1
|
Zhou C, Qin Z, Zhang H, Xiao H, Zhang H. Proinflammatory cytokines interleukin-18 and interleukin-6 mediate anorexia induction by trichothecene deoxynivalenol and its congeners. Front Vet Sci 2024; 11:1521424. [PMID: 39691381 PMCID: PMC11649634 DOI: 10.3389/fvets.2024.1521424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 12/19/2024] Open
Abstract
As the common foodborne mycotoxins with the highest pollution rate, deoxynivalenol (DON, also named "vomitoxin") can harm the health of humans and animals by causing anorectic response. It has four congeners: 3-acetyldeoxynivalenol (3-ADON), 15-acetyldeoxynivalenol (15-ADON), nivalenol (NIV), and fusarenon X (FX). These five mycotoxins have been associated with the detrimental effect on food intake. However, its underlying mechanism of anorexia remains unclear. The goal of this research was to compare the anorectic responses to these five mycotoxins and relate these effects to proinflammatory cytokines interleukin-18 (IL-18) and interleukin-6 (IL-6) following intraperitoneal (IP) and oral exposure to a common dose at 2.5 mg/kg BW in mice. Plasma IL-18 and IL-6 were elevated within 1-2 h and returned to basal levels at 6 h after exposure to DON, 3-ADON and 15-ADON. FX promoted IL-18 expression at 6 h. Whereas, FX only promoted IL-6 at 6 h. When NIV was injected intraperitoneally, IL-18 started to rise at 1 h and peaked at 6 h. Whereas, NIV only promoted IL-18 at 2 h following oral exposure. IP exposure to NIV induced an increase in IL-6 that occurred only at 2 h. No effect on IL-6 when exposed orally to NIV. In conclusion, the data indicate that IL-18 and IL-6 play critical roles in anorectic response induced by DON and its four congeners 3-ADON, 15-ADON, NIV, FX.
Collapse
Affiliation(s)
- Chuang Zhou
- Jiangsu Vocational College of Agriculture and Forestry, Jurong, China
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Zihui Qin
- School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, China
- Food Laboratory of Zhongyuan, Luohe, China
| | - Huayue Zhang
- School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, China
- Food Laboratory of Zhongyuan, Luohe, China
| | - Huiping Xiao
- School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, China
- Food Laboratory of Zhongyuan, Luohe, China
| | - Hua Zhang
- Jiangsu Vocational College of Agriculture and Forestry, Jurong, China
| |
Collapse
|
2
|
Li L, Ling ZQ. Mechanisms of cancer cachexia and targeted therapeutic strategies. Biochim Biophys Acta Rev Cancer 2024; 1879:189208. [PMID: 39542382 DOI: 10.1016/j.bbcan.2024.189208] [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: 07/27/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
Tumor cachexia is a multifactorial syndrome characterized by systemic dysfunction, including anorexia and severe weight loss that is resistant to standard nutritional interventions. It is estimated that approximately 20 % of cancer patients succumb to cachexia in the later stages of their disease. Thus, understanding its pathogenesis is vital for improving therapeutic outcomes. Recent research has focused on the imbalance between energy intake and expenditure in cachexia. Clinically, cachexia presents with anorexia, adipose tissue atrophy, and skeletal muscle wasting, each driven by distinct mechanisms. Anorexia arises primarily from tumor-secreted factors and cancer-induced hormonal disruptions that impair hypothalamic regulation of appetite. Adipose tissue atrophy is largely attributed to enhanced lipolysis, driven by increased activity of enzymes such as adipose triglyceride lipase and hormone-sensitive lipase, coupled with decreased lipoprotein lipase activity. The browning of white adipose tissue, facilitated by uncoupling protein 1, further accelerates fat breakdown by increasing energy expenditure. Skeletal muscle atrophy, a hallmark of cachexia, results from dysregulated protein turnover via the ubiquitin-proteasome and autophagy-lysosomal pathways, as well as mitochondrial dysfunction. Additionally, chemotherapy can exacerbate cachexia. This review examines the molecular mechanisms underlying cancer cachexia and discusses current therapeutic strategies, aiming to inform future research and improve treatment approaches.
Collapse
Affiliation(s)
- Long Li
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; The Second School of Clinical Medicine, Wenzhou Medical University, No. 109 Xueyuan West Road, Wenzhou 325027, Zhejiang, China
| | - Zhi-Qiang Ling
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
3
|
Yilmaz M, Aksin Ş, Balsak D, Aboalhasan Y, Batmaz İ. Comparison of orexigenic and anorexigenic neuropeptide levels in hyperemesis gravidarum patients with normal pregnant women: A prospective cohort study. Medicine (Baltimore) 2024; 103:e40069. [PMID: 39432622 PMCID: PMC11495732 DOI: 10.1097/md.0000000000040069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 09/25/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND The aim of this study was to determine whether orexigenic neuropeptides, orexin and galanin, and anorexigenic neuropeptides, alpha-melanocyte-stimulating hormone (α-MSH) and cocaine- and amphetamine-regulated transcript (CART), are implicated in hyperemesis gravidarum (HG). METHODS Fifty pregnant women who had been diagnosed with HG between April 2022 and February 2023 at the Siirt University Faculty of Medicine Training and Research Hospital (tertiary center) were recruited for this study. An equal number of pregnant women without an HG diagnosis were included in the study as the control group. Participants' age, pregnancy history, medical history, thyroid function test results, complete blood count results, and electrolyte levels were recorded, and their orexin, galanin, α-MSH, and CART serum levels were analyzed using an enzyme-linked immunosorbent assay. RESULTS No statistically significant differences in orexigenic neuropeptides (orexin and galanin) were observed between the HG and control groups. A statistical difference was found between an anorexigenic neuropeptide (α-MSH) and the control group (P = .012). Based on a receiver operating characteristic analysis, the α-MSH parameter was statistically significant for distinguishing between participants with an HG diagnosis and those without, with a sensitivity of 63.6%, specificity of 65.9%, and cutoff value of 11769.3 pg/mL (P = .012, area under curve: 0.655). Based on the severity classification of ketonuria (ketonuria levels of +1 or +2 were classified as mild, whereas levels of +3 or +4 were classified as moderate to severe), the anorexigenic CART neuropeptide was found to be a statistically significant diagnostic indicator of severe ketonuria (P = .020). CONCLUSION α-MSH and CART levels were found to be related in HG patients and in HG patients with severe ketonuria.
Collapse
Affiliation(s)
- Mehmet Yilmaz
- Siirt University Faculty of Medicine, Obstetrics and Gynecology, Siirt, Turkey
| | - Şerif Aksin
- Siirt University Faculty of Medicine, Obstetrics and Gynecology, Siirt, Turkey
| | - Deniz Balsak
- Siirt University Faculty of Medicine, Obstetrics and Gynecology, Siirt, Turkey
| | - Yasmin Aboalhasan
- Siirt Training and Research Hospital, Obstetrics and Gynecology, Siirt, Turkey
| | - İbrahim Batmaz
- Mardin Artuklu University Faculty of Medicine, Obstetrics and Gynecology, Mardin, Turkey
| |
Collapse
|
4
|
Wensveen FM, Šestan M, Polić B. The immunology of sickness metabolism. Cell Mol Immunol 2024; 21:1051-1065. [PMID: 39107476 PMCID: PMC11364700 DOI: 10.1038/s41423-024-01192-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/29/2024] [Indexed: 09/01/2024] Open
Abstract
Everyone knows that an infection can make you feel sick. Although we perceive infection-induced changes in metabolism as a pathology, they are a part of a carefully regulated process that depends on tissue-specific interactions between the immune system and organs involved in the regulation of systemic homeostasis. Immune-mediated changes in homeostatic parameters lead to altered production and uptake of nutrients in circulation, which modifies the metabolic rate of key organs. This is what we experience as being sick. The purpose of sickness metabolism is to generate a metabolic environment in which the body is optimally able to fight infection while denying vital nutrients for the replication of pathogens. Sickness metabolism depends on tissue-specific immune cells, which mediate responses tailored to the nature and magnitude of the threat. As an infection increases in severity, so do the number and type of immune cells involved and the level to which organs are affected, which dictates the degree to which we feel sick. Interestingly, many alterations associated with metabolic disease appear to overlap with immune-mediated changes observed following infection. Targeting processes involving tissue-specific interactions between activated immune cells and metabolic organs therefore holds great potential for treating both people with severe infection and those with metabolic disease. In this review, we will discuss how the immune system communicates in situ with organs involved in the regulation of homeostasis and how this communication is impacted by infection.
Collapse
Affiliation(s)
| | - Marko Šestan
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
5
|
Stumpff JP, Kim SY, McFadden MI, Nishida A, Shirazi R, Steuerman Y, Gat-Viks I, Forero A, Nair MG, Morrison J. Pleural macrophages translocate to the lung during infection to promote improved influenza outcomes. Proc Natl Acad Sci U S A 2023; 120:e2300474120. [PMID: 38100417 PMCID: PMC10743374 DOI: 10.1073/pnas.2300474120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Seasonal influenza results in 3 to 5 million cases of severe disease and 250,000 to 500,000 deaths annually. Macrophages have been implicated in both the resolution and progression of the disease, but the drivers of these outcomes are poorly understood. We probed mouse lung transcriptomic datasets using the Digital Cell Quantifier algorithm to predict immune cell subsets that correlated with mild or severe influenza A virus (IAV) infection outcomes. We identified a unique lung macrophage population that transcriptionally resembled small serosal cavity macrophages and whose presence correlated with mild disease. Until now, the study of serosal macrophage translocation in the context of viral infections has been neglected. Here, we show that pleural macrophages (PMs) migrate from the pleural cavity to the lung after infection with IAV. We found that the depletion of PMs increased morbidity and pulmonary inflammation. There were increased proinflammatory cytokines in the pleural cavity and an influx of neutrophils within the lung. Our results show that PMs are recruited to the lung during IAV infection and contribute to recovery from influenza. This study expands our knowledge of PM plasticity and identifies a source of lung macrophages independent of monocyte recruitment and local proliferation.
Collapse
Affiliation(s)
- James P. Stumpff
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA92521
| | - Sang Yong Kim
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA92521
| | - Matthew I. McFadden
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH43210
- Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, WA98109
| | - Roksana Shirazi
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA92521
| | - Yael Steuerman
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
| | - Irit Gat-Viks
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
| | - Adriana Forero
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH43210
- Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| | - Meera G. Nair
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA92521
| | - Juliet Morrison
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA92521
| |
Collapse
|
6
|
Queen NJ, Huang W, Zou X, Mo X, Cao L. AAV-BDNF gene therapy ameliorates a hypothalamic neuroinflammatory signature in the Magel2-null model of Prader-Willi syndrome. Mol Ther Methods Clin Dev 2023; 31:101108. [PMID: 37766791 PMCID: PMC10520877 DOI: 10.1016/j.omtm.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
Individuals with Prader-Willi syndrome (PWS) exhibit several metabolic and behavioral abnormalities associated with excessive food-seeking activity. PWS is thought to be driven in part by dysfunctional hypothalamic circuitry and blunted responses to peripheral signals of satiety. Previous work described a hypothalamic transcriptomic signature of individuals with PWS. Notably, PWS patients exhibited downregulation of genes involved in neuronal development and an upregulation of neuroinflammatory genes. Deficiencies of brain-derived neurotrophic factor (BDNF) and its receptor were identified as potential drivers of PWS phenotypes. Our group recently applied an adeno-associated viral (AAV)-BDNF gene therapy within a preclinical PWS model, Magel2-null mice, to improve metabolic and behavioral function. While this proof-of-concept project was promising, it remained unclear how AAV-BDNF was influencing the hypothalamic microenvironment and how its therapeutic effect was mediated. To investigate, we hypothalamically injected AAV-BDNF to wild type and Magel2-null mice and performed mRNA sequencing on hypothalamic tissue. Here, we report that (1) Magel2 deficiency is associated with neuroinflammation in the hypothalamus and (2) AAV-BDNF gene therapy reverses this neuroinflammation. These data newly reveal Magel2-null mice as a valid model of PWS-related neuroinflammation and furthermore suggest that AAV-BDNF may modulate obesity-related neuroinflammatory phenotypes through direct or indirect means.
