions in the immune system through the release of reactive nitrogen species, including nitric oxide (NO). The enzyme responsible for NO synthesis in PMN is inducible nitric oxide synthase (iNOS) that is regulated by various signaling pathways, e.g. PI3K-Akt/PKB, and transcription factors. N-Nitrosodimethylamine (NDMA), a xenobiotic widespread in the human environment, affects immune cells. The study objective here was to examine the role of the PI3K-Akt/PKB pathway in induction of NO synthesis (with involvement of iNOS) in human PMN, as well as in autologous mononuclear cells (PBMC), exposed to NDMA. Isolated cells were incubated for 2 h with a sub-lethal dose of NDMA and then the expression of several select proteins in the cell cytoplasmic and nuclear fractions were determined by Western blot analyses. The results indicated that NDMA enhanced expression of iNOS, phospho-PI3K, and phospho-IκBα in the cytoplasmic fraction of the PMN and PBMC. The nuclear fraction of these cells also had a higher NF-κB expression. Moreover, in PMN, NDMA caused an increased expression of phospho-Akt (T308), phospho-Akt (S473), and phospho-IKKαβ in the cytoplasm, and c-Jun and FosB in the nuclear fraction. Blocking of PI3K caused a decrease in expression of all these proteins in NDMA-exposed PMN. However, inhibition of PI3K led to a drop in expression of iNOS, phospho-PI3K, and phospho-IκBα in the cytoplasm, and in NF-κB in the nuclear fraction, of PBMC. The results of these studies indicated to us that NDMA activates the PI3K-Akt/PKB pathway in human PMN and that this, in turn, contributes to the activation of transcription factors NF-κB, c-Jun, and FosB involved in NO production (through modulation of iNOS expression).
Collapse
|
23
|
Inhibitory effect of citrinin on lipopolisaccharide-induced nitric oxide production by mouse macrophage cells. Mycotoxin Res 2013; 29:229-34. [DOI: 10.1007/s12550-013-0175-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 07/09/2013] [Accepted: 07/12/2013] [Indexed: 01/07/2023]
|
24
|
Shanmuganathan MV, Krishnan S, Fu X, Prasadarao NV. Attenuation of biopterin synthesis prevents Escherichia coli K1 invasion of brain endothelial cells and the development of meningitis in newborn mice. J Infect Dis 2013; 207:61-71. [PMID: 23100563 PMCID: PMC3523800 DOI: 10.1093/infdis/jis656] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 08/10/2012] [Indexed: 11/12/2022] Open
Abstract
Elevated levels of pterins and nitric oxide (NO) are observed in patients with septic shock and bacterial meningitis. We demonstrate that Escherichia coli K1 infection of human brain microvascular endothelial cells (HBMECs) induces the expression of guanosine triphosphate cyclohydrolase (GCH1), the rate-limiting enzyme in pterin synthesis, thereby elevating levels of biopterin. DAHP (2,4-diamino hydroxyl pyrimidine), a specific inhibitor of GCH1, prevented biopterin and NO production and invasion of E. coli K1 in HBMECs. GCH1 interaction with Ecgp96, the receptor for outer membrane protein A of E. coli K1, also increases on infection, and suppression of Ecgp96 expression prevents GCH1 activation and biopterin synthesis. Pretreatment of newborn mice with DAHP prevented the production of biopterin and the development of meningitis. These results suggest a novel role for biopterin synthesis in the pathogenesis of E. coli K1 meningitis.
Collapse
Affiliation(s)
| | | | - Xiaowei Fu
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, California
- Keck School of Medicine, University of Southern California, Los Angeles
| | - Nemani V. Prasadarao
- Division of Infectious Diseases, Department of Pediatrics
- Department of Surgery
- Keck School of Medicine, University of Southern California, Los Angeles
| |
Collapse
|
25
|
Song S, Zhou F, Chen WR, Xing D. PDT-induced HSP70 externalization up-regulates NO production via TLR2 signal pathway in macrophages. FEBS Lett 2012; 587:128-35. [PMID: 23247210 DOI: 10.1016/j.febslet.2012.11.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 10/31/2012] [Accepted: 11/16/2012] [Indexed: 02/07/2023]
Abstract
We studied the molecular mechanism underlying PDT-induced apoptosis-dependent macrophage activation, particularly through NO production. We demonstrate that NO production is initially induced by HSP70 on the apoptotic cell surface, and is further enhanced by macrophage phagocytosis. Additionally, we found that apoptotic cells, through TLR2, could activate PI3K, and this could be either dependent or independent of the activation of MyD88. These results reveal a novel pathway linking innate immune signalling to apoptotic cells and point at HSP70 as an important antitumor immunostimulant. They also indicate that PDT-induced apoptosis has an important role in macrophage innate immunity.
