101
|
Wang L, Hu J, Zhou H. Macrophage and Adipocyte Mitochondrial Dysfunction in Obesity-Induced Metabolic Diseases. World J Mens Health 2020; 39:606-614. [PMID: 33151047 PMCID: PMC8443980 DOI: 10.5534/wjmh.200163] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022] Open
Abstract
Obesity is one of major health burdens of modern society as it contributes to the growing prevalence of its related comorbidities, such as diabetes, cardiovascular diseases, and some cancers. A series of innate immune cells, especially macrophages, and adipocytes have been implicated in the pathogenesis of obesity. Mitochondrial dysfunction, which is induced by obesity, are critical mediators in initiating inflammation in macrophages and adipocytes, and subsequent systemic insulin resistance. In this review, we discuss new findings on how obesity impairs mitochondrial function in macrophages and adipocytes and how this dysfunction contributes to obesity and its comorbidities. We also summarize drugs that treat metabolic diseases by targeting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Liwen Wang
- Department of Metabolism and Endocrinology, the Second Xiangya Hospital, Central South University, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, the Second Xiangya Hospital, Central South University, Hunan, China
| | - Jie Hu
- Department of Metabolism and Endocrinology, the Second Xiangya Hospital, Central South University, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, the Second Xiangya Hospital, Central South University, Hunan, China
| | - Haiyan Zhou
- Department of Metabolism and Endocrinology, the Second Xiangya Hospital, Central South University, Hunan, China.,National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Metabolic Syndrome Research Center, the Second Xiangya Hospital, Central South University, Hunan, China.
| |
Collapse
|
102
|
Nieman DC, Pence BD. Exercise immunology: Future directions. JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:432-445. [PMID: 32928447 PMCID: PMC7498623 DOI: 10.1016/j.jshs.2019.12.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 05/07/2023]
Abstract
Several decades of research in the area of exercise immunology have shown that the immune system is highly responsive to acute and chronic exercise training. Moderate exercise bouts enhance immunosurveillance and when repeated over time mediate multiple health benefits. Most of the studies prior to 2010 relied on a few targeted outcomes related to immune function. During the past decade, technologic advances have created opportunities for a multi-omics and systems biology approach to exercise immunology. This article provides an overview of metabolomics, lipidomics, and proteomics as they pertain to exercise immunology, with a focus on immunometabolism. This review also summarizes how the composition and diversity of the gut microbiota can be influenced by exercise, with applications to human health and immunity. Exercise-induced improvements in immune function may play a critical role in countering immunosenescence and the development of chronic diseases, and emerging omics technologies will more clearly define the underlying mechanisms. This review summarizes what is currently known regarding a multi-omics approach to exercise immunology and provides future directions for investigators.
Collapse
Affiliation(s)
- David C Nieman
- Human Performance Laboratory, Appalachian State University, North Carolina Research Campus, Kannapolis, NC 28081, USA.
| | - Brandt D Pence
- School of Health Studies, University of Memphis, Memphis, TN 38152, USA
| |
Collapse
|
103
|
Gojanovich GS, Jacobson DL, Jao J, Russell JS, Van Dyke RB, Libutti DE, Sharma TS, Geffner ME, Gerschenson M. Mitochondrial Dysfunction and Insulin Resistance in Pubertal Youth Living with Perinatally Acquired HIV. AIDS Res Hum Retroviruses 2020; 36:703-711. [PMID: 32586116 DOI: 10.1089/aid.2020.0067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial dysfunction (MD) is linked to cardiometabolic complications, such as obesity and insulin resistance (IR), the frequencies of which are higher in adults living with HIV infection and receiving combination antiretroviral therapies (ARV). ARV-treated youth living with perinatally acquired HIV infection (YLPHIV) may be especially susceptible to IR due to long-term exposure to both factors. Medical histories, fasting blood chemistry panels, and mitochondrial function in banked peripheral blood mononuclear cells (PBMCs) were assessed in eligible YLPHIV from the Pediatric HIV/AIDS Cohort Study (PHACS)/Adolescent Master Protocol (AMP) Mitochondrial Determinants Component cohort, stratified by Homeostatic Model Assessment of IR (HOMA-IR) score: case (score ≥4, n = 39) or control (score <4, n = 105). PBMCs were sources for mitochondrial (mt) DNA copies/cell; mtRNA transcript levels of oxidative phosphorylation (OXPHOS) subunits NADH dehydrogenases 1 and 6, and cytochrome B; and enzymatic activities of OXPHOS Complexes I (CI) and IV (CIV). Logistic regression models were fit to estimate the odds of IR case diagnosis, adjusted for sex, race/ethnicity, body mass index (BMI) z-score, and Tanner stage. IR cases were similar to controls by age, sex, and race/ethnicity. Cases had higher median levels of peak HIV viral load, lactate, pyruvate, triglycerides, and BMI z-scores. OXPHOS CI enzymatic activity was lower in cases (log10 1.62 vs. 1.70) and inversely correlated with HOMA-IR score (r = -0.157, p = .061), but did not associate with IR in adjusted models. Fully adjusted models indicated associations of nadir CD4% [odds ratio (OR) = 0.95, 95% confidence intervals (CIs) = 0.90-1.00] or peak HIV load (OR = 3.48, 95% CIs = 1.70-10.79) with IR. IR in YLPHIV was strongly associated with morphometrics, but early virologic and immunologic factors may also influence MD.
Collapse
Affiliation(s)
- Greg S. Gojanovich
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Denise L. Jacobson
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jennifer Jao
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jonathan S. Russell
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Russell B. Van Dyke
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Daniel E. Libutti
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Tanvi S. Sharma
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mitchell E. Geffner
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | | |
Collapse
|
104
|
Li M, Duan L, Cai YL, Li HY, Hao BC, Chen JQ, Liu HB. Growth differentiation factor-15 is associated with cardiovascular outcomes in patients with coronary artery disease. Cardiovasc Diabetol 2020; 19:120. [PMID: 32746821 PMCID: PMC7398317 DOI: 10.1186/s12933-020-01092-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022] Open
Abstract
Background Growth differentiation factor-15 (GDF-15) is a marker of inflammation, oxidative stress and it is associated with adverse prognosis in cardiovascular disease. The aim of the present cohort study is to investigate the prognostic value of GDF-15 in patients with coronary artery disease (CAD) during long-term follow up. Methods A total of 3641 consecutive patients with CAD were prospectively enrolled into the study and followed up for major adverse cardiovascular events (MACEs) and all-cause death up to 5.3–7.6 years. Plasma GDF-15 was measured and clinical data and long-term events were registered. The patients were subsequently divided into three groups by the levels of GDF-15 and the prognostic value of GDF-15 level with MACEs and all-cause death was evaluated. Results After a median follow-up at 6.4 years later, 775 patients (event rate of 21%) had developed MACEs and 275 patients died (event rate of 7.55%). Kaplan–Meier analysis indicated that the patients with GDF-15 > 1800 ng/L were significantly associated with an increased risk of MACEs and all-cause death. Cox regression analysis indicated that GDF-15 > 1800 ng/L were independently associated with the composite of MACEs (HR 1.74; 95% CI 1.44–2.02; P < 0.001) and all-cause death (HR 2.04; 95% CI 1.57–2.61; P < 0.001). For MACEs, GDF-15 significantly improved the C-statistic (area under the curve, 0.583 [95% CI 0.559–0.606] to 0.628 [0.605–0.651]; P < 0.001), net reclassification index (0.578; P = 0.031), and integrated discrimination index (0.021; P = 0.027). For all-cause death, GDF-15 significantly improved the C-statistic (0.728 [95% CI 0.694–0.761] to 0.817 [0.781–0.846]; P < 0.001), net reclassification index (0.629; P = 0.001), and integrated discrimination index (0.035; P = 0.002). Conclusions In the setting of CAD, GDF-15 is associated with long-term MACEs and all-cause death, and provides incremental prognostic value beyond traditional risks factors.