Collapse
Affiliation(s)
- Nicholas J. Queen
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Wei Huang
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Xunchang Zou
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Xiaokui Mo
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
7
|
He Y, Zheng J, Ye B, Dai Y, Nie K. Chemotherapy-induced gastrointestinal toxicity: Pathogenesis and current management. Biochem Pharmacol 2023; 216:115787. [PMID: 37666434 DOI: 10.1016/j.bcp.2023.115787] [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: 07/24/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Chemotherapy is the most common treatment for malignant tumors. However, chemotherapy-induced gastrointestinal toxicity (CIGT) has been a major concern for cancer patients, which reduces their quality of life and leads to treatment intolerance and even cessation. Nevertheless, prevention and treatment for CIGT are challenging, due to the prevalence and complexity of the condition. Chemotherapeutic drugs directly damage gastrointestinal mucosa to induce CIGT, including nausea, vomiting, anorexia, gastrointestinal mucositis, and diarrhea, etc. The pathogenesis of CIGT involves multiple factors, such as gut microbiota disorders, inflammatory responses and abnormal neurotransmitter levels, that synergistically contribute to its occurrence and development. In particular, the dysbiosis of gut microbiota is usually linked to abnormal immune responses that increases inflammatory cytokines' expression, which is a common characteristic of many types of CIGT. Chemotherapy-induced intestinal neurotoxicity is also a vital concern in CIGT. Currently, modern medicine is the dominant treatment of CIGT, however, traditional Chinese medicine (TCM) has attracted interest as a complementary and alternative therapy that can greatly alleviate CIGT. Accordingly, this review aimed to comprehensively summarize the pathogenesis and current management of CIGT using PubMed and Google Scholar databases, and proposed that future research for CIGT should focus on the gut microbiota, intestinal neurotoxicity, and promising TCM therapies, which may help to develop more effective interventions and optimize managements of CIGT.
Collapse
Affiliation(s)
- Yunjing He
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jingrui Zheng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Binbin Ye
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yongzhao Dai
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ke Nie
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
8
|
Liu K, Zhen W, Bai D, Tan H, He X, Li Y, Liu Y, Zhang Y, Ito K, Zhang B, Ma Y. Lipopolysaccharide-induced immune stress negatively regulates broiler chicken growth via the COX-2-PGE 2-EP4 signaling pathway. Front Immunol 2023; 14:1193798. [PMID: 37207231 PMCID: PMC10189118 DOI: 10.3389/fimmu.2023.1193798] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 04/24/2023] [Indexed: 05/21/2023] Open
Abstract
Aims Immune stress in broiler chickens is characterized by the development of persistent pro-inflammatory responses that contribute to degradation of production performance. However, the underlying mechanisms that cause growth inhibition of broilers with immune stress are not well defined. Methods A total of 252 1-day-old Arbor Acres(AA) broilers were randomly allocated to three groups with six replicates per group and 14 broilers per replicate. The three groups comprised a saline control group, an Lipopolysaccharide (LPS) (immune stress) group, and an LPS and celecoxib group corresponding to an immune stress group treated with a selective COX-2 inhibitor. Birds in LPS group and saline group were intraperitoneally injected with the same amount of LPS or saline from 14d of age for 3 consecutive days. And birds in the LPS and celecoxib group were given a single intraperitoneal injection of celecoxib 15 min prior to LPS injection at 14 d of age. Results The feed intake and body weight gain of broilers were suppressed in response to immune stress induced by LPS which is an intrinsic component of the outer membrane of Gram-negative bacteria. Cyclooxygenase-2 (COX-2), a key enzyme that mediates prostaglandin synthesis, was up-regulated through MAPK-NF-κB pathways in activated microglia cells in broilers exposed to LPS. Subsequently, the binding of prostaglandin E2 (PGE2) to the EP4 receptor maintained the activation of microglia and promoted the secretion of cytokines interleukin-1β and interleukin-8, and chemokines CX3CL1 and CCL4. In addition, the expression of appetite suppressor proopiomelanocortin protein was increased and the levels of growth hormone-releasing hormone were reduced in the hypothalamus. These effects resulted in decreased expression of insulin-like growth factor in the serum of stressed broilers. In contrast, inhibition of COX-2 normalized pro-inflammatory cytokine levels and promoted the expression of Neuropeptide Y and growth hormone-releasing hormone in the hypothalamus which improved the growth performance of stressed broilers. Transcriptomic analysis of the hypothalamus of stressed broilers showed that inhibition of COX-2 activity significantly down-regulated the expression of the TLR1B, IRF7, LY96, MAP3K8, CX3CL1, and CCL4 genes in the MAPK-NF-κB signaling pathway. Conclusion This study provides new evidence that immune stress mediates growth suppression in broilers by activating the COX-2-PGE2-EP4 signaling axis. Moreover, growth inhibition is reversed by inhibiting the activity of COX-2 under stressed conditions. These observations suggest new approaches for promoting the health of broiler chickens reared in intensive conditions.
Collapse
Affiliation(s)
- Kexin Liu
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Wenrui Zhen
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Dongying Bai
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Haiqiu Tan
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Xianglong He
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Yuqian Li
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Yanhao Liu
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Yi Zhang
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Koichi Ito
- Department of Food and Physiological Models, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ibaraki, Japan
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yanbo Ma
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Longmen Laboratory, Science & Technology Innovation Center for Completed Set Equipment, Luoyang, China
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- *Correspondence: Yanbo Ma,
| |
Collapse
|
9
|
Liu Y, Guo YF, Peng H, Zhou HY, Su T, Yang M, Guo Q, Ye X, Huang Y, Jiang TJ. Hypothalamic Hnscr regulates glucose balance by mediating central inflammation and insulin signal. Cell Prolif 2023; 56:e13332. [PMID: 36042571 DOI: 10.1111/cpr.13332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES Hypothalamic dysfunction leads to glucose metabolic imbalance; however, the mechanisms still need clarification. Our current study was to explore the role of hypothalamic Hnscr in glucose metabolism. MATERIALS AND METHODS Using Hnscr knockout or htNSC-specific Hnscr overexpression mice, we evaluated the effects of Hnscr on glucose metabolism through GTTs, ITTs, serum indicator measurements, etc. Immunofluorescence staining and Western blotting were performed to test inflammation levels and insulin signalling in hypothalamus. Conditioned medium intervene were used to investigate the effects of htNSCs on neuronal cell line. We also detected the glucose metabolism of mice with htNSCs implantation. RESULTS Hnscr expression decreased in the hypothalamus after high-fat diet feed. Hnscr-null mice displayed aggravated systematic insulin resistance, while mice with htNSC-specific Hnscr overexpression had the opposite phenotype. Notably, Hnscr-null mice had increased NF-κB signal in htNSCs, along with enhanced inflammation and damaged insulin signal in neurons located in arcuate nucleus of hypothalamus. The secretions, including sEVs, of Hnscr-deficient htNSCs mediated the detrimental effects on the CNS cell line. Locally implantation with Hnscr-depleted htNSCs disrupted glucose homeostasis. CONCLUSIONS This study demonstrated that decreased Hnscr in htNSCs led to systematic glucose imbalance through activating NF-κB signal and dampening insulin signal in hypothalamic neurons.
Collapse
Affiliation(s)
- Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yi-Fan Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hai-Yan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiao Ye
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Tie-Jian Jiang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
10
|
Wieting J, Jahn K, Buchholz V, Lichtinghagen R, Deest-Gaubatz S, Bleich S, Eberlein CK, Deest M, Frieling H. Alteration of serum leptin and LEP/LEPR promoter methylation in Prader-Willi syndrome. Psychoneuroendocrinology 2022; 143:105857. [PMID: 35803048 DOI: 10.1016/j.psyneuen.2022.105857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/29/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022]
Abstract
Prader-Willi syndrome (PWS) is a rare neurodevelopmental disorder based on a loss of paternally expressed but maternally imprinted genes in chromosome region 15q11-13. PWS individuals typically show insatiable appetite with subsequent obesity representing the major mortality factor unless food intake is inhibited. The neurobiological basis of PWS-typical hyperphagia has remained poorly understood. Many PWS-typical abnormalities are based on hypothalamic dysregulation, a region in which hunger and satiety are hormonally regulated, with the hormone leptin being a main long-term regulator of satiety. Previous studies in PWS have inconsistently shown leptin alterations solely in early childhood, without investigating the leptin system on an epigenetic level. The present study investigates serum leptin levels (S-leptin) and DNA methylation of the leptin (LEP) and leptin receptor gene (LEPR) promoter in 24 individuals with PWS compared to 13 healthy controls matched for sex, age, and body mass index (BMI) and relates the results to the extent of hyperphagia in PWS. S-Leptin levels were obtained by Enzyme-linked Immunosorbent Assay. LEP/LEPR-promoter DNA methylation was assessed by bisulfite-sequencing, hyperphagia by Hyperphagia Questionnaire for Clinical Trials (HQ-CT). PWS and control groups differed significantly in S-leptin levels with higher S-leptin in PWS. Methylation analysis showed significant differences in mean promoter methylation rate both for LEP and LEPR with a lower methylation rate in PWS. LEPR, but not LEP methylation correlated significantly with S-leptin levels. S-leptin and both LEP and LEPR methylation did not correlate with HQ-CT scores in PWS. The present study is the first to show significantly elevated S-leptin levels in an adult PWS cohort combined with an altered, downregulated LEP and LEPR promoter methylation status compared to sex-, age- and BMI-matched controls. Analogous to previous studies, no link to the behavioral dimension could be drawn. Overall, the results suggest an increased leptin dysregulation in PWS, whereby the findings partly mirror those seen in non-syndromic obesity.
Collapse
Affiliation(s)
- Jelte Wieting
- Hannover Medical School, Department for Psychiatry, Social Psychiatry and Psychotherapy, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Kirsten Jahn
- Hannover Medical School, Department for Psychiatry, Social Psychiatry and Psychotherapy, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Vanessa Buchholz
- Hannover Medical School, Department for Psychiatry, Social Psychiatry and Psychotherapy, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Ralf Lichtinghagen
- Hannover Medical School, Department for Clinical Chemistry, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Stephanie Deest-Gaubatz
- Hannover Medical School, Department for Psychiatry, Social Psychiatry and Psychotherapy, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Stefan Bleich
- Hannover Medical School, Department for Psychiatry, Social Psychiatry and Psychotherapy, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Christian K Eberlein
- Hannover Medical School, Department for Psychiatry, Social Psychiatry and Psychotherapy, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Maximilian Deest
- Hannover Medical School, Department for Psychiatry, Social Psychiatry and Psychotherapy, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Helge Frieling
- Hannover Medical School, Department for Psychiatry, Social Psychiatry and Psychotherapy, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
11
|
Li Z, Zhang J, Duan X, Zhao G, Zhang M. Celastrol: A Promising Agent Fighting against Cardiovascular Diseases. Antioxidants (Basel) 2022; 11:antiox11081597. [PMID: 36009315 PMCID: PMC9405053 DOI: 10.3390/antiox11081597] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular diseases (CVD) are leading causes of morbidity and mortality worldwide; therefore, seeking effective therapeutics to reduce the global burden of CVD has become increasingly urgent. Celastrol, a bioactive compound isolated from the roots of the plant Tripterygium wilfordii (TW), has been attracting increasing research attention in recent years, as it exerts cardiovascular treatment benefits targeting both CVD and their associated risk factors. Substantial evidence has revealed a protective role of celastrol against a broad spectrum of CVD including obesity, diabetes, atherosclerosis, cerebrovascular injury, calcific aortic valve disease and heart failure through complicated and interlinked mechanisms such as direct protection against cardiomyocyte hypertrophy and death, and indirect action on oxidation and inflammation. This review will mainly summarize the beneficial effects of celastrol against CVD, largely based on in vitro and in vivo preclinical studies, and the potential underlying mechanisms. We will also briefly discuss celastrol’s pharmacokinetic limitations, which hamper its further clinical applications, and prospective future directions.
