1
|
Shi Q, Ran S, Song L, Yang H, Wang W, Liu H, Liu Q. NLRP6 overexpression improves nonalcoholic fatty liver disease by promoting lipid oxidation and decomposition in hepatocytes through the AMPK/CPT1A/PGC1A pathway. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:118-125. [PMID: 39819720 PMCID: PMC11744278 DOI: 10.12122/j.issn.1673-4254.2025.01.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVES To investigate the regulatory role of nucleotide-bound oligomerized domain-like receptor containing pyrin-domain protein 6 (NLRP6) in liver lipid metabolism and non-alcoholic fatty liver disease (NAFLD). METHODS Mouse models with high-fat diet (HFD) feeding for 16 weeks (n=6) or with methionine choline-deficient diet (MCD) feeding for 8 weeks (n=6) were examined for the development of NAFLD using HE and oil red O staining, and hepatic expressions of NLRP6 were detected with RT-qPCR, Western blotting, and immunohistochemical staining. Cultured human hepatocytes (LO2 cells) with adenovirus-mediated NLRP6 overexpression or knock-down were treated with palmitic acid (PA) in the presence or absence of compound C (an AMPK inhibitor), and the changes in cellular lipid metabolism were examined by measuring triglyceride, ATP and β-hydroxybutyrate levels and using oil red staining, RT-qPCR, and Western blotting. RESULTS HFD and MCD feeding both resulted in the development of NAFLD in mice, which showed significantly decreased NLRP6 expression in the liver. In PA-treated LO2 cells, NLRP6 overexpression significantly decreased cellular TG content and lipid deposition, while NLRP6 knockdown caused the opposite effects. NLRP6 overexpression in PA-treated LO2 cells also increased mRNA and protein expressions of PGC1A and CPT1A, levels of ATP and β-hydroxybutyrate, and the phosphorylation level of AMPK pathway; the oxidative decomposition of lipids induced by Ad-NLRP6 was inhibited by the use of AMPK inhibitors. CONCLUSIONS NLRP6 overexpression promotes lipid oxidation and decomposition through AMPK/CPT1A/PGC1A to alleviate lipid deposition in hepatocytes.
Collapse
Affiliation(s)
- Qing Shi
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Suye Ran
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Lingyu Song
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Hong Yang
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Wenjuan Wang
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Hanlin Liu
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Qi Liu
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| |
Collapse
|
2
|
Damiecki M, Naha R, Schaumkessel Y, Westhoff P, Atanelov N, Stefanski A, Petzsch P, Stühler K, Köhrer K, Weber AP, Anand R, Reichert AS, Kondadi AK. Mitochondrial apolipoprotein MIC26 is a metabolic rheostat regulating central cellular fuel pathways. Life Sci Alliance 2024; 7:e202403038. [PMID: 39393820 PMCID: PMC11472510 DOI: 10.26508/lsa.202403038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024] Open
Abstract
Mitochondria play central roles in metabolism and metabolic disorders such as type 2 diabetes. MIC26, a mitochondrial contact site and cristae organising system complex subunit, was linked to diabetes and modulation of lipid metabolism. Yet, the functional role of MIC26 in regulating metabolism under hyperglycemia is not understood. We used a multi-omics approach combined with functional assays using WT and MIC26 KO cells cultured in normoglycemia or hyperglycemia, mimicking altered nutrient availability. We show that MIC26 has an inhibitory role in glycolysis and cholesterol/lipid metabolism under normoglycemic conditions. Under hyperglycemia, this inhibitory role is reversed demonstrating that MIC26 is critical for metabolic adaptations. This is partially mediated by alterations of mitochondrial metabolite transporters. Furthermore, MIC26 deletion led to a major metabolic rewiring of glutamine use and oxidative phosphorylation. We propose that MIC26 acts as a metabolic "rheostat," that modulates mitochondrial metabolite exchange via regulating mitochondrial cristae, allowing cells to cope with nutrient overload.