Collapse
Affiliation(s)
- Man Li
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lei Duan
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yu-Lun Cai
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hui-Ying Li
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ben-Chuan Hao
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jian-Qiao Chen
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong-Bin Liu
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China. .,Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Beijing, China.
| |
Collapse
|
105
|
Moon JS, Goeminne LJE, Kim JT, Tian JW, Kim S, Nga HT, Kang SG, Kang BE, Byun J, Lee Y, Jeon J, Shong M, Auwerx J, Ryu D, Yi H. Growth differentiation factor 15 protects against the aging-mediated systemic inflammatory response in humans and mice. Aging Cell 2020; 19:e13195. [PMID: 32691494 PMCID: PMC7431835 DOI: 10.1111/acel.13195] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial dysfunction is associated with aging-mediated inflammatory responses, leading to metabolic deterioration, development of insulin resistance, and type 2 diabetes. Growth differentiation factor 15 (GDF15) is an important mitokine generated in response to mitochondrial stress and dysfunction; however, the implications of GDF15 to the aging process are poorly understood in mammals. In this study, we identified a link between mitochondrial stress-induced GDF15 production and protection from tissue inflammation on aging in humans and mice. We observed an increase in serum levels and hepatic expression of GDF15 as well as pro-inflammatory cytokines in elderly subjects. Circulating levels of cell-free mitochondrial DNA were significantly higher in elderly subjects with elevated serum levels of GDF15. In the BXD mouse reference population, mice with metabolic impairments and shorter survival were found to exhibit higher hepatic Gdf15 expression. Mendelian randomization links reduced GDF15 expression in human blood to increased body weight and inflammation. GDF15 deficiency promotes tissue inflammation by increasing the activation of resident immune cells in metabolic organs, such as in the liver and adipose tissues of 20-month-old mice. Aging also results in more severe liver injury and hepatic fat deposition in Gdf15-deficient mice. Although GDF15 is not required for Th17 cell differentiation and IL-17 production in Th17 cells, GDF15 contributes to regulatory T-cell-mediated suppression of conventional T-cell activation and inflammatory cytokines. Taken together, these data reveal that GDF15 is indispensable for attenuating aging-mediated local and systemic inflammation, thereby maintaining glucose homeostasis and insulin sensitivity in humans and mice.
Collapse
Affiliation(s)
- Ji Sun Moon
- Research Center for Endocrine and Metabolic DiseasesChungnam National University HospitalChungnam National University School of MedicineDaejeonRepublic of Korea
| | - Ludger J. E. Goeminne
- Laboratory of Integrative Systems PhysiologyÉcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Jung Tae Kim
- Research Center for Endocrine and Metabolic DiseasesChungnam National University HospitalChungnam National University School of MedicineDaejeonRepublic of Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonRepublic of Korea
| | - Jing Wen Tian
- Research Center for Endocrine and Metabolic DiseasesChungnam National University HospitalChungnam National University School of MedicineDaejeonRepublic of Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonRepublic of Korea
| | - Seok‐Hwan Kim
- Department of SurgeryChungnam National University School of MedicineDaejeonRepublic of Korea
| | - Ha Thi Nga
- Research Center for Endocrine and Metabolic DiseasesChungnam National University HospitalChungnam National University School of MedicineDaejeonRepublic of Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonRepublic of Korea
| | - Seul Gi Kang
- Research Center for Endocrine and Metabolic DiseasesChungnam National University HospitalChungnam National University School of MedicineDaejeonRepublic of Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonRepublic of Korea
| | - Baeki E. Kang
- Department of Molecular Cell BiologySungkyunkwan University School of MedicineSuwonRepublic of Korea
| | - Jin‐Seok Byun
- Department of Oral MedicineSchool of DentistryKyungpook National UniversityDaeguRepublic of Korea
| | - Young‐Sun Lee
- Department of Internal MedicineKorea University College of MedicineSeoulRepublic of Korea
| | - Jae‐Han Jeon
- Department of Internal MedicineSchool of MedicineKyungpook National UniversityDaeguKorea
| | - Minho Shong
- Research Center for Endocrine and Metabolic DiseasesChungnam National University HospitalChungnam National University School of MedicineDaejeonRepublic of Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonRepublic of Korea
| | - Johan Auwerx
- Laboratory of Integrative Systems PhysiologyÉcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Dongryeol Ryu
- Department of Molecular Cell BiologySungkyunkwan University School of MedicineSuwonRepublic of Korea
- Biomedical Institute for Convergence at SKKU (BICS)Sungkyunkwan UniversitySuwonRepublic of Korea
- Samsung Biomedical Research InstituteSamsung Medical CenterSeoulRepublic of Korea
| | - Hyon‐Seung Yi
- Research Center for Endocrine and Metabolic DiseasesChungnam National University HospitalChungnam National University School of MedicineDaejeonRepublic of Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonRepublic of Korea
| |
Collapse
|
106
|
Pereiro P, Librán-Pérez M, Figueras A, Novoa B. Conserved function of zebrafish (Danio rerio) Gdf15 as a sepsis tolerance mediator. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 109:103698. [PMID: 32289326 DOI: 10.1016/j.dci.2020.103698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 06/11/2023]
Abstract
GDF15 is frequently detected in patients suffering from various diseases, especially those associated with pro-inflammatory processes and/or metabolic disorders. Accordingly, sepsis, whose major complications are related to metabolic alterations and systemic inflammation, significantly increases the secretion of GDF15. Indeed, this cytokine could be considered a marker of sepsis severity. However, until the last several years, the involvement of GDF15 in these disorders had not been widely characterized. In mice, GDF15 was recently described as a pivotal inducer of sepsis tolerance by mediating metabolic alterations that reduce tissue damage. In this work we describe a zebrafish gdf15 gene. We found that gdf15 follows an expression pattern similar to that observed in mammals, being highly expressed in the liver and kidney and induced after pro-inflammatory stimuli. Moreover, larvae overexpressing gdf15 were more resistant to bacterial and viral challenges without affecting the pathogen load. Consequently, Gdf15 also protected zebrafish larvae against LPS-induced mortality. As in mice, zebrafish Gdf15 seems to induce sepsis tolerance by altering the metabolic parameters of the individuals.
Collapse
Affiliation(s)
- Patricia Pereiro
- Instituto de Investigaciones Marinas (IIM-CSIC), C/Eduardo Cabello, 6, 36208, Vigo, Spain.
| | - Marta Librán-Pérez
- Instituto de Investigaciones Marinas (IIM-CSIC), C/Eduardo Cabello, 6, 36208, Vigo, Spain.
| | - Antonio Figueras
- Instituto de Investigaciones Marinas (IIM-CSIC), C/Eduardo Cabello, 6, 36208, Vigo, Spain.
| | - Beatriz Novoa
- Instituto de Investigaciones Marinas (IIM-CSIC), C/Eduardo Cabello, 6, 36208, Vigo, Spain.
| |
Collapse
|
107
|
Chang JY, Hong HJ, Kang SG, Kim JT, Zhang BY, Shong M. The Role of Growth Differentiation Factor 15 in Energy Metabolism. Diabetes Metab J 2020; 44:363-371. [PMID: 32613776 PMCID: PMC7332323 DOI: 10.4093/dmj.2020.0087] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is receiving great interest beyond its role as an aging and disease-related biomarker. Recent discovery of its receptor, glial cell line-derived neurotrophic factor (GDNF) family receptor α-like (GFRAL), suggests a central role in appetite regulation. However, there is also considerable evidence that GDF15 may have peripheral activity through an as-of-yet undiscovered mode of action. This raises the question as to whether increased GDF15 induction during pathophysiologic conditions also suppresses appetite. The present review will briefly introduce the discovery of GDF15 and describe the different contexts under which GDF15 is induced, focusing on its induction during mitochondrial dysfunction. We will further discuss the metabolic role of GDF15 under various pathophysiological conditions and conclude with possible therapeutic applications.