Collapse
Affiliation(s)
- Zhexi Li
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, China
| | - Jingyi Zhang
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Research Excellence, London SE5 9NU, UK
| | - Xulei Duan
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, China
| | - Guoan Zhao
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, China
| | - Min Zhang
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, China
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Research Excellence, London SE5 9NU, UK
- Correspondence: ; Tel.: +44-207848-5319; Fax: +44-207848-5193
| |
Collapse
|
12
|
Nampoothiri S, Nogueiras R, Schwaninger M, Prevot V. Glial cells as integrators of peripheral and central signals in the regulation of energy homeostasis. Nat Metab 2022; 4:813-825. [PMID: 35879459 PMCID: PMC7613794 DOI: 10.1038/s42255-022-00610-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/15/2022] [Indexed: 01/03/2023]
Abstract
Communication between the periphery and the brain is key for maintaining energy homeostasis. To do so, peripheral signals from the circulation reach the brain via the circumventricular organs (CVOs), which are characterized by fenestrated vessels lacking the protective blood-brain barrier (BBB). Glial cells, by virtue of their plasticity and their ideal location at the interface of blood vessels and neurons, participate in the integration and transmission of peripheral information to neuronal networks in the brain for the neuroendocrine control of whole-body metabolism. Metabolic diseases, such as obesity and type 2 diabetes, can disrupt the brain-to-periphery communication mediated by glial cells, highlighting the relevance of these cell types in the pathophysiology of such complications. An improved understanding of how glial cells integrate and respond to metabolic and humoral signals has become a priority for the discovery of promising therapeutic strategies to treat metabolic disorders. This Review highlights the role of glial cells in the exchange of metabolic signals between the periphery and the brain that are relevant for the regulation of whole-body energy homeostasis.
Collapse
Affiliation(s)
- Sreekala Nampoothiri
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, Lille, France
| | - Ruben Nogueiras
- Universidade de Santiago de Compostela-Instituto de Investigation Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutrition, Santiago de Compostela, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, Lille, France.
| |
Collapse
|
13
|
Huisman C, Norgard MA, Levasseur PR, Krasnow SM, van der Wijst MGP, Olson B, Marks DL. Critical changes in hypothalamic gene networks in response to pancreatic cancer as found by single-cell RNA sequencing. Mol Metab 2022; 58:101441. [PMID: 35031523 PMCID: PMC8851272 DOI: 10.1016/j.molmet.2022.101441] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Cancer cachexia is a devastating chronic condition characterized by involuntary weight loss, muscle wasting, abnormal fat metabolism, anorexia, and fatigue. However, the molecular mechanisms underlying this syndrome remain poorly understood. In particular, the hypothalamus may play a central role in cachexia, given that it has direct access to peripheral signals because of its anatomical location and attenuated blood-brain barrier. Furthermore, this region has a critical role in regulating appetite and metabolism. METHODS To provide a detailed analysis of the hypothalamic response to cachexia, we performed single-cell RNA-seq combined with RNA-seq of the medial basal hypothalamus (MBH) in a mouse model for pancreatic cancer. RESULTS We found many cell type-specific changes, such as inflamed endothelial cells, stressed oligodendrocyes and both inflammatory and moderating microglia. Lcn2, a newly discovered hunger suppressing hormone, was the highest induced gene. Interestingly, cerebral treatment with LCN2 not only induced many of the observed molecular changes in cachexia but also affected gene expression in food-intake decreasing POMC neurons. In addition, we found that many of the cachexia-induced molecular changes found in the hypothalamus mimic those at the primary tumor site. CONCLUSION Our data reveal that multiple cell types in the MBH are affected by tumor-derived factors or host factors that are induced by tumor growth, leading to a marked change in the microenvironment of neurons critical for behavioral, metabolic, and neuroendocrine outputs dysregulated during cachexia. The mechanistic insights provided in this study explain many of the clinical features of cachexia and will be useful for future therapeutic development.
Collapse
Affiliation(s)
- Christian Huisman
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States; Knight Cancer Institute, Oregon Health & Science University, Portland, United States.
| | - Mason A Norgard
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States
| | - Peter R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States
| | - Stephanie M Krasnow
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States
| | - Monique G P van der Wijst
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Brennan Olson
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States; Medical Scientist Training Program, Oregon Health & Science University, Portland, United States
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States; Knight Cancer Institute, Oregon Health & Science University, Portland, United States; Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, United States.
| |
Collapse
|
14
|
Wang XJ, Li D, Jiao HC, Zhao JP, Lin H. Lipopolysaccharide inhibits hypothalamic Agouti-related protein gene expression via activating mechanistic target of rapamycin signaling in chicks. Gen Comp Endocrinol 2021; 313:113876. [PMID: 34371009 DOI: 10.1016/j.ygcen.2021.113876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/25/2021] [Accepted: 08/02/2021] [Indexed: 11/16/2022]
Abstract
Lipopolysaccharide (LPS) induces profound anorexia in birds. However, the neuronal regulatory network underlying LPS-provoked anorexia is unclear. To determine whether any cross talk occurs among hypothalamic mechanistic target of rapamycin (mTOR) and LPS in the regulation of appetite, we performed an intracerebroventricular injection of rapamycin (an mTOR inhibitor) on LPS-treated chicks. The results indicate that peripheral administrations of LPS decreased the agouti-related protein (AgRP) mRNA level, but increased the phosphorylated mTOR and nuclear factor-кB (NF-кB) protein level. Blocking mTOR significantly attenuated LPS-induced anorexia, AgRP suppression, and p-NF-кB increase. Thus, the results suggest that LPS causes anorexia via the mTOR-AgRP signaling pathway, and mTOR signaling is also associated with the regulation of LPS in p-NF-кB.
Collapse
Affiliation(s)
- X J Wang
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong 271018, China
| | - D Li
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong 271018, China
| | - H C Jiao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong 271018, China
| | - J P Zhao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong 271018, China
| | - H Lin
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong 271018, China.
| |
Collapse
|
15
|
Gong T, Hashimoto AC, Sasuclark AR, Khadka VS, Gurary A, Pitts MW. Selenoprotein M Promotes Hypothalamic Leptin Signaling and Thioredoxin Antioxidant Activity. Antioxid Redox Signal 2021; 35:775-787. [PMID: 30648404 PMCID: PMC8617589 DOI: 10.1089/ars.2018.7594] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Aims: Selenoproteins are an essential class of proteins involved in redox signaling and energy metabolism. However, the functions of many selenoproteins are not clearly established. Selenoprotein M (SELENOM), an endoplasmic reticulum (ER)-resident oxidoreductase bearing structural similarity to thioredoxin (TXN), is among those yet to be fully characterized. This protein is highly expressed in hypothalamic regions involved in leptin signaling and has been previously linked to energy metabolism. Herein, we performed a series of studies using in vivo and in vitro models to probe the specific influence of SELENOM on hypothalamic leptin signaling and assess SELENOM-regulated pathways. Innovation and Results: Our initial experiment in vivo demonstrated that (i) leptin promotes hypothalamic expression of SELENOM and (ii) leptin-induced STAT3 phosphorylation is impeded by SELENOM deficiency. Additional in vitro studies using mHypoE-44 immortalized hypothalamic neurons corroborated these findings, as SELENOM deficiency obstructed downstream STAT3 phosphorylation and cytosolic calcium responses evoked by leptin treatment. Correspondingly, SELENOM overexpression enhanced leptin sensitivity. Microarray analysis conducted in parallel on hypothalamic tissue and mHypoE-44 cells revealed multiple genes significantly affected by SELENOM deficiency, including thioredoxin interacting protein, a negative regulator of the TXN system. Further analysis determined that (i) SELENOM itself possesses intrinsic TXN activity and (ii) SELENOM deficiency leads to a reduction in overall TXN activity. Finally, mHypoE-44 cells lacking SELENOM displayed diminished activation of the nuclear factor kappa-light-chain enhancer of activated B-cells (NF-κB) signaling pathway and increased susceptibility to ER stress-mediated cell death. Conclusion: In sum, these findings establish SELENOM as a positive regulator of leptin signaling and TXN antioxidant activity in the hypothalamus. Antioxid. Redox Signal. 35, 775-787.
Collapse
Affiliation(s)
- Ting Gong
- Department of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, Hawaii, USA
| | - Ann C Hashimoto
- Department of Cell and Molecular Biology, Medical Microbiolgy, and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Alexandru R Sasuclark
- Department of Cell and Molecular Biology, Medical Microbiolgy, and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Vedbar S Khadka
- Bioinformatics Core in the Department of Complementary and Integrative Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Alexandra Gurary
- Department of Tropical Medicine, Medical Microbiolgy, and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Matthew W Pitts
- Department of Cell and Molecular Biology, Medical Microbiolgy, and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
16
|
Galmiche M, Achamrah N, Déchelotte P, Ribet D, Breton J. Role of microbiota-gut-brain axis dysfunctions induced by infections in the onset of anorexia nervosa. Nutr Rev 2021; 80:381-391. [PMID: 34010427 DOI: 10.1093/nutrit/nuab030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Anorexia nervosa (AN) is an eating disorder characterized by low food intake, severe body weight loss, intense fear of gaining weight, and dysmorphophobia. This chronic disease is associated with both psychiatric and somatic comorbidities. Over the years, clinical studies have accumulated evidence that viral or bacterial infections may promote the onset of eating disorders such as AN. This review aims to describe how infections and the subsequent immune responses affect food intake regulation in the short term and also how these processes may lead to long-term intestinal disorders, including gut barrier disruption and gut microbiota dysbiosis, even after the clearance of the pathogens. We discuss in particular how infection-mediated intestinal dysbiosis may promote the onset of several AN symptoms and comorbidities, including appetite dysregulation, functional gastrointestinal disorders, and mood disorders.
Collapse
Affiliation(s)
- Marie Galmiche
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| | - Najate Achamrah
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| | - Pierre Déchelotte
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| | - David Ribet
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| | - Jonathan Breton
- M. Galmiche, N. Achamrah, P. Déchelotte, and J. Breton are with Nutrition Department, CHU Rouen, F-76000 Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, INSERM UMR 1073, Nutrition, Gut and Brain Laboratory, Rouen, France. N. Achamrah, P. Déchelotte, D. Ribet, and J. Breton are with the UNIROUEN, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| |
Collapse
|
17
|
Enhanced lipid utilization is coupled to the sickness responses triggered by lipopolysaccharide. Biochem Biophys Res Commun 2021; 558:44-50. [PMID: 33895550 DOI: 10.1016/j.bbrc.2021.04.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
Sickness symptoms exerted via inflammatory responses occur in several infectious and chronic diseases. A growing body of evidence suggests that altered nutrient availability and metabolism are tightly coupled to inflammatory processes. However, the relationship between metabolic shifts and the development of the sickness response has not been explored fully. Therefore, we aimed to evaluate metabolic phenotypes with a mouse model showing sickness symptoms via systemic administration of lipopolysaccharide (LPS) in the present study. LPS injection elevated the lipid utilization and circulating levels of fatty acids. It also increased the levels of β-hydroxybutyric acid, a ketone body produced from fatty acids. We confirmed the functional connectivity between nutrient utilization and inflammatory responses and demonstrated enhanced lipid utilization in the hypothalamus providing insights into hypothalamic control of sickness responses. Collectively, these findings could help develop new therapeutic strategies to treat patients with severe sickness symptoms associated with infectious and chronic human diseases.