Collapse
Affiliation(s)
- Melissa Damiecki
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ritam Naha
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Yulia Schaumkessel
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Westhoff
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Plant Metabolism and Metabolomics Laboratory, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University, Düsseldorf, Germany
| | - Nika Atanelov
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Anja Stefanski
- Molecular Proteomics Laboratory, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Patrick Petzsch
- Genomics and Transcriptomics Laboratory, BMFZ, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Molecular Medicine, Protein Research, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karl Köhrer
- Genomics and Transcriptomics Laboratory, BMFZ, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas Pm Weber
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Plant Metabolism and Metabolomics Laboratory, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University, Düsseldorf, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Arun Kumar Kondadi
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
3
|
Bai Y, Tang Q, Zheng L, He J, Wang W, Li L, Yu J. Elevated expression of APOO as a potential prognostic marker in breast cancer: insights from bioinformatic analysis and experimental validation. BMC Med Genomics 2024; 17:271. [PMID: 39558346 PMCID: PMC11572147 DOI: 10.1186/s12920-024-02047-7] [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: 03/18/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
OBJECTIVE Apolipoprotein O (APOO) has been identified through bioinformatic prediction analysis as being highly expressed in various tumors, including breast cancer (BRCA). However, further investigations are required to understand and confirm APOO's biological role in BRCA. METHODS Bioinformatic analyses were employed to identify genes' expression statuses and their relationship with the prognoses of patients. The genes' functions were determined in cell line by gain or loss of function assays. Mechanistic studies were carried out by western blot. RESULTS Our study reveals a correlation between increased APOO expression and poorer clinical outcomes in BRCA patients. The diagnostic value of APOO was demonstrated by Receiver Operating Characteristic (ROC) curve analysis, showing a notable area under the curve (AUC) of 0.937. Additionally, we observed that APOO knockdown impedes cell proliferation and migration. Gene Set Enrichment Analysis (GSEA) suggests that APOO expression is associated with the regulation of apoptosis and autophagy signaling pathways. Experimentally, modifying APOO expression in vitro influenced apoptosis and autophagy in BRCA cells. In conclusion, our findings indicate a significant link between APOO expression and BRCA progression, mediated through APOO's impact on cellular apoptosis and autophagy. CONCLUSIONS Our data show that APOO controls BRCA process through apoptosis and autophagy signal pathway, which might provide multiple promising choices for the treatment of BRCA.
Collapse
Affiliation(s)
- Yang Bai
- Laboratory of Department of Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Qian Tang
- Department of Anesthesiology, Guiqian International General Hospital, Guiyang, 550000, Guizhou, China
| | - Liang Zheng
- Department of Thyroid Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Jun He
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Wenjian Wang
- Laboratory of Department of Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Liqi Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - Ju Yu
- Department of Thyroid Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
4
|
Chen J, Hu J, Guo X, Yang Y, Qin D, Tang X, Huang Z, Wang F, Hu D, Peng D, Yu B. Apolipoprotein O modulates cholesterol metabolism via NRF2/CYB5R3 independent of LDL receptor. Cell Death Dis 2024; 15:389. [PMID: 38830896 PMCID: PMC11148037 DOI: 10.1038/s41419-024-06778-4] [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: 08/03/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/05/2024]
Abstract
Apolipoprotein O (APOO) plays a critical intracellular role in regulating lipid metabolism. Here, we investigated the roles of APOO in metabolism and atherogenesis in mice. Hepatic APOO expression was increased in response to hyperlipidemia but was inhibited after simvastatin treatment. Using a novel APOO global knockout (Apoo-/-) model, it was found that APOO depletion aggravated diet-induced obesity and elevated plasma cholesterol levels. Upon crossing with low-density lipoprotein receptor (LDLR) and apolipoprotein E (APOE) knockout hyperlipidemic mouse models, Apoo-/- Apoe-/- and Apoo-/- Ldlr-/- mice exhibited elevated plasma cholesterol levels, with more severe atherosclerotic lesions than littermate controls. This indicated the effects of APOO on cholesterol metabolism independent of LDLR and APOE. Moreover, APOO deficiency reduced cholesterol excretion through bile and feces while decreasing phospholipid unsaturation by inhibiting NRF2 and CYB5R3. Restoration of CYB5R3 expression in vivo by adeno-associated virus (AAV) injection reversed the reduced degree of phospholipid unsaturation while decreasing blood cholesterol levels. This represents the first in vivo experimental validation of the role of APOO in plasma cholesterol metabolism independent of LDLR and elucidates a previously unrecognized cholesterol metabolism pathway involving NRF2/CYB5R3. APOO may be a metabolic regulator of total-body cholesterol homeostasis and a target for atherosclerosis management. Apolipoprotein O (APOO) regulates plasma cholesterol levels and atherosclerosis through a pathway involving CYB5R3 that regulates biliary and fecal cholesterol excretion, independently of the LDL receptor. In addition, down-regulation of APOO may lead to impaired mitochondrial function, which in turn aggravates diet-induced obesity and fat accumulation.