Collapse
Affiliation(s)
- Joon Young Chang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyun Jung Hong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seul Gi Kang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Jung Tae Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ben Yuan Zhang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea.
| |
Collapse
|
108
|
Wang X, Wang Q, Li W, Zhang Q, Jiang Y, Guo D, Sun X, Lu W, Li C, Wang Y. TFEB-NF-κB inflammatory signaling axis: a novel therapeutic pathway of Dihydrotanshinone I in doxorubicin-induced cardiotoxicity. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:93. [PMID: 32448281 PMCID: PMC7245789 DOI: 10.1186/s13046-020-01595-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Doxorubicin is effective in a variety of solid and hematological malignancies. Unfortunately, clinical application of doxorubicin is limited due to a cumulative dose-dependent cardiotoxicity. Dihydrotanshinone I (DHT) is a natural product from Salvia miltiorrhiza Bunge with multiple anti-tumor activity and anti-inflammation effects. However, its anti-doxorubicin-induced cardiotoxicity (DIC) effect, either in vivo or in vitro, has not been elucidated yet. This study aims to explore the anti-inflammation effects of DHT against DIC, and to elucidate the potential regulatory mechanism. METHODS Effects of DHT on DIC were assessed in zebrafish, C57BL/6 mice and H9C2 cardiomyocytes. Echocardiography, histological examination, flow cytometry, immunochemistry and immunofluorescence were utilized to evaluate cardio-protective effects and anti-inflammation effects. mTOR agonist and lentivirus vector carrying GFP-TFEB were applied to explore the regulatory signaling pathway. RESULTS DHT improved cardiac function via inhibiting the activation of M1 macrophages and the excessive release of pro-inflammatory cytokines both in vivo and in vitro. The activation and nuclear localization of NF-κB were suppressed by DHT, and the effect was abolished by mTOR agonist with concomitant reduced expression of nuclear TFEB. Furthermore, reduced expression of nuclear TFEB is accompanied by up-regulated phosphorylation of IKKα/β and NF-κB, while TFEB overexpression reversed these changes. Intriguingly, DHT could upregulate nuclear expression of TFEB and reduce expressions of p-IKKα/β and p-NF-κB. CONCLUSIONS Our results demonstrated that DHT can be applied as a novel cardioprotective compound in the anti-inflammation management of DIC via mTOR-TFEB-NF-κB signaling pathway. The current study implicates TFEB-IKK-NF-κB signaling axis as a previously undescribed, druggable pathway for DIC.
Collapse
Affiliation(s)
- Xiaoping Wang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Qiyan Wang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Weili Li
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Qian Zhang
- grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yanyan Jiang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Dongqing Guo
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Xiaoqian Sun
- grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Wenji Lu
- grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Chun Li
- grid.24695.3c0000 0001 1431 9176Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yong Wang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China ,grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| |
Collapse
|
109
|
Wischhusen J, Melero I, Fridman WH. Growth/Differentiation Factor-15 (GDF-15): From Biomarker to Novel Targetable Immune Checkpoint. Front Immunol 2020; 11:951. [PMID: 32508832 PMCID: PMC7248355 DOI: 10.3389/fimmu.2020.00951] [Citation(s) in RCA: 302] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Growth/differentiation factor-15 (GDF-15), also named macrophage inhibitory cytokine-1, is a divergent member of the transforming growth factor β superfamily. While physiological expression is barely detectable in most somatic tissues in humans, GDF-15 is abundant in placenta. Elsewhere, GDF-15 is often induced under stress conditions, seemingly to maintain cell and tissue homeostasis; however, a moderate increase in GDF-15 blood levels is observed with age. Highly elevated GDF-15 levels are mostly linked to pathological conditions including inflammation, myocardial ischemia, and notably cancer. GDF-15 has thus been widely explored as a biomarker for disease prognosis. Mechanistically, induction of anorexia via the brainstem-restricted GDF-15 receptor GFRAL (glial cell-derived neurotrophic factor [GDNF] family receptor α-like) is well-documented. GDF-15 and GFRAL have thus become attractive targets for metabolic intervention. Still, several GDF-15 mediated effects (including its physiological role in pregnancy) are difficult to explain via the described pathway. Hence, there is a clear need to better understand non-metabolic effects of GDF-15. With particular emphasis on its immunomodulatory potential this review discusses the roles of GDF-15 in pregnancy and in pathological conditions including myocardial infarction, autoimmune disease, and specifically cancer. Importantly, the strong predictive value of GDF-15 as biomarker may plausibly be linked to its immune-regulatory function. The described associations and mechanistic data support the hypothesis that GDF-15 acts as immune checkpoint and is thus an emerging target for cancer immunotherapy.
Collapse
Affiliation(s)
- Jörg Wischhusen
- Experimental Tumor Immunology, Department of Obstetrics and Gynecology, University of Würzburg Medical School, Würzburg, Germany
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain
- Immunology and Immunotherapy Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Wolf Herman Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Université de Paris, Sorbonne Université Team Cancer, Immune Control and Escape, Paris, France
| |
Collapse
|
110
|
Growth differentiation factor 15 facilitates lung fibrosis by activating macrophages and fibroblasts. Exp Cell Res 2020; 391:112010. [PMID: 32305327 DOI: 10.1016/j.yexcr.2020.112010] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
Lung fibrosis is a devastating disease characterized by fibroblast accumulation and extracellular matrix deposition in lungs. However, its molecular and cellular pathogenesis is not fully understood and the current therapeutic strategies are ineffective. Bleomycin-induced lung fibrosis is the most widely used experimental model for research aimed at in-depth analysis of lung fibrosis mechanisms. The present study aimed to analyse the effects of growth differentiation factor 15 (GDF15), which is associated with many diseases, in lung fibrosis. GDF15 mRNA expression was elevated in the lungs of bleomycin-treated mice, revealed by comprehensive gene analysis. Its protein levels were also increased in the lungs, bronchoalveolar lavage fluid, and plasma obtained from bleomycin-treated mice as compared to those in saline-treated mice. Bleomycin administration in mice resulted in a marked increase in senescence-associated β-galactosidase-positive and p16INK4a-positive lung structural cells including alveolar epithelial cells and macrophages. Immunohistochemical staining using anti-GDF15 antibody and increased mRNA expression of GDF15 in bleomycin-induced senescent A549 cells indicated that GDF15 is produced from alveolar epithelial cells undergoing bleomycin-induced cellular senescence. GDF15 was also implicated in the augmentation of interleukin-4/interleukin-13-induced mRNA expression of M2 markers including arginase 1 and chitinase-3-like protein and was also responsible for increased α-smooth muscle actin expression through the ALK5-Smad2/3 pathway in WI-38 lung fibroblasts. Therefore, GDF15 secreted from senescent alveolar epithelial cells might act as a profibrotic factor through activation of M2 macrophages and fibroblasts. This implies that GDF15 could be a potential therapeutic target and a predictor of lung fibrosis progression.