Collapse
|
18
|
Deshpande D, Agarwal N, Fleming T, Gaveriaux-Ruff C, Klose CSN, Tappe-Theodor A, Kuner R, Nawroth P. Loss of POMC-mediated antinociception contributes to painful diabetic neuropathy. Nat Commun 2021; 12:426. [PMID: 33462216 PMCID: PMC7814083 DOI: 10.1038/s41467-020-20677-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Painful neuropathy is a frequent complication in diabetes. Proopiomelanocortin (POMC) is an endogenous opioid precursor peptide, which plays a protective role against pain. Here, we report dysfunctional POMC-mediated antinociception in sensory neurons in diabetes. In streptozotocin-induced diabetic mice the Pomc promoter is repressed due to increased binding of NF-kB p50 subunit, leading to a loss in basal POMC level in peripheral nerves. Decreased POMC levels are also observed in peripheral nervous system tissue from diabetic patients. The antinociceptive pathway mediated by POMC is further impaired due to lysosomal degradation of μ-opioid receptor (MOR). Importantly, the neuropathic phenotype of the diabetic mice is rescued upon viral overexpression of POMC and MOR in the sensory ganglia. This study identifies an antinociceptive mechanism in the sensory ganglia that paves a way for a potential therapy for diabetic neuropathic pain.
Collapse
Affiliation(s)
- Divija Deshpande
- grid.5253.10000 0001 0328 4908Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410 Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Institute of Pharmacology, Heidelberg University, INF 366, Heidelberg, 69120 Germany ,grid.6363.00000 0001 2218 4662Department of Microbiology, Infectious Diseases and Immunology, Charité -Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Nitin Agarwal
- grid.7700.00000 0001 2190 4373Institute of Pharmacology, Heidelberg University, INF 366, Heidelberg, 69120 Germany
| | - Thomas Fleming
- grid.5253.10000 0001 0328 4908Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410 Heidelberg, Germany ,grid.452622.5German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Claire Gaveriaux-Ruff
- grid.420255.40000 0004 0638 2716Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, Illkirch, France ,grid.420255.40000 0004 0638 2716Université de Strasbourg, Illkirch, France ,grid.4444.00000 0001 2112 9282Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U1258 Illkirch, France ,grid.418692.00000 0004 0610 0264Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Christoph S. N. Klose
- grid.6363.00000 0001 2218 4662Department of Microbiology, Infectious Diseases and Immunology, Charité -Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Anke Tappe-Theodor
- grid.7700.00000 0001 2190 4373Institute of Pharmacology, Heidelberg University, INF 366, Heidelberg, 69120 Germany
| | - Rohini Kuner
- grid.7700.00000 0001 2190 4373Institute of Pharmacology, Heidelberg University, INF 366, Heidelberg, 69120 Germany
| | - Peter Nawroth
- grid.5253.10000 0001 0328 4908Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410 Heidelberg, Germany ,grid.452622.5German Center for Diabetes Research (DZD), Neuherberg, Germany ,Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Zentrum, 85764 Neuherberg, Germany
| |
Collapse
|
19
|
Li Y, Jiang Q, Wang L. Appetite Regulation of TLR4-Induced Inflammatory Signaling. Front Endocrinol (Lausanne) 2021; 12:777997. [PMID: 34899611 PMCID: PMC8664591 DOI: 10.3389/fendo.2021.777997] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/04/2021] [Indexed: 12/20/2022] Open
Abstract
Appetite is the basis for obtaining food and maintaining normal metabolism. Toll-like receptor 4 (TLR4) is an important receptor expressed in the brain that induces inflammatory signaling after activation. Inflammation is considered to affect the homeostatic and non-homeostatic systems of appetite, which are dominated by hypothalamic and mesolimbic dopamine signaling. Although the pathological features of many types of inflammation are known, their physiological functions in appetite are largely unknown. This review mainly addresses several key issues, including the structures of the homeostatic and non-homeostatic systems. In addition, the mechanism by which TLR4-induced inflammatory signaling contributes to these two systems to regulate appetite is also discussed. This review will provide potential opportunities to develop new therapeutic interventions that control appetite under inflammatory conditions.
Collapse
Affiliation(s)
- Yongxiang Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, China
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, China
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- *Correspondence: Lina Wang, ; Qingyan Jiang,
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, China
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- *Correspondence: Lina Wang, ; Qingyan Jiang,
| |
Collapse
|
20
|
Tominaga M, Ichikawa S, Sakashita F, Tadaishi M, Shimizu M, Kobayashi-Hattori K. Anorexic action of fusarenon-x in the hypothalamus and intestine. Toxicon 2020; 187:57-64. [PMID: 32882257 DOI: 10.1016/j.toxicon.2020.08.028] [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: 05/23/2020] [Revised: 08/05/2020] [Accepted: 08/30/2020] [Indexed: 10/23/2022]
Abstract
There is a lack of information available on the anorexic action of fusarenon-x (FX), which is a sesquiterpenoid mycotoxin. In this study, we investigated the changes in the hypothalamus and small intestine related to appetite after oral FX exposure. The time-course change of food intake after oral FX exposure (0.5, 1.0, and 2.5 mg/kg bw) in B6C3F1 mice showed that 2.5 mg/kg bw of FX significantly suppressed food intake during 3-6 h compared to the control. Furthermore, the total food intake for 24 h was lower in the group exposed to FX than in the control. The FX exposure (2.5 mg/kg bw for 3 h) significantly increased mRNA levels of anorexic hormones (pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcription (CART)) without changing the mRNA levels of orexigenic hormones. In addition, FX exposure indicated significantly higher mRNA levels of possible downstream targets of anorexic POMC neurons, such as the melanocortin 4 receptor (MC4R), brain-derived neurotrophic factor (BDNF) and tyrosine kinase receptor B (TrkB), in the hypothalamus compared to the control. FX exposure also significantly increased the mRNA level of inflammatory cytokines (tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β)) and activated nuclear factor-kappa B (NF-κB), which is a regulatory factor for POMC in the hypothalamus. In the intestine, FX exposure did not affect the mRNA level of anorexic peptide YY but significantly elevated that of anorexic cholecystokinin (CCK) and regulatory factors for CCK (calcium-sensing receptor (CaSR), the transient receptor potential ankyrin-1 channel (TRPA1), and transient receptor potential cation channel subfamily M member 5 (TRPM5)). These results suggest that FX sequentially induces inflammatory cytokine expression, NF-κB activation, and POMC expression in the hypothalamus. FX also induces CCK expression in the intestine possibly via induction of CaSR, TRPM5, and TRPA1 expression. These changes will eventually lead to the anorexic action of FX.
Collapse
Affiliation(s)
- Misa Tominaga
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Saori Ichikawa
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Fumiko Sakashita
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Miki Tadaishi
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Makoto Shimizu
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Kazuo Kobayashi-Hattori
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan.
| |
Collapse
|
21
|
Böttcher M, Müller-Fielitz H, Sundaram SM, Gallet S, Neve V, Shionoya K, Zager A, Quan N, Liu X, Schmidt-Ullrich R, Haenold R, Wenzel J, Blomqvist A, Engblom D, Prevot V, Schwaninger M. NF-κB signaling in tanycytes mediates inflammation-induced anorexia. Mol Metab 2020; 39:101022. [PMID: 32446877 PMCID: PMC7292913 DOI: 10.1016/j.molmet.2020.101022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Infections, cancer, and systemic inflammation elicit anorexia. Despite the medical significance of this phenomenon, the question of how peripheral inflammatory mediators affect the central regulation of food intake is incompletely understood. Therefore, we have investigated the sickness behavior induced by the prototypical inflammatory mediator IL-1β. METHODS IL-1β was injected intravenously. To interfere with IL-1β signaling, we deleted the essential modulator of NF-κB signaling (Nemo) in astrocytes and tanycytes. RESULTS Systemic IL-1β increased the activity of the transcription factor NF-κB in tanycytes of the mediobasal hypothalamus (MBH). By activating NF-κB signaling, IL-1β induced the expression of cyclooxygenase-2 (Cox-2) and stimulated the release of the anorexigenic prostaglandin E2 (PGE2) from tanycytes. When we deleted Nemo in astrocytes and tanycytes, the IL-1β-induced anorexia was alleviated whereas the fever response and lethargy response were unchanged. Similar results were obtained after the selective deletion of Nemo exclusively in tanycytes. CONCLUSIONS Tanycytes form the brain barrier that mediates the anorexic effect of systemic inflammation in the hypothalamus.
Collapse
Affiliation(s)
- Mareike Böttcher
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany
| | - Helge Müller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Sivaraj M Sundaram
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany
| | - Sarah Gallet
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Lille, France; University of Lille, FHU 1000 days for Health, School of Medicine, U1172, Lille, France
| | - Vanessa Neve
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany
| | - Kiseko Shionoya
- Department of Clinical and Experimental Medicine, Linköping University, S-581 85, Linköping, Sweden
| | - Adriano Zager
- Department of Clinical and Experimental Medicine, Linköping University, S-581 85, Linköping, Sweden
| | - Ning Quan
- Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Xiaoyu Liu
- Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Ruth Schmidt-Ullrich
- Department of Signal Transduction in Tumor Cells, Max-Delbrück-Center (MDC) for Molecular Medicine, 13125, Berlin, Germany
| | - Ronny Haenold
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), 07745, Jena, Germany; Matthias Schleiden Institute of Genetics, Bioinformatics and Molecular Botany, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Jan Wenzel
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Anders Blomqvist
- Department of Clinical and Experimental Medicine, Linköping University, S-581 85, Linköping, Sweden
| | - David Engblom
- Department of Clinical and Experimental Medicine, Linköping University, S-581 85, Linköping, Sweden
| | - Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Lille, France; University of Lille, FHU 1000 days for Health, School of Medicine, U1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23562, Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany.
| |
Collapse
|
22
|
Aviello G, Cristiano C, Luckman SM, D'Agostino G. Brain control of appetite during sickness. Br J Pharmacol 2020; 178:2096-2110. [DOI: 10.1111/bph.15189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Gabriella Aviello
- Department of Pharmacy, School of Medicine and Surgery University of Naples Federico II Naples Italy
| | - Claudia Cristiano
- Department of Pharmacy, School of Medicine and Surgery University of Naples Federico II Naples Italy
| | - Simon M. Luckman
- Faculty of Biology, Medicine and Health, School of Medical Sciences University of Manchester Manchester UK
| | - Giuseppe D'Agostino
- Faculty of Biology, Medicine and Health, School of Medical Sciences University of Manchester Manchester UK
| |
Collapse
|
23
|
Duan K, Gao X, Zhu D. The clinical relevance and mechanism of skeletal muscle wasting. Clin Nutr 2020; 40:27-37. [PMID: 32788088 DOI: 10.1016/j.clnu.2020.07.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/15/2020] [Accepted: 07/21/2020] [Indexed: 02/08/2023]
Abstract
Skeletal muscle wasting occurs in both chronic and acute diseases. Increasing evidence has shown this debilitating process is associated with short- and long-term outcomes in critical, cancer and surgical patients. Both muscle quantity and quality, as reflected by the area and density of a given range of attenuation in CT scan, impact the patient prognosis. In addition, ultrasound and bioelectrical impedance analysis (BIA) are also widely used in the assessment of body composition due to their bedside viability and no radioactivity. Mechanism researches have revealed complicated pathways are involved in muscle wasting, which include altered IGF1-Akt-FoxO signaling, elevated levels of myostatin and activin A, activation of NF-κB pathway and glucocorticoid effects. Particularly, central nervous system (CNS) has been proven to participate in regulating muscle wasting in various conditions, such as infection and tumor. Several promising therapeutic agents have been under developing in the treatment of muscle atrophy, such as myostatin antagonist, ghrelin analog, non-steroidal selective androgen receptor modulators (SARMs). Notably, nutritional therapy is still the fundamental support in combating muscle wasting. However, the optimizing and tailored nutrition regimen relies on accurate metabolism measurement and large clinical trials in the future. Here, we will discuss the current understanding of muscle wasting and potential treatment in clinical practice.