Collapse
Affiliation(s)
- Jin Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Jiarui Hu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, NO.139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Xin Guo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Yang Yang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Donglu Qin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Xiaoyu Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Zhijie Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Fengjiao Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Die Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Bilian Yu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Research Institute of Blood Lipid and Atherosclerosis, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China.
- Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
- FuRong Laboratory, Changsha, 410078, Hunan, China.
| |
Collapse
|
5
|
Li Y, Yang M, Yuan L, Li T, Zhong X, Guo Y. Associations between a polygenic risk score and the risk of gestational diabetes mellitus in a Chinese population: a case-control study. Endocr J 2023; 70:1159-1168. [PMID: 37779084 DOI: 10.1507/endocrj.ej23-0245] [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] [Indexed: 10/03/2023] Open
Abstract
Our objective was to construct a polygenic risk score (PRS) and assess its utility and effectiveness in predicting the risk of gestational diabetes mellitus (GDM) in a Chinese population. We performed a case-control study involving 638 patients with GDM and 1,062 healthy controls. Genotyping was conducted utilizing a genome-wide association study (GWAS), and a PRS was constructed. We identified 12 susceptibility loci that exhibited significant associations with the risk of GDM at a p-value threshold of ≤5.0 × 10-8, of which four loci were newly discovered. A higher PRS was associated with an increased risk of GDM (OR: 1.44; 95% CI: 1.03, 2.01 for the highest quartile compared to the lowest quartile). The PRS demonstrated a clear linear relationship with the fasting plasma glucose (FPG), 1-hour postprandial glucose (1hPG), and 2-hour postprandial glucose (2hPG) levels. The maximally adjusted β coefficients and their corresponding 95% CIs were 0.181 (0.041, 0.320) for FPG, 0.225 (0.103, 0.346) for 1hPG, and 0.172 (0.036, 0.307) for 2hPG. Among the genetic variants examined, TCF7L2 rs7903146 displayed the strongest association with GDM risk (logOR = 0.18, p = 2.37 × 10-19), followed by ADAMTSL1 rs10963767 (logOR = 0.14, p = 3.58 × 10-15). The areas under the curve (AUCs) was significantly increased from 0.703 (0.678, 0.728) in the traditional risk factor model to 0.765 (0.741, 0.788) by including PRS. These findings indicate that pregnant women with a higher PRS could potentially derive considerable advantages from the implementation of a feasible PRS-based GDM screening program aimed at delivering precision prevention strategies within Chinese populations.