Collapse
|
111
|
Choi MJ, Jung SB, Lee SE, Kang SG, Lee JH, Ryu MJ, Chung HK, Chang JY, Kim YK, Hong HJ, Kim H, Kim HJ, Lee CH, Mardinoglu A, Yi HS, Shong M. An adipocyte-specific defect in oxidative phosphorylation increases systemic energy expenditure and protects against diet-induced obesity in mouse models. Diabetologia 2020; 63:837-852. [PMID: 31925461 DOI: 10.1007/s00125-019-05082-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/30/2019] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS Mitochondrial oxidative phosphorylation (OxPhos) is essential for energy production and survival. However, the tissue-specific and systemic metabolic effects of OxPhos function in adipocytes remain incompletely understood. METHODS We used adipocyte-specific Crif1 (also known as Gadd45gip1) knockout (AdKO) mice with decreased adipocyte OxPhos function. AdKO mice fed a normal chow or high-fat diet were evaluated for glucose homeostasis, weight gain and energy expenditure (EE). RNA sequencing of adipose tissues was used to identify the key mitokines affected in AdKO mice, which included fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15). For in vitro analysis, doxycycline was used to pharmacologically decrease OxPhos in 3T3L1 adipocytes. To identify the effects of GDF15 and FGF21 on the metabolic phenotype of AdKO mice, we generated AdKO mice with global Gdf15 knockout (AdGKO) or global Fgf21 knockout (AdFKO). RESULTS Under high-fat diet conditions, AdKO mice were resistant to weight gain and exhibited higher EE and improved glucose tolerance. In vitro pharmacological and in vivo genetic inhibition of OxPhos in adipocytes significantly upregulated mitochondrial unfolded protein response-related genes and secretion of mitokines such as GDF15 and FGF21. We evaluated the metabolic phenotypes of AdGKO and AdFKO mice, revealing that GDF15 and FGF21 differentially regulated energy homeostasis in AdKO mice. Both mitokines had beneficial effects on obesity and insulin resistance in the context of decreased adipocyte OxPhos, but only GDF15 regulated EE in AdKO mice. CONCLUSIONS/INTERPRETATION The present study demonstrated that the adipose tissue adaptive mitochondrial stress response affected systemic energy homeostasis via cell-autonomous and non-cell-autonomous pathways. We identified novel roles for adipose OxPhos and adipo-mitokines in the regulation of systemic glucose homeostasis and EE, which facilitated adaptation of an organism to local mitochondrial stress.
Collapse
Affiliation(s)
- Min Jeong Choi
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Saet-Byel Jung
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
| | - Seong Eun Lee
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
| | - Seul Gi Kang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Ju Hee Lee
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, 35015, South Korea
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Hyo Kyun Chung
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
| | - Joon Young Chang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Yong Kyung Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
| | - Hyun Jung Hong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Hyun Jin Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, 35015, South Korea
| | - Chul-Ho Lee
- Animal Model Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Hyon-Seung Yi
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, 35015, South Korea.
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea.
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, 35015, South Korea.
| |
Collapse
|
112
|
Xiang LX, Ran Q, Chen L, Xiang Y, Li FJ, Zhang XM, Xiao YN, Zou LY, Zhong JF, Li SC, Li ZJ. CR6-interacting factor-1 contributes to osteoclastogenesis by inducing receptor activator of nuclear factor κB ligand after radiation. World J Stem Cells 2020; 12:222-240. [PMID: 32266053 PMCID: PMC7118287 DOI: 10.4252/wjsc.v12.i3.222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/09/2020] [Accepted: 03/15/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Radiation induces rapid bone loss and enhances bone resorption and adipogenesis, leading to an increased risk of bone fracture. There is still a lack of effective preventive or therapeutic method for irradiation-induced bone injury. Receptor activator of nuclear factor κB ligand (RANKL) provides the crucial signal to induce osteoclast differentiation and plays an important role in bone resorption. However, the mechanisms of radiation-induced osteoporosis are not fully understood. AIM To investigate the role of CR6-interacting factor-1 (Crif1) in osteoclastogenesis after radiation and its possible mechanism. METHODS C57BL/6 mice were exposed to Co-60 gamma rays and received 5 Gy of whole-body sublethal irradiation at a rate of 0.69 Gy/min. For in vitro study, mouse bone marrow mesenchymal stem/stromal cells (BM-MSCs) were irradiated with Co-60 at a single dose of 9 Gy. For osteoclast induction, monocyte-macrophage RAW264.7 cells were cocultured with mouse BM-MSCs for 7 d. ClusPro and InterProSurf were used to investigate the interaction interface in Crif1 and protein kinase cyclic adenosine monophosphate (cAMP)-activited catalytic subunit alpha complex. Virtual screening using 462608 compounds from the Life Chemicals database around His120 of Crif1 was carried out using the program Autodock_vina. A tetrazolium salt (WST-8) assay was carried out to study the toxicity of compounds to different cells, including human BM-MSCs, mouse BM-MSCs, and Vero cells. RESULTS Crif1 expression increased in bone marrow cells after radiation in mice. Overexpression of Crif1 in mouse BM-MSCs and radiation exposure could increase RANKL secretion and promote osteoclastogenesis in vitro. Deletion of Crif1 in BM-MSCs could reduce both adipogenesis and RANKL expression, resulting in the inhibition of osteoclastogenesis. Deletion of Crif1 in RAW264.7 cells did not affect the receptor activator of nuclear factor κB expression or osteoclast differentiation. Following treatment with protein kinase A (PKA) agonist (forskolin) and inhibitor (H-89) in mouse BM-MSCs, Crif1 induced RANKL secretion via the cAMP/PKA pathway. Moreover, we identified the Crif1-protein kinase cyclic adenosine monophosphate-activited catalytic subunit alpha interaction interface by in silico studies and shortlisted interface inhibitors through virtual screening on Crif1. Five compounds dramatically suppressed RANKL secretion and adipogenesis by inhibiting the cAMP/PKA pathway. CONCLUSION Crif1 promotes RANKL expression via the cAMP/PKA pathway, which induces osteoclastogenesis by binding to receptor activator of nuclear factor κB on monocytes-macrophages in the mouse model. These results suggest a role for Crif1 in modulating osteoclastogenesis and provide insights into potential therapeutic strategies targeting the balance between osteogenesis and adipogenesis for radiation-induced bone injury.