Collapse
Affiliation(s)
- Sheng Song
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | | | | | | |
Collapse
|
26
|
Abstract
TLR agonists initiate a rapid activation program in dendritic cells (DCs) that requires support from metabolic and bioenergetic resources. We found previously that TLR signaling promotes aerobic glycolysis and a decline in oxidative phosphorylation (OXHPOS) and that glucose restriction prevents activation and leads to premature cell death. However, it remained unclear why the decrease in OXPHOS occurs under these circumstances. Using real-time metabolic flux analysis, in the present study, we show that mitochondrial activity is lost progressively after activation by TLR agonists in inflammatory blood monocyte-derived DCs that express inducible NO synthase. We found that this is because of inhibition of OXPHOS by NO and that the switch to glycolysis is a survival response that serves to maintain ATP levels when OXPHOS is inhibited. Our data identify NO as a profound metabolic regulator in inflammatory monocyte-derived DCs.
Collapse
|
27
|
Activation of liver X receptors attenuates endotoxin-induced liver injury in mice with nonalcoholic fatty liver disease. Dig Dis Sci 2012; 57:390-8. [PMID: 21948338 DOI: 10.1007/s10620-011-1902-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 08/31/2011] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIMS Nonalcoholic fatty liver disease (NAFLD) is classically associated with insulin resistance and the inflammatory response, especially in the nonalcoholic steatohepatitis phase. The liver X receptors (LXRs) play a critical role in the regulation of cholesterol metabolism and inflammatory processes. METHODS Wild-type C57BL/6 mice were fed a normal diet (ND) or a high-fat (HF) diet for 8 weeks. Some ND- and HF-fed mice were treated (i.p.) with the LXR agonist T0901317 (30 mg/kg/day) for 7 days. Lipopolysaccharide (LPS, 50 μg/mouse) was then injected intraperitoneally to induce liver injury. The activation of MAPKs, NF-κB and the PI3K pathway was evaluated using Western blot. Bone marrow-derived macrophages (MDMs) were isolated from the femurs of C57BL/6 mice and cultured with or without T0901317 (20 μmol/l). The expression of tumor necrosis factor-alpha (TNF-α) and inducible nitric oxide synthase (iNOS) was evaluated in vitro or in vivo using real-time PCR, immunohistochemistry, or Western blot. RESULTS The LXR agonist T0901317 attenuated LPS-induced liver injury in a murine model of NAFLD, reflected by reduced serum alanine aminotransferase and aspartate aminotransferase levels, and reduced liver histology changes. Activation of LXRs reduced TNF-α and iNOS expression through inhibiting JNK and the PI3K signaling pathway. An in vitro study demonstrated that the activation of LXR inhibited the expression of TNF-α and iNOS in the MDMs of mice. CONCLUSIONS Activation of LXRs attenuates LPS-induced liver injury in murine NAFLD through inhibiting the pro-inflammatory activity of macrophages.
Collapse
|
28
|
Tsai KD, Chang WW, Lin CC, Hsu SC, Lee YJ, Chen W, Shieh JC, Lin TH. Differential effects of LY294002 and wortmannin on inducible nitric oxide synthase expression in glomerular mesangial cells. Int Immunopharmacol 2012; 12:471-80. [PMID: 22240122 DOI: 10.1016/j.intimp.2011.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 11/26/2011] [Accepted: 12/22/2011] [Indexed: 01/07/2023]
Abstract
Nitric oxide (NO) that is produced by inducible nitric oxide synthase (iNOS) is associated with the pathophysiology of glomerulonephritis. Numerous studies have focused on the regulation of NO production by iNOS to reduce NO-mediated cytotoxicity. In the present study, we demonstrated the differential effects of two phosphatidylinositol 3-kinase (PI3K) inhibitors, LY294002 and wortmannin, on lipopolysaccharide- (LPS) and interferon (IFN)-γ-induced NO production in a glomerular mesangial cell line, MES-13 cells. At dosages without affecting cell viability of MES-13 cells, 5μM LY294002 showed a more-significant inhibitory effect on LPS/IFN-γ-induced NO production, and iNOS protein and gene expressions than did 1μM wortmannin. Akt phosphorylation in MES-13 cells declined upon the addition of wortmannin, but not upon treatment with LY294002. Suppression of PI3K expression by small interfering (si)RNA exhibited no effect on LPS/IFN-γ-stimulated NO production or iNOS protein expression in MES-13 cells. Neither LY294002 nor wortmannin reduced IFN-γ-induced STAT-1α phosphorylation. LY294002 exhibited a more-significant inhibitory effect on NF-κB luciferase activities than wortmannin in LPS/IFN-γ-stimulated MES-13 cells. Moreover, LY294002, but not wortmannin, accelerated iNOS protein degradation and reduced the iNOS dimer/monomer ratio in MES-13 cells. Although both LY294002 and wortmannin are known as PI3K inhibitors, their differential effects on iNOS expression in MES-13 cells indicate that the effects of LY294002 on inhibiting NF-κB activation and accelerating iNOS protein degradation are through a mechanism independent of PI3K.