Collapse
Affiliation(s)
- Ying Li
- Department of Graduate School, Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Mengjiao Yang
- Department of Laboratory, The First People's Hospital of Shuangliu District, Chengdu, 610200, Sichuan, China
| | - Lu Yuan
- Department of Endocrinology, The First People's Hospital of Shuangliu District, Chengdu, 610200, Sichuan, China
| | - Ting Li
- Department of Endocrinology, The First People's Hospital of Shuangliu District, Chengdu, 610200, Sichuan, China
| | - Xinli Zhong
- Department of Gynaecology and Obstetrics, The First People's Hospital of Shuangliu District, Chengdu, 610200, Sichuan, China
| | - Yanying Guo
- Department of Endocrinology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, Xinjiang, China
| |
Collapse
|
6
|
Peifer-Weiß L, Kurban M, David C, Lubeck M, Kondadi AK, Nemer G, Reichert AS, Anand R. A X-linked nonsense APOO/MIC26 variant causes a lethal mitochondrial disease with progeria-like phenotypes. Clin Genet 2023; 104:659-668. [PMID: 37649161 DOI: 10.1111/cge.14420] [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: 06/13/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023]
Abstract
APOO/MIC26 is a subunit of the MICOS complex required for mitochondrial cristae morphology and function. Here, we report a novel variant of the APOO/MIC26 gene that causes a severe mitochondrial disease with overall progeria-like phenotypes in two patients. Both patients developed partial agenesis of the corpus callosum, bilateral congenital cataract, hypothyroidism, and severe immune deficiencies. The patients died at an early age of 12 or 18 months. Exome sequencing revealed a mutation (NM_024122.5): c.532G>T (p.E178*) in the APOO/MIC26 gene that causes a nonsense mutation leading to the loss of 20 C-terminal amino acids. This mutation resulted in a highly unstable and degradation prone MIC26 protein, yet the remaining minute amounts of mutant MIC26 correctly localized to mitochondria and interacted physically with other MICOS subunits. MIC26 KO cells expressing MIC26 harboring the respective APOO/MIC26 mutation showed mitochondria with perturbed cristae architecture and fragmented morphology resembling MIC26 KO cells. We conclude that the novel mutation found in the APOO/MIC26 gene is a loss-of-function mutation impairing mitochondrial morphology and cristae morphogenesis.
Collapse
Affiliation(s)
- Leon Peifer-Weiß
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mazen Kurban
- Department Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
- Department of Dermatology, American University of Beirut, Beirut, Lebanon
| | - Céline David
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Melissa Lubeck
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Arun Kumar Kondadi
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Georges Nemer
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
7
|
Tang X, Huang Z, Wang F, Chen J, Qin D, Peng D, Yu B. Macrophage-specific deletion of MIC26 (APOO) mitigates advanced atherosclerosis by increasing efferocytosis. Atherosclerosis 2023; 386:117374. [PMID: 37995600 DOI: 10.1016/j.atherosclerosis.2023.117374] [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: 05/16/2023] [Revised: 10/01/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND AND AIMS Recent studies have suggested that MIC26 (apolipoprotein O, APOO), a novel mitochondrial inner membrane protein, is involved in inflammation. Thus, the role of macrophage MIC26 in acute inflammation and chronic inflammatory disease atherosclerosis was investigated. METHODS Macrophage-specific MIC26 knockout mice (MIC26LysM) were generated by crossing Apooflox/flox and LysMcre+/- mice. An endotoxemia mouse model was generated to explore the effects of macrophage MIC26 deficiency on acute inflammation, while an atherosclerosis mouse model was constructed by crossing MIC26LysM mice with Apoe-/- mice and challenged with a Western diet. Atherosclerotic plaques, primary macrophage function, and mitochondrial structure and function were analyzed. RESULTS MIC26 knockout did not affect the median survival time and post-injection serum interleukin 1β concentrations in mice with endotoxemia. Mice with MIC26 deficiency in an Apoe-/- background had smaller atherosclerotic lesions and necrotic core than the control group. In vitro studies found that the loss of MIC26 did not affect macrophage polarization, apoptosis, or lipid handling capacity, but increased efferocytosis (the ability to clear apoptotic cells). An in situ efferocytosis assay of plaques also showed that the ratio of macrophage-associated apoptotic cells to free apoptotic cells was higher in the MIC26-deficient group than in the control group, indicating increased efferocytosis. In addition, an in vivo thymus efferocytosis assay indicated that MIC26 deletion promoted efferocytosis. Mechanistically, the loss of MIC26 resulted in an abnormal mitochondrial inner membrane structure, increased mitochondrial fission, and decreased mitochondrial membrane potential. Loss of MIC26 reduced mitochondria optic atrophy type 1 (OPA1) protein, and OPA1 silencing in macrophages promoted efferocytosis. Overexpression of OPA1 abolished the increase in efferocytosis produced by MIC26 deficiency. CONCLUSIONS Macrophage MIC26 deletion alleviated advanced atherosclerosis and necrotic core expansion by promoting efferocytosis. This mechanism may be related to the increased mitochondrial fission caused by reduced mitochondrial OPA1.