Collapse
Affiliation(s)
- Li-Xin Xiang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Qian Ran
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Li Chen
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yang Xiang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Feng-Jie Li
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xiao-Mei Zhang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yan-Ni Xiao
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ling-Yun Zou
- Bioinformatics Center, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Jiang F Zhong
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Shengwen Calvin Li
- CHOC Children's Research Institute, Children's Hospital of Orange County, University of California, Irvine, CA 92868, United States
| | - Zhong-Jun Li
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
113
|
Nakayasu ES, Syed F, Tersey SA, Gritsenko MA, Mitchell HD, Chan CY, Dirice E, Turatsinze JV, Cui Y, Kulkarni RN, Eizirik DL, Qian WJ, Webb-Robertson BJM, Evans-Molina C, Mirmira RG, Metz TO. Comprehensive Proteomics Analysis of Stressed Human Islets Identifies GDF15 as a Target for Type 1 Diabetes Intervention. Cell Metab 2020; 31:363-374.e6. [PMID: 31928885 PMCID: PMC7319177 DOI: 10.1016/j.cmet.2019.12.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 09/03/2019] [Accepted: 12/12/2019] [Indexed: 01/03/2023]
Abstract
Type 1 diabetes (T1D) results from the progressive loss of β cells, a process propagated by pro-inflammatory cytokine signaling that disrupts the balance between pro- and anti-apoptotic proteins. To identify proteins involved in this process, we performed comprehensive proteomics of human pancreatic islets treated with interleukin-1β and interferon-γ, leading to the identification of 11,324 proteins, of which 387 were significantly regulated by treatment. We then tested the function of growth/differentiation factor 15 (GDF15), which was repressed by the treatment. We found that GDF15 translation was blocked during inflammation, and it was depleted in islets from individuals with T1D. The addition of exogenous GDF15 inhibited interleukin-1β+interferon-γ-induced apoptosis of human islets. Administration of GDF15 reduced by 53% the incidence of diabetes in NOD mice. Our approach provides a unique resource for the identification of the human islet proteins regulated by cytokines and was effective in discovering a potential target for T1D therapy.
Collapse
Affiliation(s)
- Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Farooq Syed
- Center for Diabetes and Metabolic Diseases and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sarah A Tersey
- Center for Diabetes and Metabolic Diseases and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Hugh D Mitchell
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Chi Yuet Chan
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ercument Dirice
- Department of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, and Harvard Stem Cell Institute, Boston, MA, USA
| | - Jean-Valery Turatsinze
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Yi Cui
- Environmental and Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Rohit N Kulkarni
- Department of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, and Harvard Stem Cell Institute, Boston, MA, USA
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Bobbie-Jo M Webb-Robertson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA; Computing and Analytics Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Raghavendra G Mirmira
- Center for Diabetes and Metabolic Diseases and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
114
|
Lou K, Huang P, Ma H, Wang X, Xu H, Wang W. Orlistat increases arsenite tolerance in THP-1 derived macrophages through the up-regulation of ABCA1. Drug Chem Toxicol 2019; 45:274-282. [PMID: 31665930 DOI: 10.1080/01480545.2019.1683571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Orlistat is an FDA-approved over-the-counter drug to treat obesity through the inhibition of lipase activity. Macrophages, which express high levels of lipoprotein lipase (LPL), are important phagocytes in the innate immune system. Our previous studies indicated that environmentally relevant concentrations of arsenite (As+3) could inhibit the major immune functions of macrophages. As the down-regulation of LPL is known to increase the expression of ABCA1, the cholesterol exporter demonstrated to be related to the resistance of arsenic toxicity. We examined if orlistat could reverse the inhibitive effects of As+3 on macrophage functions. The results showed that 50 μM orlistat reversed As+3-induced suppressions on phagocytosis, NO production and cytokine secretion in THP-1 derived macrophages. The expression of ABCA1 was significantly increased by orlistat in As+3 co-treated macrophages, which was associated with decreased intracellular As+3 levels. Collectively, these results indicated that orlistat could reverse the suppressive effects induced by As+3 in macrophages through the increased expression of ABCA1, which has the potential to be developed as a therapeutic agent for arsenic-induced immunosuppression.
Collapse
Affiliation(s)
- Kaiyan Lou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Department of Pharmaceutical Sciences, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Ping Huang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Department of Pharmaceutical Sciences, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Huijuan Ma
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Department of Pharmaceutical Sciences, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiaolei Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Department of Pharmaceutical Sciences, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Huan Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Department of Pharmaceutical Sciences, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wei Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Department of Pharmaceutical Sciences, School of Pharmacy, East China University of Science and Technology, Shanghai, China.,Department of Pharmacology and Toxicology and BIO5 Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
115
|
Tang M, Luo M, Lu W, Wang S, Zhang R, Liang W, Gu J, Yu X, Zhang X, Hu C. Serum growth differentiation factor 15 is associated with glucose metabolism in the third trimester in Chinese pregnant women. Diabetes Res Clin Pract 2019; 156:107823. [PMID: 31446114 DOI: 10.1016/j.diabres.2019.107823] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/17/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Growth differentiation factor 15 (GDF15) has been demonstrated to increase in diabetes as a protective factor. However, studies assessing relationships between GDF15 levels and gestational diabetes mellitus (GDM) are limited. In this study, we aimed to investigate whether GDF15 levels are related to GDM in Chinese subjects. METHODS We included 200 GDM patients and 200 matched normal controls in the second trimester as well as 130 GDM patients and 130 matched normal controls in the third trimester. Serum GDF15 levels of all participants were determined using an enzyme-linked immunosorbent assay (ELISA). Then, according to GDF15 levels, we equally divided the participants in the second and third trimesters into four subgroups respectively. The relationships of serum GDF15 levels with glucolipid metabolism indicators were analyzed. RESULTS In the third trimester, GDF15 levels were significantly higher in the GDM patients than in the normal controls (P < 0.001). Additionally, fasting blood glucose (FBG), 1-h postprandial glucose (1h-PG), 2-h postprandial glucose (2h-PG), hemoglobin A1C (HbA1c) and area under curve of glucose (AUCG) from the 75-g oral glucose tolerance test (OGTT) were positively associated with GDF15 levels (P < 0.05), even after adjusting for age, pregestational BMI, changes of BMI until the third trimester, gestational age, twin and family history of diabetes. Moreover, GDF15 levels were higher in the third trimester than in the second trimester (P < 0.001). No significant relationships were found between GDF15 levels and glucolipid metabolism in the second trimester (P > 0.05). CONCLUSIONS Serum GDF15 levels were positively correlated with glucose metabolism in the third trimester in Chinese pregnant women.
Collapse
Affiliation(s)
- Mengyang Tang
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Mingjuan Luo
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Endocrinology, University of Hong Kong Shenzhen Hospital, China
| | - Wenqian Lu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shiyun Wang
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Wei Liang
- Department of Endocrinology, University of Hong Kong Shenzhen Hospital, China
| | - Jianfen Gu
- Department of Endocrinology, University of Hong Kong Shenzhen Hospital, China
| | - Xuemei Yu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xueli Zhang
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Cheng Hu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China; Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China.
| |
Collapse
|
116
|
Kang YE, Kim HJ, Shong M. Regulation of Systemic Glucose Homeostasis by T Helper Type 2 Cytokines. Diabetes Metab J 2019; 43:549-559. [PMID: 31694077 PMCID: PMC6834846 DOI: 10.4093/dmj.2019.0157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/16/2019] [Indexed: 12/25/2022] Open
Abstract
Obesity results in an inflammatory microenvironment in adipose tissue, leading to the deterioration of tissue protective mechanisms. Although recent studies suggested the importance of type 2 immunity in an anti-inflammatory microenvironment in adipose tissue, the regulatory effects of T helper 2 (Th2) cytokines on systemic metabolic regulation are not fully understood. Recently, we identified the roles of the Th2 cytokine (interleukin 4 [IL-4] and IL-13)-induced adipokine, growth differentiation factor 15 (GDF15), in adipose tissue in regulating systemic glucose metabolism via signal transducer and activator of transcription 6 (STAT6) activation. Moreover, we showed that mitochondrial oxidative phosphorylation is required to maintain these macrophage-regulating autocrine and paracrine signaling pathways via Th2 cytokine-induced secretion of GDF15. In this review, we discuss how the type 2 immune response and Th2 cytokines regulate metabolism in adipose tissue. Specifically, we review the systemic regulatory roles of Th2 cytokines in metabolic disease and the role of mitochondria in maintenance of type 2 responses in adipose tissue homeostasis.