Collapse
Affiliation(s)
- Kuen-Daw Tsai
- Department of Internal Medicine, China Medical University Beigang Hospital, 123 Sinde Road, Beigang Township, Yunlin County 65152, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Reis J, Guan XQ, Kisselev AF, Papasian CJ, Qureshi AA, Morrison DC, Van Way CW, Vogel SN, Qureshi N. LPS-induced formation of immunoproteasomes: TNF-α and nitric oxide production are regulated by altered composition of proteasome-active sites. Cell Biochem Biophys 2011; 60:77-88. [PMID: 21455682 DOI: 10.1007/s12013-011-9182-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Stimulation of mouse macrophages with LPS leads to tumor necrosis factor (TNF-α) secretion and nitric oxide (NO) release at different times through independent signaling pathways. While the precise regulatory mechanisms responsible for these distinct phenotypic responses have not been fully delineated, results of our recent studies strongly implicate the cellular cytoplasmic ubiquitin-proteasome pathway as a key regulator of LPS-induced macrophage inflammatory responses. Our objective in this study was to define the relative contribution of specific proteasomal active-sites in induction of TNF-α and NO after LPS treatment of RAW 264.7 macrophages using selective inhibitors of these active sites. Our data provide evidence that LPS stimulation of mouse macrophages triggers a selective increase in the levels of gene and protein expression of the immunoproteasomes, resulting in a modulation of specific functional activities of the proteasome and a corresponding increase in NO production as compared to untreated controls. These findings suggest the LPS-dependent induction of immunoproteasome. In contrast, we also demonstrate that TNF-α expression is primarily dependent on both the chymotrypsin- and the trypsin-like activities of X, Y, Z subunits of the proteasome. Proteasome-associated post-acidic activity alone also contributes to LPS-induced expression of TNF-α. Taken together; our results indicate that LPS-induced TNF-α in macrophages is differentially regulated by each of the three proteasome activities. Since addition of proteasome inhibitors to mouse macrophages profoundly affects the degradation of proteins involved in signal transduction, we conclude that proteasome-specific degradation of several signaling proteins is likely involved in differential regulation of LPS-dependent secretion of proinflammatory mediators.
Collapse
Affiliation(s)
- Julia Reis
- Department of Basic Medical Science, Shock/Trauma Research Center, School of Medicine, University of Missouri, 2411 Holmes Street, Kansas City, MO 64108, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Phosphatidylinositol 3-kinase inhibitor suppresses inducible nitric oxide synthase expression in bronchiole epithelial cells in asthmatic rats. Mol Cell Biochem 2011; 359:293-9. [PMID: 21847581 DOI: 10.1007/s11010-011-1023-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 07/29/2011] [Indexed: 02/06/2023]
Abstract
Inducible nitric oxide synthase (iNOS) is known to produce nitric oxide (NO), which is a main contributor to asthmatic airway inflammation. Recent studies have shown that phosphatidylinositol 3-kinase (PI3K) is ubiquitously expressed in airway epithelial cells and its inhibition could relieve airway inflammation and hyperresponsiveness. This study aimed to explore the interaction of PI3K and NO signaling in allergic asthma. We investigated the effects of PI3K inhibitor wortmannin on iNOS expression in bronchiole epithelial cells and NO, IL-4 and IFN-γ levels in lung tissues of asthmatic rat model, which was prepared by 10% OVA solution sensitization and 1% OVA aerosol challenge. Our results showed that the ratio of eosinophils to total cells in BALF, PI3K activity, NO and IL-4 levels in lung tissues was increased after OVA sensitization and challenge, but then was attenuated by the administration of wortmannin. In contrast, IFN-γ level in lung tissues was decreased after OVA sensitization and challenge and increased after the administration of wortmannin. The expression of iNOS protein in bronchiole epithelial cells, iNOS mRNA level and iNOS activity in lung tissues was markedly upregulated after OVA sensitization and challenge, but the upregulation was significantly antagonized by wortmannin. Taken together, these data provide evidence that PI3K functions upstream to modulate iNOS/NO signaling, which then promotes the development of airway inflammation in asthmatic animal model. PI3K inhibitor wortmannin could lead to reduced iNOS expression and NO production, therefore inhibiting airway inflammatory responses.
Collapse
|
31
|
He T, Smith LA, Lu T, Joyner MJ, Katusic ZS. Activation of peroxisome proliferator-activated receptor-{delta} enhances regenerative capacity of human endothelial progenitor cells by stimulating biosynthesis of tetrahydrobiopterin. Hypertension 2011; 58:287-94. [PMID: 21709207 PMCID: PMC3143017 DOI: 10.1161/hypertensionaha.111.172189] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The mechanisms underlying the regenerative capacity of endothelial progenitor cells (EPCs) are not fully understood. We hypothesized that biosynthesis of tetrahydrobiopterin is an important mechanism responsible for the stimulatory effects of peroxisome proliferator-activated receptor-δ (PPARδ) activation on regenerative function of human EPCs. Treatment of human EPCs with a selective PPARδ agonist GW501516 for 24 hours increased the levels of mRNA, protein, and enzymatic activity of GTP cyclohydrolase I (GTPCH I), as well as the production of tetrahydrobiopterin. The effects of GW501516 were mediated by suppression of PTEN expression, thereby increasing phosphorylation of AKT. The AKT signaling also mediated GW501516-induced phosphorylation of endothelial NO synthase. In addition, activation of PPARδ significantly enhanced proliferation of EPCs. This effect was abolished by the GTPCH I inhibitor, 2,4-diamino-6-hydroxypyrimidine, or genetic inactivation of GTPCH I with small interfering RNA but not by inhibition of endothelial NO synthase with N(G)-nitro-l-arginine methyl ester. Supplementation of NO did not reverse 2,4-diamino-6-hydroxypyrimidine-inhibited 5-bromodeoxyuridine incorporation. Furthermore, transplantation of human EPCs stimulated re-endothelialization in a mouse model of carotid artery injury. Pretreatment of EPCs with GW501516 significantly enhanced the ability of transplanted EPCs to repair denuded endothelium. GTPCH I-small interfering RNA transfection significantly inhibited in vivo regenerative capacity of EPCs stimulated with GW501516. Thus, in human EPCs, activation of PPARδ stimulates expression and activity of GTPCH I and biosynthesis of tetrahydrobiopterin via PTEN-AKT signaling pathway. This effect enhances the regenerative function of EPCs.