Collapse
Affiliation(s)
- Xiaoyu Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Systemic Autoimmune Diseases in Hunan Province, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Zhijie Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Fengjiao Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Jin Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Donglu Qin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| | - Bilian Yu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China; FuRong Laboratory, Changsha, 410078, Hunan, China.
| |
Collapse
|
8
|
Liu J, Liu J, Qin G, Li J, Fu Z, Li J, Li M, Guo C, Zhao M, Zhang Z, Li F, Zhao X, Wang L, Zhang Y. MDSCs-derived GPR84 induces CD8 + T-cell senescence via p53 activation to suppress the antitumor response. J Immunother Cancer 2023; 11:e007802. [PMID: 38016719 PMCID: PMC10685939 DOI: 10.1136/jitc-2023-007802] [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] [Accepted: 10/17/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUNDS G-protein-coupled receptor 84 (GPR84) marks a subset of myeloid-derived suppressor cells (MDSCs) with stronger immunosuppression in the tumor microenvironment. Yet, how GPR84 endowed the stronger inhibition of MDSCs to CD8+ T cells function is not well established. In this study, we aimed to identify the underlying mechanism behind the immunosuppression of CD8+ T cells by GPR84+ MDSCs. METHODS The role and underlying mechanism that MDSCs or exosomes (Exo) regulates the function of CD8+ T cells were investigated using immunofluorescence, fluorescence activating cell sorter (FACS), quantitative real-time PCR, western blot, ELISA, Confocal, RNA-sequencing (RNA-seq), etc. In vivo efficacy and mechanistic studies were conducted with wild type, GPR84 and p53 knockout C57/BL6 mice. RESULTS Here, we showed that the transfer of GPR84 from MDSCs to CD8+ T cells via the Exo attenuated the antitumor response. This inhibitory effect was also observed in GPR84-overexpressed CD8+ T cells, whereas depleting GPR84 elevated CD8+ T cells proliferation and function in vitro and in vivo. RNA-seq analysis of CD8+ T cells demonstrated the activation of the p53 signaling pathway in CD8+ T cells treated with GPR84+ MDSCs culture medium. While knockout p53 did not induce senescence in CD8+ T cells treated with GPR84+ MDSCs. The per cent of GPR84+ CD8+ T cells work as a negative indicator for patients' prognosis and response to chemotherapy. CONCLUSIONS These data demonstrated that the transfer of GPR84 from MDSCs to CD8+ T cells induces T-cell senescence via the p53 signaling pathway, which could explain the strong immunosuppression of GPR84 endowed to MDSCs.