Collapse
Affiliation(s)
- Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyun Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, Korea
| | - Minho Shong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, Korea.
| |
Collapse
|
117
|
Campderrós L, Moure R, Cairó M, Gavaldà-Navarro A, Quesada-López T, Cereijo R, Giralt M, Villarroya J, Villarroya F. Brown Adipocytes Secrete GDF15 in Response to Thermogenic Activation. Obesity (Silver Spring) 2019; 27:1606-1616. [PMID: 31411815 DOI: 10.1002/oby.22584] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/11/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Transcriptomic analysis of gene expression in brown adipose tissue (BAT) from mice in response to cold revealed strong induction of growth and differentiation factor 15 (GDF15). This study aimed to characterize GDF15 as a brown adipokine released in response to thermogenic activation and to determine its target functions. METHODS GDF15 expression was measured in adipose tissues from mice in response to physiological and pharmacological modulators of thermogenesis. Brown and beige cell cultures were used to dissect the mechanisms regulating GDF15 expression. Brown adipocyte cellular models of fibroblast growth factor 21 and β-klotho invalidation were employed to identify the autocrine regulators of GDF15. RAW 264.7 macrophages were used to explore the targeting of GDF15 released by brown adipocytes. RESULTS Cold exposure of mice strongly induced GDF15 expression in BAT. Norepinephrine and cyclic adenosine monophosphate induced GDF15 expression and release by cells through protein kinase A-mediated mechanisms. Noradrenergic regulation of GDF15 required the active fibroblast growth factor 21 pathway in brown adipocytes. GDF15 released by brown adipocytes targeted macrophages and downregulated the expression of proinflammatory genes. CONCLUSIONS GDF15 is a brown adipokine released by brown and beige cells in response to thermogenic activity. GDF15 released by BAT targets macrophages and may mediate downregulation of local inflammatory pathways.
Collapse
Affiliation(s)
- Laura Campderrós
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Ricardo Moure
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Montserrat Cairó
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Aleix Gavaldà-Navarro
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Tania Quesada-López
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Rubén Cereijo
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Marta Giralt
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| | - Joan Villarroya
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Catalonia, Spain
- Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Madrid, Spain
| |
Collapse
|
118
|
He C, Larson-Casey JL, Davis D, Hanumanthu VS, Longhini ALF, Thannickal VJ, Gu L, Carter AB. NOX4 modulates macrophage phenotype and mitochondrial biogenesis in asbestosis. JCI Insight 2019; 4:126551. [PMID: 31434799 DOI: 10.1172/jci.insight.126551] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 07/16/2019] [Indexed: 12/17/2022] Open
Abstract
Macrophage activation is implicated in the development of pulmonary fibrosis by generation of profibrotic molecules. Although NADPH oxidase 4 (NOX4) is known to contribute to pulmonary fibrosis, its effects on macrophage activation and mitochondrial redox signaling are unclear. Here, we show that NOX4 is crucial for lung macrophage profibrotic polarization and fibrotic repair after asbestos exposure. NOX4 was elevated in lung macrophages from subjects with asbestosis, and mice harboring a deletion of NOX4 in lung macrophages were protected from asbestos-induced fibrosis. NOX4 promoted lung macrophage profibrotic polarization and increased production of profibrotic molecules that induce collagen deposition. Mechanistically, NOX4 further augmented mitochondrial ROS production and induced mitochondrial biogenesis. Targeting redox signaling and mitochondrial biogenesis prevented the profibrotic polarization of lung macrophages by reducing the production of profibrotic molecules. These observations provide evidence that macrophage NOX4 is a potentially novel therapeutic target to halt the development of asbestos-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Chao He
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, and
| | | | - Dana Davis
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, and
| | - Vidya Sagar Hanumanthu
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ana Leda F Longhini
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor J Thannickal
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, and.,Birmingham Veterans Administration Medical Center, Birmingham, Alabama, USA
| | - Linlin Gu
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, and
| | - A Brent Carter
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, and.,Birmingham Veterans Administration Medical Center, Birmingham, Alabama, USA
| |
Collapse
|
119
|
Hassanpour Golakani M, Mohammad MG, Li H, Gamble J, Breit SN, Ruitenberg MJ, Brown DA. MIC-1/GDF15 Overexpression Is Associated with Increased Functional Recovery in Traumatic Spinal Cord Injury. J Neurotrauma 2019; 36:3410-3421. [PMID: 31232176 DOI: 10.1089/neu.2019.6421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) has devastating consequences, with limited therapeutic options; therefore, improving its functional outcome is a major goal. The outcome of SCI is contributed to by neuroinflammation, which may be a target for improved recovery and quality of life after injury. Macrophage inhibitory cytokine-1/growth differentiation factor 15 (MIC-1/GDF15) has been identified as a potential novel therapy for central nervous system (CNS) injury because it is an immune regulatory cytokine with neurotrophic properties. Here we used MIC-1/GDF15 knockout (KO) and overexpressing/transgenic (Tg) and wild type (WT) animals to explore its putative therapeutic benefits in a mouse model of contusive SCI. MIC-1/GDF15 Tg mice had superior locomotor recovery and reduced secondary tissue loss at 28 days compared with their KO and WT counterparts. Overexpression of MIC-1/GDF15 coincided with increased expression of monocyte chemoattractant protein-1 (MCP-1)/C-C Motif Chemokine Ligand 2 (CCL2) at the lesion site (28 days post-SCI) and enhanced recruitment of inflammatory cells to the injured spinal cord. This inflammatory cellular infiltrate included an increased frequency of macrophages and dendritic cells (DCs) that mostly preceded recruitment of cluster of differentiation (CD)4+ and CD8+ T cells. Collectively, our findings suggest hat MIC-1/GDF15 is associated with beneficial changes in the clinical course of SCI that are characterized by altered post-injury inflammation and improved functional outcome. Further investigation of MIC-1/GDF15 as a novel therapeutic target for traumatic SCI appears warranted.
Collapse
Affiliation(s)
- Masoud Hassanpour Golakani
- St. Vincent's Centre for Applied Medical Research (AMR), St Vincent's Hospital and University of New South Wales (UNSW), Sydney, New South Wales, Australia.,The Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Mohammad G Mohammad
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates. Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Hui Li
- St. Vincent's Centre for Applied Medical Research (AMR), St Vincent's Hospital and University of New South Wales (UNSW), Sydney, New South Wales, Australia.,The Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Joanne Gamble
- The Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Samuel N Breit
- St. Vincent's Centre for Applied Medical Research (AMR), St Vincent's Hospital and University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - David A Brown
- St. Vincent's Centre for Applied Medical Research (AMR), St Vincent's Hospital and University of New South Wales (UNSW), Sydney, New South Wales, Australia.,The Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia.,Department of Immunopathology, Institute for Clinical Pathology and Medical Research-New South Wales Health Pathology, Westmead Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
120
|
Weinstock A, Brown EJ, Garabedian ML, Pena S, Sharma M, Lafaille J, Moore KJ, Fisher EA. Single-Cell RNA Sequencing of Visceral Adipose Tissue Leukocytes Reveals that Caloric Restriction Following Obesity Promotes the Accumulation of a Distinct Macrophage Population with Features of Phagocytic Cells. IMMUNOMETABOLISM 2019; 1:e190008. [PMID: 31396408 PMCID: PMC6687332 DOI: 10.20900/immunometab20190008] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity can lead to type 2 diabetes and is an epidemic. A major contributor to its adverse effects is inflammation of the visceral adipose tissue (VAT). Life-long caloric restriction (CR), in contrast, results in extended lifespan, enhanced glucose tolerance/insulin sensitivity, and other favorable phenotypes. The effects of CR following obesity are incompletely established, but studies show multiple benefits. Many leukocyte types, macrophages predominantly, reside in VAT in homeostatic and pathological states. CR following obesity transiently increases VAT macrophage content prior to resolution of inflammation and obesity, suggesting that macrophage content and phenotype play critical roles. Here, we examined the heterogeneity of VAT leukocytes and the effects of obesity and CR. In general, our single-cell RNA-sequencing data demonstrate that macrophages are the most abundant and diverse subpopulation of leukocytes in VAT. Obesity induced significant transcriptional changes in all 15 leukocyte subpopulations, with many genes showing coordinated changes in expression across the leukocyte subpopulations. Additionally, obese VAT displayed expansion of one major macrophage subpopulation, which, in silico, was enriched in lipid binding and metabolic processes. This subpopulation returned from dominance in obesity to lean proportions after only 2 weeks of CR, although the pattern of gene expression overall remained similar. Surprisingly, CR VAT is dominated by a different macrophage subpopulation, which is absent in lean conditions. This subpopulation is enriched in genes related to phagocytosis and we postulate that its function includes clearance of dead cells, as well as excess lipids, contributing to limiting VAT inflammation and restoration of the homeostatic state.