Collapse
Affiliation(s)
- Tongrong He
- Department of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Rochester, Minnesota
| | - Leslie A. Smith
- Department of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Rochester, Minnesota
| | - Tong Lu
- Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Michael J. Joyner
- Department of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Rochester, Minnesota
| | - Zvonimir S. Katusic
- Department of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Rochester, Minnesota
| |
Collapse
|
32
|
Cheng C, Ho WE, Goh FY, Guan SP, Kong LR, Lai WQ, Leung BP, Wong WSF. Anti-malarial drug artesunate attenuates experimental allergic asthma via inhibition of the phosphoinositide 3-kinase/Akt pathway. PLoS One 2011; 6:e20932. [PMID: 21695271 PMCID: PMC3111464 DOI: 10.1371/journal.pone.0020932] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 05/13/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Phosphoinositide 3-kinase (PI3K)/Akt pathway is linked to the development of asthma. Anti-malarial drug artesunate is a semi-synthetic derivative of artemisinin, the principal active component of a medicinal plant Artemisia annua, and has been shown to inhibit PI3K/Akt activity. We hypothesized that artesunate may attenuate allergic asthma via inhibition of the PI3K/Akt signaling pathway. METHODOLOGY/PRINCIPAL FINDINGS Female BALB/c mice sensitized and challenged with ovalbumin (OVA) developed airway inflammation. Bronchoalveolar lavage fluid was assessed for total and differential cell counts, and cytokine and chemokine levels. Lung tissues were examined for cell infiltration and mucus hypersecretion, and the expression of inflammatory biomarkers. Airway hyperresponsiveness was monitored by direct airway resistance analysis. Artesunate dose-dependently inhibited OVA-induced increases in total and eosinophil counts, IL-4, IL-5, IL-13 and eotaxin levels in bronchoalveolar lavage fluid. It attenuated OVA-induced lung tissue eosinophilia and airway mucus production, mRNA expression of E-selectin, IL-17, IL-33 and Muc5ac in lung tissues, and airway hyperresponsiveness to methacholine. In normal human bronchial epithelial cells, artesunate blocked epidermal growth factor-induced phosphorylation of Akt and its downstream substrates tuberin, p70S6 kinase and 4E-binding protein 1, and transactivation of NF-κB. Similarly, artesunate blocked the phosphorylation of Akt and its downstream substrates in lung tissues from OVA-challenged mice. Anti-inflammatory effect of artesunate was further confirmed in a house dust mite mouse asthma model. CONCLUSION/SIGNIFICANCE Artesunate ameliorates experimental allergic airway inflammation probably via negative regulation of PI3K/Akt pathway and the downstream NF-κB activity. These findings provide a novel therapeutic value for artesunate in the treatment of allergic asthma.
Collapse
Affiliation(s)
- Chang Cheng
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
- Immunology Program, Life Science Institute; National University of Singapore, Singapore
| | - W. Eugene Ho
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
- Immunology Program, Life Science Institute; National University of Singapore, Singapore
| | - Fera Y. Goh
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
- Immunology Program, Life Science Institute; National University of Singapore, Singapore
| | - Shou Ping Guan
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
- Immunology Program, Life Science Institute; National University of Singapore, Singapore
| | - Li Ren Kong
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Wen-Qi Lai
- Departments of Physiology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Bernard P. Leung
- Departments of Physiology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - W. S. Fred Wong
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
- Immunology Program, Life Science Institute; National University of Singapore, Singapore
| |
Collapse
|
33
|
Herbst S, Schaible UE, Schneider BE. Interferon gamma activated macrophages kill mycobacteria by nitric oxide induced apoptosis. PLoS One 2011; 6:e19105. [PMID: 21559306 PMCID: PMC3085516 DOI: 10.1371/journal.pone.0019105] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 03/17/2011] [Indexed: 02/03/2023] Open
Abstract
Mycobacterium tuberculosis is an intracellular pathogen of macrophages and escapes the macrophages' bactericidal effectors by interfering with phagosome-lysosome fusion. IFN-γ activation renders the macrophages capable of killing intracellular mycobacteria by overcoming the phagosome maturation block, nutrient deprivation and exposure to microbicidal effectors including nitric oxide (NO). While the importance about NO for the control of mycobacterial infection in murine macrophages is well documented, the underlying mechanism has not been revealed yet. In this study we show that IFN-γ induced apoptosis in mycobacteria-infected macrophages, which was strictly dependent on NO. Subsequently, NO-mediated apoptosis resulted in the killing of intracellular mycobacteria independent of autophagy. In fact, killing of mycobacteria was susceptible to the autophagy inhibitor 3-methyladenine (3-MA). However, 3-MA also suppressed NO production, which is an important off-target effect to be considered in autophagy studies using 3-MA. Inhibition of caspase 3/7 activation, as well as NO production, abolished apoptosis and elimination of mycobacteria by IFN-γ activated macrophages. In line with the finding that drug-induced apoptosis kills intracellular mycobacteria in the absence of NO, we identified NO-mediated apoptosis as a new defense mechanism of activated macrophages against M. tuberculosis.