Collapse
Affiliation(s)
- Kaipeng Duan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Xin Gao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Dongming Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China.
| |
Collapse
|
24
|
Peroxisome Proliferator-Activated Receptors and Caloric Restriction-Common Pathways Affecting Metabolism, Health, and Longevity. Cells 2020; 9:cells9071708. [PMID: 32708786 PMCID: PMC7407644 DOI: 10.3390/cells9071708] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR) is a traditional but scientifically verified approach to promoting health and increasing lifespan. CR exerts its effects through multiple molecular pathways that trigger major metabolic adaptations. It influences key nutrient and energy-sensing pathways including mammalian target of rapamycin, Sirtuin 1, AMP-activated protein kinase, and insulin signaling, ultimately resulting in reductions in basic metabolic rate, inflammation, and oxidative stress, as well as increased autophagy and mitochondrial efficiency. CR shares multiple overlapping pathways with peroxisome proliferator-activated receptors (PPARs), particularly in energy metabolism and inflammation. Consequently, several lines of evidence suggest that PPARs might be indispensable for beneficial outcomes related to CR. In this review, we present the available evidence for the interconnection between CR and PPARs, highlighting their shared pathways and analyzing their interaction. We also discuss the possible contributions of PPARs to the effects of CR on whole organism outcomes.
Collapse
|
25
|
Jin S, Kim KK, Park BS, Kim DH, Jeong B, Kang D, Lee TH, Park JW, Kim JG, Lee BJ. Function of astrocyte MyD88 in high-fat-diet-induced hypothalamic inflammation. J Neuroinflammation 2020; 17:195. [PMID: 32560726 PMCID: PMC7304177 DOI: 10.1186/s12974-020-01846-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/19/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND A growing body of evidence shows that hypothalamic inflammation is an important factor in the initiation of obesity. In particular, reactive gliosis accompanied by inflammatory responses in the hypothalamus are pivotal cellular events that elicit metabolic abnormalities. In this study, we examined whether MyD88 signaling in hypothalamic astrocytes controls reactive gliosis and inflammatory responses, thereby contributing to the pathogenesis of obesity. METHODS To analyze the role of astrocyte MyD88 in obesity pathogenesis, we used astrocyte-specific Myd88 knockout (KO) mice fed a high-fat diet (HFD) for 16 weeks or injected with saturated free fatty acids. Astrocyte-specific gene expression in the hypothalamus was determined using real-time PCR with mRNA purified by the Ribo-Tag system. Immunohistochemistry was used to detect the expression of glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, phosphorylated signal transducer and activator of transcription 3, and α-melanocyte-stimulating hormone in the hypothalamus. Animals' energy expenditure was measured using an indirect calorimetry system. RESULTS The astrocyte-specific Myd88 KO mice displayed ameliorated hypothalamic reactive gliosis and inflammation induced by injections of saturated free fatty acids and a long-term HFD. Accordingly, the KO mice were resistant to long-term HFD-induced obesity and showed an improvement in HFD-induced leptin resistance. CONCLUSIONS These results suggest that MyD88 in hypothalamic astrocytes is a critical molecular unit for obesity pathogenesis that acts by mediating HFD signals for reactive gliosis and inflammation.
Collapse
Affiliation(s)
- Sungho Jin
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
- Present address: Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Kwang Kon Kim
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Byong Seo Park
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Dong Hee Kim
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Bora Jeong
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Dasol Kang
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Tae Hwan Lee
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea.
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea.
| |
Collapse
|
26
|
Kang D, Kim HR, Kim KK, Kim DH, Jeong B, Jin S, Park JW, Seong JY, Lee BJ. Brain-specific chemokine FAM19A5 induces hypothalamic inflammation. Biochem Biophys Res Commun 2020; 523:829-834. [PMID: 31954515 DOI: 10.1016/j.bbrc.2019.12.119] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/25/2019] [Indexed: 12/20/2022]
Abstract
The cytokine-like protein FAM19A5 is highly expressed in the brain, but little is known about its functions there. Here, we found that FAM19A5 was expressed in mouse hypothalamic cells expressing proopiomelanocortin (POMC) and neuropeptide Y (NPY)/agouti-related peptide (AgRP), and in the microglia. Tumor necrosis factor-α (TNF-α), which induces inflammatory sickness responses, greatly increased hypothalamic expression of FAM19A5. Knockdown of FAM19A5 expression resulted in decreased TNF-α-induced anorexia, body weight loss and TNF-α-induced expression of inflammatory factors. In contrast, intracerebroventricular administration of FAM19A5 induced anorexia, body weight loss and hyperthermia, together with increased expression of inflammatory factors. FAM19A5 injection also induced increases in c-fos activation and POMC mRNA level in hypothalamic POMC neurons. Together, these results suggest that FAM19A5 plays an important role in hypothalamic inflammatory responses.
Collapse
Affiliation(s)
- Dasol Kang
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Han Rae Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, 44610, South Korea; Department of Pharmacology and Physiology, School of Medicine & Health Sciences, The George Washington University, USA, 20037
| | - Kwang Kon Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Dong Hee Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Bora Jeong
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Sungho Jin
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Jeong Woo Park
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, 44610, South Korea
| | - Jae Young Seong
- Graduate School of Medicine, Korea University, Seoul, 02841, South Korea
| | - Byung Ju Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan, 44610, South Korea.
| |
Collapse
|
27
|
Chowen JA, Garcia-Segura LM. Microglia, neurodegeneration and loss of neuroendocrine control. Prog Neurobiol 2020; 184:101720. [PMID: 31715222 DOI: 10.1016/j.pneurobio.2019.101720] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/19/2019] [Accepted: 11/02/2019] [Indexed: 02/07/2023]
|
28
|
Yamawaki Y, Shirawachi S, Mizokami A, Nozaki K, Ito H, Asano S, Oue K, Aizawa H, Yamawaki S, Hirata M, Kanematsu T. Phospholipase C-related catalytically inactive protein regulates lipopolysaccharide-induced hypothalamic inflammation-mediated anorexia in mice. Neurochem Int 2019; 131:104563. [PMID: 31589911 DOI: 10.1016/j.neuint.2019.104563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 09/09/2019] [Accepted: 10/02/2019] [Indexed: 11/29/2022]
Abstract
Peripheral lipopolysaccharide (LPS) injection induces systemic inflammation through the activation of the inhibitor of nuclear factor kappa B (NF-κB) kinase (IKK)/NF-κB signaling pathway, which promotes brain dysfunction resulting in conditions including anorexia. LPS-mediated reduction of food intake is associated with activation of NF-κB signaling and phosphorylation of the transcription factor signal transducer and activator of transcription 3 (STAT3) in the hypothalamus. We recently reported phospholipase C-related catalytically inactive protein (PRIP) as a new negative regulator of phosphatidylinositol 3-kinase/AKT signaling. AKT regulates the IKK/NF-κB signaling pathway; therefore, this study aimed to investigate the role of PRIP/AKT signaling in LPS-mediated neuroinflammation-induced anorexia. PRIP gene (Prip1 and Prip2) knockout (Prip-KO) mice intraperitoneally (ip) administered with LPS exhibited increased anorexia responses compared with wild-type (WT) controls. Although few differences were observed between WT and Prip-KO mice in LPS-elicited plasma pro-inflammatory cytokine elevation, hypothalamic pro-inflammatory cytokines were significantly upregulated in Prip-KO rather than WT mice. Hypothalamic AKT and IKK phosphorylation and IκB degradation were significantly increased in Prip-KO rather than WT mice, indicating further promotion of AKT-mediated NF-κB signaling. Consistently, hypothalamic STAT3 was further phosphorylated in Prip-KO rather than WT mice. Furthermore, suppressor of cytokine signaling 3 (Socs3), a negative feedback regulator for STAT3 signaling, and cyclooxogenase-2 (Cox2), a candidate molecule in LPS-induced anorexigenic responses, were upregulated in the hypothalamus in Prip-KO rather than WT mice. Pro-inflammatory cytokines were upregulated in hypothalamic microglia isolated from Prip-KO rather than WT mice. Together, these findings indicate that PRIP negatively regulates LPS-induced anorexia caused by pro-inflammatory cytokine expression in the hypothalamus, which is mediated by AKT-activated NF-κB signaling. Importantly, hypothalamic microglia participate in this PRIP-mediated process. Elucidation of PRIP-mediated neuroinflammatory responses may provide novel insights into the pathophysiology of many brain dysfunctions.
Collapse
Affiliation(s)
- Yosuke Yamawaki
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan; Laboratory of Advanced Pharmacology, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka, 815-8511, Japan
| | - Satomi Shirawachi
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Akiko Mizokami
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kanako Nozaki
- Department of Neurobiology, Division of Basic Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hikaru Ito
- Department of Neurobiology, Division of Basic Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan; Center for Experimental Animals, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Kana Oue
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan; Department of Dental Anesthesiology, Division of Applied Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Hidenori Aizawa
- Department of Neurobiology, Division of Basic Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shigeto Yamawaki
- Department of Psychiatry and Neurosciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Masato Hirata
- Oral Medicine Research Center, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| | - Takashi Kanematsu
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan; Department of Cell Biology and Pharmacology, Faculty of Dental Science, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
29
|
Kim HR, Kim DH, Kim KK, Jeong B, Kang D, Lee TH, Park JW, Kwon HM, Lee BJ. Tonicity-responsive enhancer binding protein (TonEBP) regulates TNF-α-induced hypothalamic inflammation. FEBS Lett 2019; 593:2762-2770. [PMID: 31281956 DOI: 10.1002/1873-3468.13533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/19/2019] [Accepted: 06/30/2019] [Indexed: 01/20/2023]
Abstract
Tonicity-responsive enhancer binding protein (TonEBP) is a widely expressed transcription factor and is important in the regulation of inflammatory cytokines. Here, we have identified TonEBP expression in the hypothalamus, which is particularly high in proopiomelanocortin (POMC) neurons. TonEBP overexpression stimulates POMC transcription, and TonEBP haploinsufficiency in TonEBP (+/-) mice results in a decrease in hypothalamic POMC expression. TonEBP (+/-) mice show reduced sickness responses, which include anorexia and hyperthermia, that are initially induced by tumor necrosis factor (TNF)-α. TonEBP (+/-) mice also show lower levels of TNF-α-induced hypothalamic expression of POMC and pro-inflammatory cytokines. These results suggest that TonEBP is an important molecular regulator in the development of inflammatory sickness responses through the control of POMC and pro-inflammatory cytokine expression in the hypothalamus.