Collapse
Affiliation(s)
- Ada Weinstock
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Emily J Brown
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Michela L Garabedian
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Stephanie Pena
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Monika Sharma
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Juan Lafaille
- Department of Microbiology and Immunology, NYU School of Medicine, New York, NY 10016, USA
| | - Kathryn J Moore
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, NY 10016, USA
- Department of Microbiology and Immunology, NYU School of Medicine, New York, NY 10016, USA
| |
Collapse
|
121
|
Hirose M, Künstner A, Schilf P, Tietjen AK, Jöhren O, Huebbe P, Rimbach G, Rupp J, Schwaninger M, Busch H, Ibrahim SM. A Natural mtDNA Polymorphism in Complex III Is a Modifier of Healthspan in Mice. Int J Mol Sci 2019; 20:E2359. [PMID: 31085998 PMCID: PMC6539666 DOI: 10.3390/ijms20092359] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
In this study, we provide experimental evidence that a maternally inherited polymorphism in the mitochondrial cytochrome b gene (mt-Cytb; m.15124A>G, Ile-Val) in mitochondrial complex III resulted in middle-aged obesity and higher susceptibility to diet-induced obesity, as well as age-related inflammatory disease, e.g., ulcerative dermatitis, in mice. As a consequence of the gene variation, we observed alterations in body composition, metabolism and mitochondrial functions, i.e., increased mitochondrial oxygen consumption rate and higher levels of reactive oxygen species, as well as in the commensal bacterial composition in the gut, with higher abundance of Proteobacteria in mice carrying the variant. These observations are in line with the previously described links of the mitochondrial complex III gene with obesity and metabolic diseases in humans. Given that these functional changes by the G variant at m.15124 in the mt-Cytb are already present in young mice that were kept under normal condition, it is plausible that the m.15124A>G variant is a disease susceptibility modifier to the diseases induced by additional stressors, i.e., dietary and/or aging stress, and that the variant results in the higher incidence of clinical diseases presentation in C57BL/6J-mt129S1/SvlmJ than C57BL/6J mice. Thus, mtDNA variants could be potential biomarkers to evaluate the healthspan.
Collapse
Affiliation(s)
- Misa Hirose
- Luebeck Institute of Experimental Dermatology, University of Luebeck, 23562 Luebeck, Germany.
| | - Axel Künstner
- Luebeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Luebeck, 23562 Luebeck, Germany.
| | - Paul Schilf
- Luebeck Institute of Experimental Dermatology, University of Luebeck, 23562 Luebeck, Germany.
| | - Anna Katharina Tietjen
- Luebeck Institute of Experimental Dermatology, University of Luebeck, 23562 Luebeck, Germany.
| | - Olaf Jöhren
- Center of Brain, Behavior & Metabolism, University of Luebeck, 23562 Luebeck, Germany.
| | - Patricia Huebbe
- Institute of Human Nutrition and Food Science, University of Kiel, 24098 Kiel, Germany.
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, 24098 Kiel, Germany.
| | - Jan Rupp
- Department of Infectious Disease and Microbiology, University of Luebeck, 23562 Luebeck, Germany.
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Luebeck, 23562 Luebeck, Germany.
| | - Hauke Busch
- Luebeck Institute of Experimental Dermatology, Institute for Cardiogenetics and Center for research of inflammatory skin disease (CRIS), University of Luebeck, 23562 Luebeck, Germany.
| | - Saleh M Ibrahim
- Luebeck Institute of Experimental Dermatology and Center for research of inflammatory skin disease (CRIS), University of Luebeck, 23562 Luebeck, Germany.
| |
Collapse
|
122
|
Zhang H, Fealy CE, Kirwan JP. Exercise training promotes a GDF15-associated reduction in fat mass in older adults with obesity. Am J Physiol Endocrinol Metab 2019; 316:E829-E836. [PMID: 30860878 PMCID: PMC6580172 DOI: 10.1152/ajpendo.00439.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity is a major risk factor for metabolic disease. Growth differentiation factor 15 (GDF15) has shown promise as a weight loss agent for obesity in animal studies. In healthy lean humans, fasting plasma GDF15 increases after acute exercise. However, the role of GDF15 in human obesity and the response of plasma GDF15 to exercise training in patients with obesity is unknown. Here, 24 sedentary volunteers with obesity [age: 65 ± 1 yr; body mass index (BMI): 35.3 ± 0.9 kg/m2] participated in a supervised 12-wk aerobic exercise intervention: 1 h/day, 5 days/wk at ~85% maximum heart rate with controlled isocaloric diet. As a result, plasma GDF15 was significantly increased (PRE: 644.1 ± 42.6 pg/ml, POST: 704.4 ± 47.2 pg/ml, P < 0.01) after the exercise intervention. Inconsistent with animal models, ΔGDF15 was not correlated with change in weight, BMI, or resting energy expenditure. However, ΔGDF15 was correlated with a reduction in total fat mass (P < 0.05), abdominal fat mass (P < 0.05), and android fat mass (P ≤ 0.05). Participants with a positive GDF15 response to exercise had increased total fat oxidation (PRE: 0.25 ± 0.05 mg·kg-1·min-1, POST: 0.43 ± 0.07 mg·kg-1·min-1, P ≤ 0.05), metabolic flexibility [PRE: -0.01 ± 0.01 delta respiratory quotient (RQ), POST: 0.06 ± 0.01 delta RQ, P < 0.001], and insulin sensitivity (PRE: 0.33 ± 0.01 QUICKI index, POST: 0.34 ± 0.01 QUICKI index, P < 0.01), suggesting a link between GDF15 and fat mass loss as well as exercise-induced metabolic improvement in humans with obesity. We conclude that the exercise-induced increase in plasma GDF15 and the association with reduced fat mass may indicate a role for GDF15 as a therapeutic target for human obesity.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University , Cleveland, Ohio
- Department of Pathobiology, Lerner Research Institution, Cleveland Clinic , Cleveland, Ohio
- Integrated Physiology and Molecular Medicine, Pennington Biomedical Research Center , Baton Rouge, Louisiana
| | - Ciarán E Fealy
- Department of Pathobiology, Lerner Research Institution, Cleveland Clinic , Cleveland, Ohio
| | - John P Kirwan
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University , Cleveland, Ohio
- Department of Pathobiology, Lerner Research Institution, Cleveland Clinic , Cleveland, Ohio
- Integrated Physiology and Molecular Medicine, Pennington Biomedical Research Center , Baton Rouge, Louisiana
| |
Collapse
|
123
|
Cui H, Banerjee S, Guo S, Xie N, Ge J, Jiang D, Zörnig M, Thannickal VJ, Liu G. Long noncoding RNA Malat1 regulates differential activation of macrophages and response to lung injury. JCI Insight 2019; 4:124522. [PMID: 30676324 DOI: 10.1172/jci.insight.124522] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/17/2019] [Indexed: 01/30/2023] Open
Abstract