Collapse
Affiliation(s)
- Susanne Herbst
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ulrich E. Schaible
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Department of Molecular Infection Biology, Research Center Borstel, Borstel, Germany
- * E-mail:
| | - Bianca E. Schneider
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Department of Molecular Infection Biology, Research Center Borstel, Borstel, Germany
| |
Collapse
|
34
|
Sugiyama KI, Kawakami H, Kamata Y, Sugita-Konishi Y. Effect of a combination of deoxynivalenol and nivalenol on lipopolisaccharide-induced nitric oxide production by mouse macrophages. Mycotoxin Res 2010; 27:57-62. [DOI: 10.1007/s12550-010-0076-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/29/2010] [Accepted: 10/30/2010] [Indexed: 10/18/2022]
|
35
|
Chen L, Zeng X, Wang J, Briggs SS, O'Neill E, Li J, Leek R, Kerr DJ, Harris AL, Cai S. Roles of tetrahydrobiopterin in promoting tumor angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2671-80. [PMID: 20847284 PMCID: PMC2966821 DOI: 10.2353/ajpath.2010.100025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/21/2010] [Indexed: 01/09/2023]
Abstract
Nitric oxide (NO), which is derived from endothelial NO synthase (eNOS), provides crucial signals for angiogenesis in the tumor microenvironment. Tetrahydrobiopterin (BH4) is an absolute requirement for eNOS activity. In this study, we investigated whether this activation is both maintained by a wild-type Ras/phosphatidylinositol 3-kinase (PI3K)/Akt-positive feedback loop in endothelial cells and affects tumor angiogenesis. We found that supplementation of BH4 (via the pterin salvage pathway with Sep) increased Akt/eNOS phosphorylation in both human eNOS-transfected COS-7 cells and endothelial cells concomitant with increases in NO production, cell proliferation, migration, and tube formation. This augmentation was abrogated by a PI3K inhibitor. Sepiapterin (Sep) also increased GTP-bound wild-type Ras and PI3K/Akt/eNOS activation, which was prevented by the eNOS inhibitor, Nω-Nitro-L-arginine methyl ester (L-NAME). Furthermore, expression of GTP cyclohydrolase I (the rate-limiting enzyme in de novo BH4 synthesis) under doxycycline control potentiated in vivo tumorigenesis, tumor cell proliferation, as well as angiogenesis. Conversely, both switching off GTP cyclohydrolase I expression as well as inhibiting its enzymatic activity significantly decreased eNOS expression and tumor vascularization. This study demonstrates an important role for BH4 synthesis in angiogenesis by the activation of eNOS for NO production, which is maintained by a PI3K/Akt-positive feedback loop through effects on wild-type Ras in endothelial cells. Our findings suggest that BH4 synthesis may be a rational target for antiangiogenesis therapy for tumors.
Collapse
Affiliation(s)
- Liye Chen
- Department of Clinical Pharmacology, University of Oxford, John Radcliffe Hospital, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Sun J, Druhan LJ, Zweier JL. Reactive oxygen and nitrogen species regulate inducible nitric oxide synthase function shifting the balance of nitric oxide and superoxide production. Arch Biochem Biophys 2010; 494:130-7. [PMID: 19932078 PMCID: PMC4073618 DOI: 10.1016/j.abb.2009.11.019] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 11/17/2009] [Indexed: 01/30/2023]
Abstract
Inducible NOS (iNOS) is induced in diseases associated with inflammation and oxidative stress, and questions remain regarding its regulation. We demonstrate that reactive oxygen/nitrogen species (ROS/RNS) dose-dependently regulate iNOS function. Tetrahydrobiopterin (BH4)-replete iNOS was exposed to increasing concentrations of ROS/RNS and activity was measured with and without subsequent BH4 addition. Peroxynitrite (ONOO(-)) produced the greatest change in NO generation rate, approximately 95% decrease, and BH4 only partially restored this loss of activity. Superoxide (O2(.-)) greatly decreased NO generation, however, BH4 addition restored this activity. Hydroxyl radical ((.)OH) mildly decreases NO generation in a BH4-dependent manner. iNOS was resistant to H2O2 with only slightly decreased NO generation with up to millimolar concentrations. In contrast to the inhibition of NO generation, ROS enhanced O2(.-) production from iNOS, while ONOO(-) had the opposite effect. Thus, ROS promote reversible iNOS uncoupling, while ONOO(-) induces irreversible enzyme inactivation and decreases both NO and O2(.-) production.