Collapse
Affiliation(s)
- Han Rae Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, South Korea
| | - Dong Hee Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, South Korea
| | - Kwang Kon Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, South Korea
| | - Bora Jeong
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, South Korea
| | - Dasol Kang
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, South Korea
| | - Tae Hwan Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, South Korea
| | - Jeong Woo Park
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, South Korea
| | - Hyug Moo Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, South Korea
| | - Byung Ju Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, South Korea
| |
Collapse
|
30
|
Abstract
Under conditions leading to aging and metabolic syndrome, the hypothalamus atypically undergoes proinflammatory signaling activation leading to a chronic and stable background inflammation, referred to as "hypothalamic microinflammation." Through the past decade of research, progress has been made to causally link this hypothalamic inflammation to the mechanism of aging as well as metabolic syndrome, promoting the "hypothalamic microinflammation" theory, which helps characterize the consensus of these epidemic health problems. In general, it is consistently appreciated that hypothalamic microinflammation emerges during the early stages of aging and metabolic syndrome and evolves to be multifaceted and advanced alongside disease progression, while inhibition of key inflammatory components in the hypothalamus has a broad range of effects in counteracting these disorders. Herein, focusing on aging and metabolic syndrome, this writing aims to provide an overview of and insights into the mediators, signaling components, cellular impacts, and physiological significance of this hypothalamic microinflammation.
Collapse
|
31
|
He M, Huang XF, Gao G, Zhou T, Li W, Hu J, Chen J, Li J, Sun T. Olanzapine-induced endoplasmic reticulum stress and inflammation in the hypothalamus were inhibited by an ER stress inhibitor 4-phenylbutyrate. Psychoneuroendocrinology 2019; 104:286-299. [PMID: 30927713 DOI: 10.1016/j.psyneuen.2019.03.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/01/2019] [Accepted: 03/19/2019] [Indexed: 01/08/2023]
Abstract
Antipsychotics are the most important treatment for schizophrenia. However, antipsychotics, particularly olanzapine and clozapine, are associated with severe weight gain/obesity side-effects. Although numerous studies have been carried out to identify the exact mechanisms of antipsychotic-induced weight gain, it is still important to consider other pathways. Endoplasmic reticulum (ER) stress signaling and its associated inflammation pathway is one of the most important pathways involved in regulation of energy balance. In the present study, we examined the role of hypothalamic protein kinase R like endoplasmic reticulum kinase- eukaryotic initiation factor 2α (PERK-eIF2α) signaling and the inflammatory IkappaB kinase β- nuclear factor kappa B (IKKβ-NFκB) signaling pathway in olanzapine-induced weight gain in female rats. In this study, we found that olanzapine significantly activated PERK-eIF2α and IKKβ-NFκB signaling in SH-SY5Y cells in a dose-dependent manner. Olanzapine treatment for 8 days in rats was associated with activated PERK-eIF2α signaling and IKKβ-NFκB signaling in the hypothalamus, accompanied by increased food intake and weight gain. Co-treatment with an ER stress inhibitor, 4-phenylbutyrate (4-PBA), decreased olanzapine-induced food intake and weight gain in a dose- and time-dependent manner. Moreover, 4-PBA dose-dependently inhibited olanzapine-induced activated PERK-eIF2α and IKKβ-NFκB signaling in the hypothalamus. These results suggested that hypothalamic ER stress may play an important role in antipsychotic-induced weight gain.
Collapse
Affiliation(s)
- Meng He
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and Centre for Translational Neuroscience, School of Medicine, University of Wollongong, NSW, 2522, Australia
| | - Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei, China
| | - Ting Zhou
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Wenting Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jinqi Hu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jia Chen
- Wuhan Seventh Hospital, Wuhan, Hubei, China
| | - Jing Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China; State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei, China.
| |
Collapse
|
32
|
Mravec B, Horvathova L, Cernackova A. Hypothalamic Inflammation at a Crossroad of Somatic Diseases. Cell Mol Neurobiol 2019; 39:11-29. [PMID: 30377908 PMCID: PMC11469881 DOI: 10.1007/s10571-018-0631-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/24/2018] [Indexed: 02/08/2023]
Abstract
Various hypothalamic nuclei function as central parts of regulators that maintain homeostasis of the organism. Recently, findings have shown that inflammation in the hypothalamus may significantly affect activity of these homeostats and consequently participate in the development of various somatic diseases such as obesity, diabetes, hypertension, and cachexia. In addition, hypothalamic inflammation may also affect aging and lifespan. Identification of the causes and mechanisms involved in the development of hypothalamic inflammation creates not only a basis for better understanding of the etiopathogenesis of somatic diseases, but for the development of new therapeutic approaches for their treatment, as well.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 813 72, Bratislava, Slovakia.
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Lubica Horvathova
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Alena Cernackova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 813 72, Bratislava, Slovakia
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
33
|
Jeong B, Kim HR, Choi NS, Park BS, Eom H, Park JW, Kim JG, Lee BJ. Role of thyroid transcription factor-1 in transcriptional regulation of heme oxygenase-1. Biochem Biophys Res Commun 2018; 496:147-152. [PMID: 29305861 DOI: 10.1016/j.bbrc.2018.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 01/03/2018] [Indexed: 02/05/2023]
Abstract
Here, we report thyroid transcription factor 1 (TTF-1) as an important transcription factor for the expression of heme oxygenase-1 (HO-1). HO-1 is a well-known cytoprotective enzyme against inflammation. We observed that HO-1 co-expressed with TTF-1 in mouse hypothalamic cells. Results from luciferase and chromatin immunoprecipitation assays revealed that TTF-1 directly activated HO-1 transcription by binding to binding domains in the 5'-flanking region of the HO-1 gene. A proinflammatory cytokine, tumor necrosis factor-alpha (TNF-α), induced nuclear translocation of TTF-1 and increased binding affinity of TTF-1 to its binding sites on the HO-1 gene. HO-1 mRNA increased with TTF-1 overexpression but decreased with RNA interference of TTF-1 expression in rat astroglial C6 cells. Together with results showing involvement of TTF-1 in the TNF-α-induced increase in interleukin 1 beta and monocyte chemotactic protein 1 production, this study suggests that TTF-1 plays an important role in the mouse hypothalamus TNF-α-induced inflammatory response for regulating HO-1 gene expression.
Collapse
Affiliation(s)
- Bora Jeong
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, South Korea
| | - Han Rae Kim
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, South Korea
| | - Nan Seong Choi
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, South Korea
| | - Byong Seo Park
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
| | - Hyejin Eom
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, South Korea
| | - Jeong Woo Park
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, South Korea
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, South Korea
| | - Byung Ju Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, South Korea.
| |
Collapse
|
34
|
Feng B, Zhang N, Duan K, Shi B. Hypothalamic POMC expression is required for peripheral insulin action on hepatic gluconeogenesis through regulating STAT3 in sepsis rats. J Cell Mol Med 2017; 22:1696-1707. [PMID: 29285858 PMCID: PMC5824389 DOI: 10.1111/jcmm.13449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/06/2017] [Indexed: 12/13/2022] Open
Abstract
Liver injury and dysregulated glucose homoeostasis are common manifestations during sepsis. Although plenty of studies reported insulin could protect against multiple organ injuries caused by critical infections among patients, little was known about the precise mechanism. We investigated whether liver inflammatory pathway and central neuropeptides were involved in the process. In sepsis rats, hepatic IKK/NF‐κB pathway and STAT3 were strongly activated, along with reduced body weight, blood glucose and suppressed hepatic gluconeogenesis (GNG). Peripheral insulin administration efficiently attenuated liver dysfunction and glucose metabolic disorders by suppressing hypothalamic anorexigenic neuropeptide proopiomelanocortin (POMC) expression, hepatic NF‐κB pathway and STAT3 phosphorylation. Furthermore, knockdown of hypothalamic POMC significantly diminished protective effect of insulin on hepatic GNG and insulin‐induced STAT3 inactivation, but not inflammation or IKK/NF‐κB pathway. These results suggest that hepatic IKK/NF‐κB pathway mediates the anti‐inflammatory effect of insulin in septic rats, and peripheral insulin treatment may improve hepatic GNG by inhibiting STAT3 phosphorylation dependent on hypothalamic POMC expression.
Collapse
Affiliation(s)
- Bin Feng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Nannan Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Kaipeng Duan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bimin Shi
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
35
|
Mechanick JI, Zhao S, Garvey WT. Leptin, An Adipokine With Central Importance in the Global Obesity Problem. Glob Heart 2017; 13:113-127. [PMID: 29248361 DOI: 10.1016/j.gheart.2017.10.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/25/2017] [Indexed: 02/08/2023] Open
Abstract
Leptin has central importance in the global obesity and cardiovascular disease problem. Leptin is principally secreted by adipocytes and acts in the hypothalamus to suppress appetite and food intake, increase energy expenditure, and regulate body weight. Based on clinical translation of specific and networked actions, leptin affects the cardiovascular system and may be a marker and driver of cardiometabolic risk factors with interventions that are actionable by cardiologists. Leptin subnetwork analysis demonstrates a statistically significant role for ethnoculturally and socioeconomically appropriate lifestyle intervention in cardiovascular disease. Emergent mechanistic components and potential diagnostic or therapeutic targets include hexokinase 3, urocortins, clusterin, sialic acid-binding immunoglobulin-like lectin 6, C-reactive protein, platelet glycoprotein VI, albumin, pentraxin 3, ghrelin, obestatin prepropeptide, leptin receptor, neuropeptide Y, and corticotropin-releasing factor receptor 1. Emergent associated symptoms include weight change, eating disorders, vascular necrosis, chronic fatigue, and chest pain. Leptin-targeted therapies are reported for lipodystrophy and leptin deficiency, but they are investigational for leptin resistance, obesity, and other chronic diseases.
Collapse
Affiliation(s)
- Jeffrey I Mechanick
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Endocrinology, Diabetes, and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Shan Zhao
- Basepaws Inc., Redondo Beach, CA, USA
| | - W Timothy Garvey
- Department of Nutritional Sciences and Diabetes Research Center, University of Alabama at Birmingham, Birmingham, AL, USA; Geriatric Research Education and Clinical Center, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| |
Collapse
|
36
|
Hypothalamic and inflammatory basis of hypertension. Clin Sci (Lond) 2017; 131:211-223. [PMID: 28057892 DOI: 10.1042/cs20160001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/07/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
Abstract
Hypertension is a major health problem with great consequences for public health. Despite its role as the primary cause of significant morbidity and mortality associated with cardiovascular disease, the pathogenesis of essential hypertension remains largely unknown. The central nervous system (CNS) in general, and the hypothalamus in particular, are intricately involved in the development and maintenance of hypertension. Over the last several decades, the understanding of the brain's role in the development of hypertension has dramatically increased. This brief review is to summarize the neural mechanisms of hypertension with a focus on neuroendocrine and neurotransmitter involvement, highlighting recent findings that suggest that hypothalamic inflammation disrupts key signalling pathways to affect the central control of blood pressure, and therefore suggesting future development of interventional strategies that exploit recent findings pertaining to the hypothalamic control of blood pressure as well as the inflammatory-sympathetic mechanisms involved in hypertension.