Collapse
Affiliation(s)
- Jinyan Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiayin Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Guohui Qin
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiahui Li
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ziyi Fu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jieyao Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Miaomiao Li
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Caijuan Guo
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ming Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Li
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuan Zhao
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & and Treatment, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Lubeck M, Derkum NH, Naha R, Strohm R, Driessen MD, Belgardt BF, Roden M, Stühler K, Anand R, Reichert AS, Kondadi AK. MIC26 and MIC27 are bona fide subunits of the MICOS complex in mitochondria and do not exist as glycosylated apolipoproteins. PLoS One 2023; 18:e0286756. [PMID: 37279200 DOI: 10.1371/journal.pone.0286756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/23/2023] [Indexed: 06/08/2023] Open
Abstract
Impairments of mitochondrial functions are linked to human ageing and pathologies such as cancer, cardiomyopathy, neurodegeneration and diabetes. Specifically, aberrations in ultrastructure of mitochondrial inner membrane (IM) and factors regulating them are linked to diabetes. The development of diabetes is connected to the 'Mitochondrial Contact Site and Cristae Organising System' (MICOS) complex which is a large membrane protein complex defining the IM architecture. MIC26 and MIC27 are homologous apolipoproteins of the MICOS complex. MIC26 has been reported as a 22 kDa mitochondrial and a 55 kDa glycosylated and secreted protein. The molecular and functional relationship between these MIC26 isoforms has not been investigated. In order to understand their molecular roles, we depleted MIC26 using siRNA and further generated MIC26 and MIC27 knockouts (KOs) in four different human cell lines. In these KOs, we used four anti-MIC26 antibodies and consistently detected the loss of mitochondrial MIC26 (22 kDa) and MIC27 (30 kDa) but not the loss of intracellular or secreted 55 kDa protein. Thus, the protein assigned earlier as 55 kDa MIC26 is nonspecific. We further excluded the presence of a glycosylated, high-molecular weight MIC27 protein. Next, we probed GFP- and myc-tagged variants of MIC26 with antibodies against GFP and myc respectively. Again, only the mitochondrial versions of these tagged proteins were detected but not the corresponding high-molecular weight MIC26, suggesting that MIC26 is indeed not post-translationally modified. Mutagenesis of predicted glycosylation sites in MIC26 also did not affect the detection of the 55 kDa protein band. Mass spectrometry of a band excised from an SDS gel around 55 kDa could not confirm the presence of any peptides derived from MIC26. Taken together, we conclude that both MIC26 and MIC27 are exclusively localized in mitochondria and that the observed phenotypes reported previously are exclusively due to their mitochondrial function.
Collapse
Affiliation(s)
- Melissa Lubeck
- Medical Faculty and University Hospital Düsseldorf, Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nick H Derkum
- Medical Faculty and University Hospital Düsseldorf, Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ritam Naha
- Medical Faculty and University Hospital Düsseldorf, Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Rebecca Strohm
- Medical Faculty and University Hospital Düsseldorf, Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marc D Driessen
- Medical Faculty and University Hospital, Institute of Molecular Medicine, Protein Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Bengt-Frederik Belgardt
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Neuherberg, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, Neuherberg, Germany
- Medical Faculty and University Hospital Düsseldorf, Department of Endocrinology and Diabetology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes, Heinrich Heine University, Düsseldorf, Germany
| | - Kai Stühler
- Medical Faculty and University Hospital, Institute of Molecular Medicine, Protein Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Molecular Proteomics Laboratory, BMFZ, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ruchika Anand
- Medical Faculty and University Hospital Düsseldorf, Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas S Reichert
- Medical Faculty and University Hospital Düsseldorf, Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Arun Kumar Kondadi
- Medical Faculty and University Hospital Düsseldorf, Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
10
|
Abstract
Apolipoproteins, the protein component of lipoproteins, play an important role in lipid transport, lipoprotein assembly, and receptor recognition. Apolipoproteins are glycosylated and the glycan moieties play an integral role in apolipoprotein function. Changes in apolipoprotein glycosylation correlate with several diseases manifesting in dyslipidemias. Despite their relevance in apolipoprotein function and diseases, the total glycan repertoire of most apolipoproteins remains undefined. This review summarizes the current knowledge and knowledge gaps regarding human apolipoprotein glycan composition, structure, glycosylation site, and functions. Given the relevance of glycosylation to apolipoprotein function, we expect that future studies of apolipoprotein glycosylation will contribute new understanding of disease processes and uncover relevant biomarkers and therapeutic targets. Considering these future efforts, we also provide a brief overview of current mass spectrometry based technologies that can be applied to define detailed glycan structures, site-specific compositions, and the role of emerging approaches for clinical applications in biomarker discovery and personalized medicine.