Collapse
Affiliation(s)
- Jian Sun
- Davis Heart and Lung Research Institute, and the Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, College of Medicine, Columbus, OH 43210, USA
| | - Lawrence J. Druhan
- Davis Heart and Lung Research Institute, and the Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, College of Medicine, Columbus, OH 43210, USA
| | - Jay L. Zweier
- Davis Heart and Lung Research Institute, and the Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
37
|
Sugiyama KI, Muroi M, Tanamoto KI, Nishijima M, Sugita-Konishi Y. Deoxynivalenol and nivalenol inhibit lipopolysaccharide-induced nitric oxide production by mouse macrophage cells. Toxicol Lett 2010; 192:150-4. [DOI: 10.1016/j.toxlet.2009.10.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 10/15/2009] [Accepted: 10/15/2009] [Indexed: 12/26/2022]
|
38
|
Du J, Wei N, Guan T, Xu H, An J, Pritchard KA, Shi Y. Inhibition of CDKS by roscovitine suppressed LPS-induced *NO production through inhibiting NFkappaB activation and BH4 biosynthesis in macrophages. Am J Physiol Cell Physiol 2009; 297:C742-9. [PMID: 19553566 PMCID: PMC2740392 DOI: 10.1152/ajpcell.00138.2009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 06/23/2009] [Indexed: 11/22/2022]
Abstract
In inflammatory diseases, tissue damage is critically associated with nitric oxide ((*)NO) and cytokines, which are overproduced in response to cellular release of endotoxins. Here we investigated the inhibitory effect of roscovitine, a selective inhibitor of cyclin-dependent kinases (CDKs) on (*)NO production in mouse macrophages. In RAW264.7 cells, we found that roscovitine abolished the production of (*)NO induced by lipopolysaccharide (LPS). Moreover, roscovitine significantly inhibited LPS-induced inducible nitric oxide synthase (iNOS) mRNA and protein expression. Our data also showed that roscovitine attenuated LPS-induced phosphorylation of IkappaB kinase beta (IKKbeta), IkappaB, and p65 but enhanced the phosphorylation of ERK, p38, and c-Jun NH(2)-terminal kinase (JNK). In addition, roscovitine dose dependently inhibited LPS-induced expression of cyclooxygenase-2 (COX)-2, IL-1beta, and IL-6 but not tumor necrosis factor (TNF)-alpha. Tetrahydrobiopterin (BH(4)), an essential cofactor for iNOS, is easily oxidized to 7,8-dihydrobiopterin (BH(2)). Roscovitine significantly inhibited LPS-induced BH(4) biosynthesis and decreased BH(4)-to-BH(2) ratio. Furthermore, roscovitine greatly reduced the upregulation of GTP cyclohydrolase-1 (GCH-1), the rate-limiting enzyme for BH(4) biosynthesis. Using other CDK inhibitors, we found that CDK1, CDK5, and CDK7, but not CDK2, significantly inhibited LPS-induced (*)NO production in macrophages. Similarly, in isolated peritoneal macrophages, roscovitine strongly inhibited (*)NO production, iNOS, and COX-2 upregulation, activation of NFkappaB, and induction of GCH-1 by LPS. Together, our data indicate that roscovitine abolishes LPS-induced (*)NO production in macrophages by suppressing nuclear factor-kappaB activation and BH(4) biosynthesis, which might be mediated by CDK1, CDK5, and CDK7. Our results also suggest that roscovitine may inhibit inflammation and that CDKs may play important roles in the mechanisms by which roscovitine attenuates inflammation.
Collapse
Affiliation(s)
- Jianhai Du
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Pieper GM, Ionova IA, Cooley BC, Migrino RQ, Khanna AK, Whitsett J, Vásquez-Vivar J. Sepiapterin decreases acute rejection and apoptosis in cardiac transplants independently of changes in nitric oxide and inducible nitric-oxide synthase dimerization. J Pharmacol Exp Ther 2009; 329:890-9. [PMID: 19307452 PMCID: PMC2683770 DOI: 10.1124/jpet.108.148569] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 03/18/2009] [Indexed: 12/30/2022] Open
Abstract
Tetrahydrobiopterin (BH(4)), a cofactor of inducible nitric-oxide synthase (iNOS), is an important post-translational regulator of NO bioactivity. We examined whether treatment of cardiac allograft recipients with sepiapterin [S-(-)-2-amino-7,8-dihydro-6-(2-hydroxy-1-oxopropyl)-4-(1H)-pteridinone], a precursor of BH(4), inhibited acute rejection and apoptosis in cardiac transplants. Heterotopic cardiac transplantation was performed in Wistar-Furth donor to Lewis recipient strain rats. Recipients were treated daily after transplantation with 10 mg/kg sepiapterin. Grafts were harvested on post-transplant day 6 for analysis of BH(4) (high-performance liquid chromatography), expression of inflammatory cytokines (reverse transcription- and real-time polymerase chain reaction), iNOS (Western blots), and NO (Griess reaction and NO analyzer). Histological rejection grade was scored, and graft function was determined by echocardiography. Apoptosis, protein nitration, and oxidative stress were determined by immunohistochemistry. Treatment of allografts with sepiapterin increased cardiac BH(4) levels by 3-fold without changing protein levels of GTP cyclohydrolase, the enzyme that regulates de novo BH(4) synthesis. Sepiapterin decreased inflammatory cell infiltrate and significantly inhibited histological rejection scores and apoptosis similar in magnitude to cyclosporine. Sepiapterin also decreased nitrative and oxidative stress. Sepiapterin caused a smaller increase in left ventricular mass versus untreated allografts but without improving fractional shortening. Sepiapterin did not alter tumor necrosis factor-alpha and interferon-gamma expression, whereas it decreased interleukin (IL)-2 expression. Sepiapterin did not change total iNOS protein or monomer levels, or plasma and tissue NO metabolites levels. It is concluded that the mechanism(s) of antirejection are due in part to decreased apoptosis, protein nitration, and oxidation of cardiomyocytes, which seems to be mediated at the immune level by limiting inflammatory cell infiltration via decreased IL-2-mediated T-lymphocyte expansion.