Collapse
|
37
|
Subadi I, Nugraha B, Laswati H, Josomuljono H. Pain Relief with Wet Cupping Therapy in Rats is Mediated by Heat Shock Protein 70 and ß-Endorphin. IRANIAN JOURNAL OF MEDICAL SCIENCES 2017; 42:384-391. [PMID: 28761205 PMCID: PMC5523046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Wet cupping therapy is a complementary therapy in pain management. The mechanism of this therapy, however, needs further elucidation. Cells injured by wet cupping therapy seem to stimulate the expression of heat shock protein 70 (HSP70). Its benefit in pain reduction could be mediated by the expression of ß-endorphin. This study aimed at determining the correlation between HSP70 and ß-endorphin after wet cupping therapy. METHODS Sixteen male Wistar rats were divided into control (CG; n=8) and treatment (TG; n=8) groups. The rats in both groups were injected with complete Freund's adjuvant (CFA) at the footpad. In the TG, wet cupping therapy was done at the left and right paralumbar regions 48 hours after the CFA injection. Twenty-four hours after therapy, the hot plate test was done to assess pain threshold. Thereafter, immunohistochemistry from the skin subjected to wet cupping therapy was conducted for HSP70 and ß-endorphin. RESULTS The expression of HSP70 was significantly higher in the keratinocytes of the TG (20.25±3.53; P<0.001) than in the keratinocytes of the CG (10.50±2.44; P<0.001). The expression of ß-endorphin was significantly higher in the keratinocytes of the TG (22.37±3.52; P<0.001) than in the keratinocytes of the CG (5.12±1.72; P<0.001). The results also revealed a high correlation between HSP70 and ß-endorphin (β=0.864; P<0.001). Pain threshold after wet cupping therapy was significantly higher in the TG (22.81±6.34 s; P=0.003) than in the CG (11.78±3.56 s). CONCLUSIONS The benefit of wet cupping therapy in terms of pain reduction in rats could be mediated by the expression of HSP70 and ß-endorphin.
Collapse
Affiliation(s)
- Imam Subadi
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Airlangga University, Surabaya, Indonesia,Correspondence: Imam Subadi, MD, PhD; Faculty of Medicine Airlangga University, Jl. Prof. Dr. Moestopo 47 Surabaya-Indonesia Tel: +62 812 3250655 Fax: +62 31 5038838
| | - Boya Nugraha
- Department of Rehabilitation Medicine, Hannover Medical School, Hannover, Germany
| | - Hening Laswati
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Harjanto Josomuljono
- Departmentof Physiology, Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| |
Collapse
|
38
|
Le Thuc O, Stobbe K, Cansell C, Nahon JL, Blondeau N, Rovère C. Hypothalamic Inflammation and Energy Balance Disruptions: Spotlight on Chemokines. Front Endocrinol (Lausanne) 2017; 8:197. [PMID: 28855891 PMCID: PMC5557773 DOI: 10.3389/fendo.2017.00197] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 07/27/2017] [Indexed: 12/20/2022] Open
Abstract
The hypothalamus is a key brain region in the regulation of energy balance as it controls food intake and both energy storage and expenditure through integration of humoral, neural, and nutrient-related signals and cues. Many years of research have focused on the regulation of energy balance by hypothalamic neurons, but the most recent findings suggest that neurons and glial cells, such as microglia and astrocytes, in the hypothalamus actually orchestrate together several metabolic functions. Because glial cells have been described as mediators of inflammatory processes in the brain, the existence of a causal link between hypothalamic inflammation and the deregulations of feeding behavior, leading to involuntary weight loss or obesity for example, has been suggested. Several inflammatory pathways that could impair the hypothalamic control of energy balance have been studied over the years such as, among others, toll-like receptors and canonical cytokines. Yet, less studied so far, chemokines also represent interesting candidates that could link the aforementioned pathways and the activity of hypothalamic neurons. Indeed, chemokines, in addition to their role in attracting immune cells to the inflamed site, have been suggested to be capable of neuromodulation. Thus, they could disrupt cellular activity together with synthesis and/or secretion of multiple neurotransmitters/mediators involved in the maintenance of energy balance. This review discusses the different inflammatory pathways that have been identified so far in the hypothalamus in the context of feeding behavior and body weight control impairments, with a particular focus on chemokines signaling that opens a new avenue in the understanding of the major role played by inflammation in obesity.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
- Helmholtz Diabetes Center (HDC), German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Katharina Stobbe
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Céline Cansell
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Jean-Louis Nahon
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Nicolas Blondeau
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Carole Rovère
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
- *Correspondence: Carole Rovère,
| |
Collapse
|
39
|
Wauman J, Zabeau L, Tavernier J. The Leptin Receptor Complex: Heavier Than Expected? Front Endocrinol (Lausanne) 2017; 8:30. [PMID: 28270795 PMCID: PMC5318964 DOI: 10.3389/fendo.2017.00030] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/01/2017] [Indexed: 12/31/2022] Open
Abstract
Under normal physiological conditions, leptin and the leptin receptor (ObR) regulate the body weight by balancing food intake and energy expenditure. However, this adipocyte-derived hormone also directs peripheral processes, including immunity, reproduction, and bone metabolism. Leptin, therefore, can act as a metabolic switch connecting the body's nutritional status to high energy consuming processes. We provide an extensive overview of current structural insights on the leptin-ObR interface and ObR activation, coupling to signaling pathways and their negative regulation, and leptin functioning under normal and pathophysiological conditions (obesity, autoimmunity, cancer, … ). We also discuss possible cross-talk with other receptor systems on the receptor (extracellular) and signaling cascade (intracellular) levels.
Collapse
Affiliation(s)
- Joris Wauman
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
| | - Lennart Zabeau
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
| | - Jan Tavernier
- Cytokine Receptor Laboratory, Faculty of Medicine and Health Sciences, Department of Biochemistry, Ghent University, Ghent, Belgium
- VIB Medical Biotechnology Center, VIB, Ghent, Belgium
- *Correspondence: Jan Tavernier,
| |
Collapse
|
40
|
Hypothalamic activation is essential for endotoxemia-induced acute muscle wasting. Sci Rep 2016; 6:38544. [PMID: 27922103 PMCID: PMC5138608 DOI: 10.1038/srep38544] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/10/2016] [Indexed: 01/04/2023] Open
Abstract
Growing evidence suggests acute skeletal muscle wasting is a key factor affecting nutritional support and prognosis in critical patients. Previously, plenty of studies of muscle wasting focused on the peripheral pathway, little was known about the central role. We tested the hypothesis whether central inflammatory pathway and neuropeptides were involved in the process. In lipopolysaccharide (LPS) treated rats, hypothalamic NF-κB pathway and inflammation were highly activated, which was accompanied with severe muscle wasting. Central inhibition of nuclear factor kappa-B (NF-κB) pathway activation by infusion of an inhibitor (PS1145) can efficiently reduce muscle wasting as well as attenuate hypothalamic neuropeptides alteration. Furthermore, knockdown the expression of anorexigenic neuropeptide proopiomelanocortin (POMC) expression with a lentiviral vector containing shRNA can significantly alleviate LPS-induced muscle wasting, whereas hypothalamic inflammation or NF-κB pathway was barely affected. Taken together, these results suggest activation of hypothalamic POMC is pivotal for acute muscle wasting caused by endotoxemia. Neuropeptide POMC expression may have mediated the contribution of hypothalamic inflammation to peripheral muscle wasting. Pharmaceuticals with the ability of inhibiting hypothalamic NF-κB pathway or POMC activation may have a therapeutic potential for acute muscle wasting and nutritional therapy in septic patients.
Collapse
|
41
|
Zhu X, Levasseur PR, Michaelis KA, Burfeind KG, Marks DL. A distinct brain pathway links viral RNA exposure to sickness behavior. Sci Rep 2016; 6:29885. [PMID: 27435819 PMCID: PMC4951726 DOI: 10.1038/srep29885] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/23/2016] [Indexed: 11/14/2022] Open
Abstract
Sickness behaviors and metabolic responses to invading pathogens are common to nearly all types of infection. These responses evolved to provide short-term benefit to the host to ward off infection, but impact on quality of life, and when prolonged lead to neurodegeneration, depression, and cachexia. Among the major infectious agents, viruses most frequently enter the brain, resulting in profound neuroinflammation. We sought to define the unique features of the inflammatory response in the brain to these infections. We demonstrate that the molecular pathway defining the central response to dsRNA is distinct from that found in the periphery. The behavioral and physical response to the dsRNA mimetic poly I:C is dependent on signaling via MyD88 when it is delivered centrally, whereas this response is mediated via the TRIF pathway when delivered peripherally. We also define the likely cellular candidates for this MyD88-dependent step. These findings suggest that symptom management is possible without ameliorating protective antiviral immune responses.
Collapse
Affiliation(s)
- Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA
| | - Pete R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA
| | - Katherine A Michaelis
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA.,MD/PhD Program, Oregon Health &Science University, Portland, OR 97239, USA
| | - Kevin G Burfeind
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA.,MD/PhD Program, Oregon Health &Science University, Portland, OR 97239, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health &Science University, Portland, OR 97239, USA
| |
Collapse
|
42
|
Jin S, Kim JG, Park JW, Koch M, Horvath TL, Lee BJ. Hypothalamic TLR2 triggers sickness behavior via a microglia-neuronal axis. Sci Rep 2016; 6:29424. [PMID: 27405276 PMCID: PMC4942617 DOI: 10.1038/srep29424] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/17/2016] [Indexed: 12/16/2022] Open
Abstract
Various pathophysiologic mechanisms leading to sickness behaviors have been proposed. For example, an inflammatory process in the hypothalamus has been implicated, but the signaling modalities that involve inflammatory mechanisms and neuronal circuit functions are ill-defined. Here, we show that toll-like receptor 2 (TLR2) activation by intracerebroventricular injection of its ligand, Pam3CSK4, triggered hypothalamic inflammation and activation of arcuate nucleus microglia, resulting in altered input organization and increased activity of proopiomelanocortin (POMC) neurons. These animals developed sickness behavior symptoms, including anorexia, hypoactivity, and hyperthermia. Antagonists of nuclear factor kappa B (NF-κB), cyclooxygenase pathway and melanocortin receptors 3/4 reversed the anorexia and body weight loss induced by TLR2 activation. These results unmask an important role of TLR2 in the development of sickness behaviors via stimulation of hypothalamic microglia to promote POMC neuronal activation in association with hypothalamic inflammation.
Collapse
Affiliation(s)
- Sungho Jin
- Department of Biological Sciences, University of Ulsan, Ulsan, 680-749, Republic of Korea
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 406-772, Republic of Korea.,Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan, 680-749, Republic of Korea
| | - Marco Koch
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Institute of Anatomy, University of Leipzig, 04103 Leipzig, Germany
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan, 680-749, Republic of Korea
| |
Collapse
|
43
|
Yamawaki Y, Oue K, Shirawachi S, Asano S, Harada K, Kanematsu T. Phospholipase C-related catalytically inactive protein can regulate obesity, a state of peripheral inflammation. JAPANESE DENTAL SCIENCE REVIEW 2016; 53:18-24. [PMID: 28408965 PMCID: PMC5390332 DOI: 10.1016/j.jdsr.2016.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 05/23/2016] [Accepted: 06/06/2016] [Indexed: 11/28/2022] Open
Abstract
Obesity is defined as abnormal or excessive fat accumulation. Chronic inflammation in fat influences the development of obesity-related diseases. Many reports state that obesity increases the risk of morbidity in many diseases, including hypertension, dyslipidemia, type 2 diabetes, coronary heart disease, stroke, sleep apnea, and breast, prostate and colon cancers, leading to increased mortality. Obesity is also associated with chronic neuropathologic conditions such as depression and Alzheimer's disease. However, there is strong evidence that weight loss reduces these risks, by limiting blood pressure and improving levels of serum triglycerides, total cholesterol, low-density lipoprotein (LDL)-cholesterol, and high-density lipoprotein (HDL)-cholesterol. Prevention and control of obesity is complex, and requires a multifaceted approach. The elucidation of molecular mechanisms driving fat metabolism (adipogenesis and lipolysis) aims at developing clinical treatments to control obesity. We recently reported a new regulatory mechanism in fat metabolism: a protein phosphatase binding protein, phospholipase C-related catalytically inactive protein (PRIP), regulates lipolysis in white adipocytes and heat production in brown adipocytes via phosphoregulation. Deficiency of PRIP in mice led to reduced fat accumulation and increased energy expenditure, resulting in a lean phenotype. Here, we evaluate PRIP as a new therapeutic target for the control of obesity.