Macrophage activation, i.e., classical M1 and the alternative M2, plays a critical role in many pathophysiological processes, such as inflammation and tissue injury and repair. Although the regulation of macrophage activation has been under extensive investigation, there is little knowledge about the role of long noncoding RNAs (lncRNAs) in this event. In this study, we found that lncRNA Malat1 expression is distinctly regulated in differentially activated macrophages in that it is upregulated in LPS-treated and downregulated in IL-4-treated cells. Malat1 knockdown attenuates LPS-induced M1 macrophage activation. In contrast, Malat1 knockdown enhanced IL-4-activated M2 differentiation as well as a macrophage profibrotic phenotype. Mechanistically, Malat1 knockdown led to decreased expression of Clec16a, silencing of which phenocopied the regulatory effect of Malat1 on M1 activation. Interestingly, Malat1 knockdown promoted IL-4 induction of mitochondrial pyruvate carriers (MPCs) and their mediation of glucose-derived oxidative phosphorylation (OxPhos), which was crucial to the Malat1 regulation of M2 differentiation and profibrotic phenotype. Furthermore, mice with either global or conditional myeloid knockout of Malat1 demonstrated diminished LPS-induced systemic and pulmonary inflammation and injury. In contrast, these mice developed more severe bleomycin-induced lung fibrosis, accompanied by alveolar macrophages displaying augmented M2 and profibrotic phenotypes. In summary, we have identified what we believe is a previously unrecognized role of Malat1 in the regulation of macrophage polarization. Our data demonstrate that Malat1 is involved in pulmonary pathogeneses in association with aberrant macrophage activation.
Collapse
Affiliation(s)
- Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sami Banerjee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sijia Guo
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Pulmonary, Allergy, and Critical Care Medicine, The Second Affiliated Hospital, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Na Xie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jing Ge
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Geriatrics and Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dingyuan Jiang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Martin Zörnig
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
124
|
Park S, Kang HJ, Jeon JH, Kim MJ, Lee IK. Recent advances in the pathogenesis of microvascular complications in diabetes. Arch Pharm Res 2019; 42:252-262. [PMID: 30771210 DOI: 10.1007/s12272-019-01130-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/31/2019] [Indexed: 12/29/2022]
Abstract
Millions of people worldwide have diabetes, which is diagnosed by fasting blood glucose levels exceeding 126 mg/dL. Regardless of the type of diabetes, prolonged hyperglycemia is damaging to several organs including eyes, kidneys, nerve, and/or heart. The damages are associated with a high risk of morbidity and mortality. Diabetes has been implicated in ischemia in the microvasculature of the target tissues, which occurs due to the insufficient perfusion of tissues. The resulting occlusion and pain affect the quality of life. Multiple therapeutic approaches have been proposed for a long time to overcome these vascular complications. Apart from systemically controlling high glucose levels, other therapeutic strategies are not well understood. In this review, we summarize the recent literature for biochemical/cellular targets that are being utilized for the treatment of diabetic microvascular diseases. These targets, which are closely associated with mitochondrial dysfunction, include the polyol and diacylglycerol-protein kinase C pathways, oxidative stress, non-enzymatic glycation and the formation of advanced glycation end products, and immune dysregulation/inflammation.
Collapse
Affiliation(s)
- Sungmi Park
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea.
| | - Hyeon-Ji Kang
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea
| | - Jae-Han Jeon
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu, South Korea
| | - Min-Ji Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu, South Korea
| | - In-Kyu Lee
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, South Korea.
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, South Korea.
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
125
|
Xu H, Wang X, Wang W. Functional suppression of macrophages derived from THP-1 cells by environmentally-relevant concentrations of arsenite. Comp Biochem Physiol C Toxicol Pharmacol 2018; 214:36-42. [PMID: 30189257 DOI: 10.1016/j.cbpc.2018.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 01/16/2023]
Abstract
Environmental exposure to arsenic is known to induce immunotoxicity. Macrophages are the professional phagocytes that are important in the immune system. In this study, we utilized the macrophages derived from the THP-1 human monocyte cell line as the experimental model to study the functional suppression induced by arsenite (As+3), one of the most prevalent forms of inorganic arsenic, at environmentally-relevant concentrations. Apoptosis was observed in the THP-1 derived macrophages treated with 500 nM As+3 for 18 h. Suppression of phagocytosis was induced by 18 h As+3 treatment starting from 100 nM. Suppressive effects on the production of two pro-inflammatory cytokines, IL-1β and TNF-α, were also found with the treatment of low to moderate doses of As+3 in lipopolysaccharides-stimulated THP-1 derived macrophages. The nitric oxide production was also inhibited by As+3 treatments, which was negatively correlated with the production of superoxide. Collectively, the results from the study demonstrated that environmentally-relevant concentrations of As+3 induced cytotoxicity and suppressed the major cellular functions in THP-1 derived macrophages. The macrophages were showed to be relatively sensitive to As+3, and could be the essential target of the toxicity induced by environmental arsenic exposures.
Collapse
Affiliation(s)
- Huan Xu
- East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai 200237, China; East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai 200237, China.
| | - Xiaolei Wang
- East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai 200237, China
| | - Wei Wang
- East China University of Science and Technology, School of Pharmacy, Department of Pharmaceutical Sciences, Shanghai 200237, China; East China University of Science and Technology, State Key Laboratory of Bioreactor Engineering, Shanghai 200237, China; University of New Mexico, Department of Chemistry and Chemical Biology, Albuquerque, NM 87131, USA.
| |
Collapse
|
126
|
Kim KH, Lee MS. Pathogenesis of Nonalcoholic Steatohepatitis and Hormone-Based Therapeutic Approaches. Front Endocrinol (Lausanne) 2018; 9:485. [PMID: 30197624 PMCID: PMC6117414 DOI: 10.3389/fendo.2018.00485] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an emerging global health problem and a potential risk factor for type 2 diabetes, cardiovascular disease, and chronic kidney disease. Nonalcoholic steatohepatitis (NASH), an advanced form of NAFLD, is a predisposing factor for development of cirrhosis and hepatocellular carcinoma. The increasing prevalence of NASH emphasizes the need for novel therapeutic approaches. Although therapeutic drugs against NASH are not yet available, fundamental insights into the pathogenesis of NASH have been made during the past few decades. Multiple therapeutic strategies have been developed and are currently being explored in clinical trials or preclinical testing. The pathogenesis of NASH involves multiple intracellular/extracellular events in various cell types in the liver or crosstalk events between the liver and other organs. Here, we review current findings and knowledge regarding the pathogenesis of NASH, focusing on the most recent advances. We also highlight hormone-based therapeutic approaches for treatment of NASH.
Collapse
Affiliation(s)
- Kook Hwan Kim
- Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Kook Hwan Kim ;
| | - Myung-Shik Lee
- Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Myung-Shik Lee
| |
Collapse
|