Collapse
Affiliation(s)
- Galen M Pieper
- Division of Transplant Surgery, Medical College of Wisconsin, 9200 West Wisconsin Ave., Milwaukee, WI 53226, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
Ionova IA, Vásquez-Vivar J, Whitsett J, Herrnreiter A, Medhora M, Cooley BC, Pieper GM. Deficient BH4 production via de novo and salvage pathways regulates NO responses to cytokines in adult cardiac myocytes. Am J Physiol Heart Circ Physiol 2008; 295:H2178-87. [PMID: 18835915 PMCID: PMC2614582 DOI: 10.1152/ajpheart.00748.2008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Accepted: 09/25/2008] [Indexed: 12/25/2022]
Abstract
Adult rat cardiac myocytes typically display a phenotypic response to cytokines manifested by low or no increases in nitric oxide (NO) production via inducible NO synthase (iNOS) that distinguishes them from other cell types. To better characterize this response, we examined the expression of tetrahydrobiopterin (BH4)-synthesizing and arginine-utilizing genes in cytokine-stimulated adult cardiac myocytes. Intracellular BH4 and 7,8-dihydrobiopterin (BH2) and NO production were quantified. Cytokines induced GTP cyclohydrolase and its feedback regulatory protein but with deficient levels of BH4 synthesis. Despite the induction of iNOS protein, cytokine-stimulated adult cardiac myocytes produced little or no increase in NO versus unstimulated cells. Western blot analysis under nonreducing conditions revealed the presence of iNOS monomers. Supplementation with sepiapterin (a precursor of BH4) increased BH4 as well as BH2, but this did not enhance NO levels or eliminate iNOS monomers. Similar findings were confirmed in vivo after treatment of rat cardiac allograft recipients with sepiapterin. It was found that expression of dihydrofolate reductase, required for full activity of the salvage pathway, was not detected in adult cardiac myocytes. Thus, adult cardiac myocytes have a limited capacity to synthesize BH4 after cytokine stimulation. The mechanisms involve posttranslational factors impairing de novo and salvage pathways. These conditions are unable to support active iNOS protein dimers necessary for NO production. These findings raise significant new questions about the prevailing understanding of how cytokines, via iNOS, cause cardiac dysfunction and injury in vivo during cardiac inflammatory disease states since cardiac myocytes are not a major source of high NO production.
Collapse
Affiliation(s)
- Irina A Ionova
- Department of Surgery (Transplant Surgery), Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Angiopoietin-Related Growth Factor Enhances Blood Flow Via Activation of the ERK1/2-eNOS-NO Pathway in a Mouse Hind-Limb Ischemia Model. Arterioscler Thromb Vasc Biol 2008; 28:827-34. [DOI: 10.1161/atvbaha.107.149674] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Objective—
Transgenic mice overexpressing angiopoietin-related growth factor (AGF) exhibit enhanced angiogenesis, suggesting that AGF may be a useful drug target in ischemic disease. Our goal was to determine whether AGF enhances blood flow in a mouse hind-limb ischemia model and to define molecular mechanisms underlying AGF signaling in endothelial cells.
Methods and Results—
Intramuscular injection of adenovirus harboring AGF into the ischemic limb increased AGF production, which increased blood flow through induction of angiogenesis and arteriogenesis, thereby reducing the necessity for limb amputation. In vitro analysis showed that exposing human umbilical venous endothelial cells to AGF increased nitric oxide (NO) production through activation of an ERK1/2-endothelial NO synthetase (eNOS) signaling pathway. AGF-stimulated eNOS phosphorylation, NO production, and endothelial cell migration were all abolished by specific MEK1/2 inhibitors. Moreover, AGF did not restore blood flow to ischemic hind-limbs of either mice receiving NOS inhibitor L-NAME or eNOS knockout mice.
Conclusion—
Activation of an ERK1/2-eNOS-NO pathway is a crucial signaling mechanism by which AGF increases blood flow through induction of angiogenesis and arteriogenesis. Further investigation of the regulation underlying AGF signaling pathway may contribute to develop a new clinical strategy for ischemic vascular diseases.