Collapse
Affiliation(s)
- Yosuke Yamawaki
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kana Oue
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan.,Department of Dental Anesthesiology, Division of Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Satomi Shirawachi
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kae Harada
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Takashi Kanematsu
- Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
44
|
O'Flanagan CH, Bowers LW, Hursting SD. A weighty problem: metabolic perturbations and the obesity-cancer link. Horm Mol Biol Clin Investig 2016; 23:47-57. [PMID: 26167982 DOI: 10.1515/hmbci-2015-0022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 06/05/2015] [Indexed: 01/03/2023]
Abstract
Obesity is an established risk factor for several cancers, including breast, colon, endometrial, ovarian, gastric, pancreatic and liver, and is increasingly a public health concern. Obese cancer patients often have poorer prognoses, reduced response to standard treatments, and are more likely to develop metastatic disease than normo-weight individuals. Many of the pathologic features of obesity promote tumor growth, such as metabolic perturbations, hormonal and growth factor imbalances, and chronic inflammation. Although obesity exacerbates tumor development, the interconnected relationship between the two conditions presents opportunities for new treatment approaches, some of which may be more successful in obese cohorts. Here, we discuss the many ways in which excess adiposity can impact cancer development and progression and address potential preventive and therapeutic strategies to reduce the burden of obesity-related cancers.
Collapse
|
45
|
Liu Y, Huang Y, Liu T, Wu H, Cui H, Gautron L. Lipopolysacharide Rapidly and Completely Suppresses AgRP Neuron-Mediated Food Intake in Male Mice. Endocrinology 2016; 157:2380-92. [PMID: 27111742 PMCID: PMC4891783 DOI: 10.1210/en.2015-2081] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although Agouti-related peptide (AgRP) neurons play a key role in the regulation of food intake, their contribution to the anorexia caused by proinflammatory insults has yet to be identified. Using a combination of neuroanatomical and pharmacogenetics experiments, this study sought to investigate the importance of AgRP neurons and downstream targets in the anorexia caused by the peripheral administration of a moderate dose of lipopolysaccharide (LPS) (100 μg/kg, ip). First, in the C57/Bl6 mouse, we demonstrated that LPS induced c-fos in select AgRP-innervated brain sites involved in feeding but not in any arcuate proopiomelanocortin neurons. Double immunohistochemistry further showed that LPS selectively induced c-Fos in a large subset of melanocortin 4 receptor-expressing neurons in the lateral parabrachial nucleus. Secondly, we used pharmacogenetics to stimulate the activity of AgRP neurons during the course of LPS-induced anorexia. In AgRP-Cre mice expressing the designer receptor hM3Dq-Gq only in AgRP neurons, the administration of the designer drug clozapine-N-oxide (CNO) induced robust food intake. Strikingly, CNO-mediated food intake was rapidly and completely blunted by the coadministration of LPS. Neuroanatomical experiments further indicated that LPS did not interfere with the ability of CNO to stimulate c-Fos in AgRP neurons. In summary, our findings combined together support the view that the stimulation of select AgRP-innervated brain sites and target neurons, rather than the inhibition of AgRP neurons themselves, is likely to contribute to the rapid suppression of food intake observed during acute bacterial endotoxemia.
Collapse
Affiliation(s)
- Yang Liu
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Ying Huang
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Tiemin Liu
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Hua Wu
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Huxing Cui
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Laurent Gautron
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| |
Collapse
|
46
|
Dwarkasing JT, Witkamp RF, Boekschoten MV, Ter Laak MC, Heins MS, van Norren K. Increased hypothalamic serotonin turnover in inflammation-induced anorexia. BMC Neurosci 2016; 17:26. [PMID: 27207102 PMCID: PMC4875640 DOI: 10.1186/s12868-016-0260-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 05/11/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Anorexia can occur as a serious complication of disease. Increasing evidence suggests that inflammation plays a major role, along with a hypothalamic dysregulation characterized by locally elevated serotonin levels. The present study was undertaken to further explore the connections between peripheral inflammation, anorexia and hypothalamic serotonin metabolism and signaling pathways. First, we investigated the response of two hypothalamic neuronal cell lines to TNFα, IL-6 and LPS. Next, we studied transcriptomic changes and serotonergic activity in the hypothalamus of mice after intraperitoneal injection with TNFα, IL-6 or a combination of TNFα and IL-6. RESULTS In vitro, we showed that hypothalamic neurons responded to inflammatory mediators by releasing cytokines. This inflammatory response was associated with an increased serotonin release. Mice injected with TNFα and IL-6 showed decreased food intake, associated with altered expression of inflammation-related genes in the hypothalamus. In addition, hypothalamic serotonin turnover showed to be elevated in treated mice. CONCLUSIONS Overall, our results underline that peripheral inflammation reaches the hypothalamus where it affects hypothalamic serotoninergic metabolism. These hypothalamic changes in serotonin pathways are associated with decreased food intake, providing evidence for a role of serotonin in inflammation-induced anorexia.
Collapse
Affiliation(s)
- J T Dwarkasing
- Nutrition and Pharmacology Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703HD, Wageningen, The Netherlands.
| | - R F Witkamp
- Nutrition and Pharmacology Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703HD, Wageningen, The Netherlands
| | - M V Boekschoten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703HD, Wageningen, The Netherlands
| | - M C Ter Laak
- Nutrition and Pharmacology Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703HD, Wageningen, The Netherlands
| | - M S Heins
- Brains On-line, P.O. Box 4030, 9701 EA, Groningen, The Netherlands
| | - K van Norren
- Nutrition and Pharmacology Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703HD, Wageningen, The Netherlands
| |
Collapse
|
47
|
Quarta C, Sánchez-Garrido MA, Tschöp MH, Clemmensen C. Renaissance of leptin for obesity therapy. Diabetologia 2016; 59:920-7. [PMID: 26983921 DOI: 10.1007/s00125-016-3906-7] [Citation(s) in RCA: 25] [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: 12/04/2015] [Accepted: 01/22/2016] [Indexed: 12/12/2022]
Abstract
Diet-induced obesity and its metabolic comorbidities constitute an overwhelming health crisis and there is an urgent need for safe and effective pharmacological interventions. Being largely shelved for decades, scientists are now revisiting the anti-obesity virtues of leptin. Whereas it remains evident that leptin as a stand-alone therapy is not an effective approach, the potential for employing sensitising pharmacology to unleash the weight-lowering properties of leptin has injected new hope into the field. Fascinatingly, these leptin-sensitising agents seem to act via distinct metabolic pathways and may thus, in parallel with their clinical development, serve as important research tools to progress our understanding of the molecular, physiological and behavioural pathways underlying energy homeostasis and obesity pathophysiology. This review summarises a presentation given at the 'Is leptin coming back?' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Thomas Meek and Gregory Morton, DOI: 10.1007/s00125-016-3898-3 , and by Gerald Shulman and colleagues, DOI: 10.1007/s00125-016-3909-4 ) and an overview by the Session Chair, Ulf Smith (DOI: 10.1007/s00125-016-3894-7 ).
Collapse
Affiliation(s)
- Carmelo Quarta
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Munich, Germany
| | - Miguel A Sánchez-Garrido
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Munich, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Munich, Germany.
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Munich, Germany.
| | - Christoffer Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Munich, Germany
| |
Collapse
|
48
|
Tominaga M, Momonaka Y, Yokose C, Tadaishi M, Shimizu M, Yamane T, Oishi Y, Kobayashi-Hattori K. Anorexic action of deoxynivalenol in hypothalamus and intestine. Toxicon 2016; 118:54-60. [PMID: 27090011 DOI: 10.1016/j.toxicon.2016.04.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/14/2016] [Indexed: 12/18/2022]
Abstract
Although deoxynivalenol (DON) suppresses food intake and subsequent weight gain, its contribution to anorexia mechanisms has not been fully clarified. Thus, we investigated the anorexic actions of DON in the hypothalamus and intestine, both organs related to appetite. When female B6C3F1 mice were orally exposed to different doses of DON, a drastic anorexic action was observed at a dose of 12.5 mg/kg body weight (bw) from 0 to 3 h after administration. Exposure to DON (12.5 mg/kg bw) for 3 h significantly increased the hypothalamic mRNA levels of anorexic pro-opiomelanocortin (POMC) and its downstream targets, including melanocortin 4 receptor, brain-derived neurotrophic factor, and tyrosine kinase receptor B; at the same time, orexigenic hormones were not affected. In addition, exposure to DON significantly elevated the hypothalamic mRNA levels of proinflammatory cytokines (IL-1β, TNF-α, and IL-6) and activated nuclear factor-kappa B (NF-κB), an upstream factor of POMC. These results suggest that DON-induced proinflammatory cytokines increased the POMC level via NF-κB activation. Moreover, exposure to DON significantly enhanced the gastrointestinal mRNA levels of anorexic cholecystokinin (CCK) and transient receptor potential ankyrin-1 channel (TRPA1), a possible target of DON; these findings suggest that DON induced anorexic action by increasing CCK production via TRPA1. Taken together, these results suggest that DON induces anorexic POMC, perhaps via NF-κB activation, by increasing proinflammatory cytokines in the hypothalamus and brings about CCK production, possibly through increasing intestinal TRPA1 expression, leading to anorexic actions.
Collapse
Affiliation(s)
- Misa Tominaga
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Yuka Momonaka
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Chihiro Yokose
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Miki Tadaishi
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Makoto Shimizu
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Takumi Yamane
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Yuichi Oishi
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Kazuo Kobayashi-Hattori
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan.
| |
Collapse
|
49
|
Liu X, Cai F, Zhang Y, Yang A, Liu L. Celastrol, an NF-κB inhibitor, ameliorates hypercalciuria and articular cartilage lesions in a mouse model of secondary osteoporosis. J Pharmacol Sci 2016; 130:204-11. [DOI: 10.1016/j.jphs.2016.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/24/2015] [Accepted: 02/01/2016] [Indexed: 01/17/2023] Open
|
50
|
Kwon O, Kim KW, Kim MS. Leptin signalling pathways in hypothalamic neurons. Cell Mol Life Sci 2016; 73:1457-77. [PMID: 26786898 PMCID: PMC11108307 DOI: 10.1007/s00018-016-2133-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/20/2015] [Accepted: 01/07/2016] [Indexed: 12/25/2022]
Abstract
Leptin is the most critical hormone in the homeostatic regulation of energy balance among those so far discovered. Leptin primarily acts on the neurons of the mediobasal part of hypothalamus to regulate food intake, thermogenesis, and the blood glucose level. In the hypothalamic neurons, leptin binding to the long form leptin receptors on the plasma membrane initiates multiple signaling cascades. The signaling pathways known to mediate the actions of leptin include JAK-STAT signaling, PI3K-Akt-FoxO1 signaling, SHP2-ERK signaling, AMPK signaling, and mTOR-S6K signaling. Recent evidence suggests that leptin signaling in hypothalamic neurons is also linked to primary cilia function. On the other hand, signaling molecules/pathways mitigating leptin actions in hypothalamic neurons have been extensively investigated in an effort to treat leptin resistance observed in obesity. These include SOCS3, tyrosine phosphatase PTP1B, and inflammatory signaling pathways such as IKK-NFκB and JNK signaling, and ER stress-mitochondrial signaling. In this review, we discuss leptin signaling pathways in the hypothalamus, with a particular focus on the most recently discovered pathways.
Collapse
Affiliation(s)
- Obin Kwon
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
- Appetite Regulation Laboratory, Asan Institute for Life Science, Seoul, 05505, Korea
| | - Ki Woo Kim
- Department of Pharmacology, Yonsei University Wonju College of Medicine, Wonju, 26426, Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, 26426, Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
- Appetite Regulation Laboratory, Asan Institute for Life Science, Seoul, 05505, Korea.
| |
Collapse
|