Collapse
|
42
|
Suh SJ, Jin UH, Kim KW, Son JK, Lee SH, Son KH, Chang HW, Lee YC, Kim CH. Triterpenoid saponin, oleanolic acid 3-O-β-d-glucopyranosyl(1→3)-α-l-rhamnopyranosyl(1→2)-α-l-arabinopyranoside (OA) from Aralia elata inhibits LPS-induced nitric oxide production by down-regulated NF-κB in raw 264.7 cells. Arch Biochem Biophys 2007; 467:227-33. [PMID: 17910942 DOI: 10.1016/j.abb.2007.08.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 08/22/2007] [Accepted: 08/25/2007] [Indexed: 02/03/2023]
Abstract
It is well known that the pro-inflammatory mediators such as nitric oxide (NO) and prostaglandin (PG)E(2) are involved in several inflammatory diseases and lipopolysaccharide (LPS) can stimulate these inflammatory responses. Oleanolic acid 3-O-beta-d-glucopyranosyl(1-->3)-alpha-l-rhamnopyranosyl(1-->2)-alpha-l-arabinopyranoside (OA) was purified from edible plant Aralia elata. OA inhibited LPS-induced NO and PGE(2) production in raw 264.7 murine macrophages in a dose-dependent manner and RT-PCR analysis indicated OA inhibited mRNA transcriptions of iNOS and COX-2 genes in LPS-induced cells. EMSA and Western blot analysis revealed that OA drastically reduced NF-kappaB translocation by the inhibition effects of LPS-induced phosphorylation of IkappaBalpha. In addition, it was found that OA inhibited the phosphorylation of ERK1/2, p38 and JNK MAPK, and the treatment of U0126 in LPS-induced raw 264.7 cells showed significant inhibition activity on the NO production and the phosphorylation of IkappaBalpha. Taken together, it is suggested that OA from A. elata has an anti-inflammatory activity via down-regulation of NF-kappaB.
Collapse
Affiliation(s)
- Seok-Jong Suh
- Department of Biological Science, Sungkyunkwan University, Chunchun-Dong 300, Jangan-Gu, Suwon City, Kyunggi-Do 440-746, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sarić A, Balog T, Sobocanec S, Marotti T. Endomorphin 1 activates nitric oxide synthase 2 activity and downregulates nitric oxide synthase 2 mRNA expression. Neuroscience 2007; 144:1454-61. [PMID: 17197099 DOI: 10.1016/j.neuroscience.2006.11.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Revised: 11/07/2006] [Accepted: 11/12/2006] [Indexed: 11/20/2022]
Abstract
Endomorphins 1 and 2 are newly discovered opioid tetrapeptides whose structure is more resistant to enzymatic degradation than that of other opioid peptides. Endomorphins 1 and 2 are considered as endogenous ligands with a high affinity for mu receptors. A number of studies have shown that opioid peptides per se can induce release of nitric oxide from rodent and human immune cells. Endomorphins seemed to be involved in the process of vasodilatation by stimulating release of nitric oxide. In our study we stimulated in vitro J774 macrophages with different concentrations of endomorphin 1 or 2 for measuring nitric oxide release and nitric oxide synthase 2 (NOS 2) mRNA expression. Results showed that 48 h incubation did not enhance nitric oxide release when measured with the Griess method. On the other hand, using real-time amperometric detection of nitric oxide release shortly after challenge with endomorphins, we showed that only 10(-6) M endomorphin 1 was able to stimulate nitric oxide release from a J774 macrophage cell line by activation of NOS 2 isoenzyme. The peak release was 1000-1500 s after stimulation and was in the range of nitric oxide release stimulated with 10 microg/ml lipopolysaccharide. In contrast to this, endomorphin 2 failed to induce nitric oxide release in all tested concentrations. Using a specific inhibitor of nitric oxide synthase 2 (N-(3-[aminomethyl]benzyl)acetamidine, 1400W) we eliminated the stimulatory effect of endomorphin 1 on nitric oxide release. The expression of mRNA for NOS 2 in J774 macrophages, after 30 min incubation with either lipopolysaccharide or 10(-6) M endomorphin 1 was not upregulated. As expected, lipopolysaccharide induced de novo NOS 2 transcription within 4 h. At the same time, in contrast to lipopolysaccharide, mRNA expression of cells treated with endomorphin 1 was downregulated. Since a mu-opioid receptor specific antagonist beta-funaltrexamine hydrochloride inhibited nitric oxide release from endomorphin 1-treated cells, the effect seemed to be mu-opioid receptor mediated.
Collapse
MESH Headings
- Animals
- Cell Line
- Dose-Response Relationship, Drug
- Down-Regulation/drug effects
- Down-Regulation/physiology
- Enzyme Activation/drug effects
- Enzyme Activation/physiology
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation, Enzymologic/physiology
- Lipopolysaccharides/pharmacology
- Macrophages/drug effects
- Macrophages/enzymology
- Mice
- Narcotic Antagonists/pharmacology
- Nitric Oxide/biosynthesis
- Nitric Oxide Synthase Type II/drug effects
- Nitric Oxide Synthase Type II/genetics
- Nitric Oxide Synthase Type II/metabolism
- Oligopeptides/metabolism
- Oligopeptides/pharmacology
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- A Sarić
- Division of Molecular Medicine, Rudjer Bosković Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | | | | | | |
Collapse
|