Collapse
|
11
|
Liu Y, Xiong Z, Zhou W, Chen Y, Huang Q, Wu Y. Role of apolipoprotein O in autophagy via the p38 mitogen-activated protein kinase signaling pathway in myocardial infarction. Clinics (Sao Paulo) 2022; 77:100046. [PMID: 35588578 PMCID: PMC9120058 DOI: 10.1016/j.clinsp.2022.100046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/10/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To explore the role and possible mechanisms of action of apolipoprotein O (APOO) in autophagy in Myocardial Infarction (MI) in vivo and in vitro. METHODS Differential gene expression and single Gene Set Enrichment Analysis (GSEA) were used to evaluate MI-related candidate genes. Animal and cell MI models were established. Sh-APOO, si-APOO, and SB203580 were used to inhibit the expression of APOO or p38MAPK. Western blot and qRT-PCR were used to analyze the expression levels of the target protein or mRNA. Apoptosis was observed using the TUNEL assay. The plasma concentrations of CK-MB and cTn-I in humans and mice were determined. RESULTS In the GSE23294 dataset, APOO mRNA was highly expressed in the left ventricle of mice with MI; GSEA revealed that APOO was positively correlated with p38MAPK, autophagy, and apoptosis. The plasma concentration of APOO in patients with MI was significantly higher than that in healthy subjects. The expression of APOO, Beclin-1, LC3, and Bax in mouse and AC16 cell MI models increased, while the level of Bcl-2 decreased. After silencing the APOO gene, the expression of APOO was downregulated; meanwhile, changes in autophagy, apoptosis and myocardial cell injury were reversed in vivo and in vitro. Furthermore, autophagy was alleviated after AC16 cells were treated with SB203580. CONCLUSIONS The increased APOO expression in mouse and cell MI models may activate autophagy and apoptosis by regulating the p38MAPK signaling pathway, thus aggravating the myocardial injury.
Collapse
Affiliation(s)
- Yue Liu
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Zhiping Xiong
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Wei Zhou
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Yuxin Chen
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Qing Huang
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China
| | - Yanqing Wu
- Nanchang University Second Affiliated Hospital, Cardiovascular Medicine, Nanchang City, Jiangxi Province, PR China.
| |
Collapse
|
12
|
Ali MB, Dreger DL, Buckley RM, Mansoor S, Khan QM, Ostrander EA. Genetic Origins of the Two Canis lupus familiaris (Dog) Freight Dog Populations. J Hered 2022; 113:160-170. [PMID: 35575082 PMCID: PMC9113510 DOI: 10.1093/jhered/esac002] [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: 10/28/2021] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Despite periodic drops in popularity, Arctic sled dogs continue to play a vital role in northern societies, providing both freight transit and recreational race activities. In this study, we selected the Mackenzie River Husky, a freight dog of complex history, and the Chinook, an American Kennel Club recognized freight dog breed whose heritage reportedly overlaps that of the MKRH, for detailed population analysis. We tested each to determine their component breeds and used admixture analysis to ascertain their population structure. We utilized haplotype analysis to identify genomic regions shared between each population and their founding breeds. Our data show that the Alaskan Malamutes and modern Greenland sled dog contributed to both populations, but there are also unexpected contributions from the German Shepherd dog and Collie. We used haplotype analysis to identify genomic regions nearing fixation in population type and identify provocative genes in each region. Finally, in response to recent reports regarding the importance of dietary lipid genes in Arctic dogs, we analyzed 8 such genes in a targeted analysis observing signatures of selection in both populations at the MLXIPL gene loci. These data highlight the genetic routes that breeds of similar function have taken toward their occupation as successful sled dogs.
Collapse
Affiliation(s)
- Muhammad Basil Ali
- National Institute for Biotechnology and Genetic Engineering College (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Dayna L Dreger
- National Human Genome Research Institute, National Institutes of Health, 50 South Drive, Building 51, Room 5351, Bethesda, MD 20892, USA
| | - Reuben M Buckley
- National Human Genome Research Institute, National Institutes of Health, 50 South Drive, Building 51, Room 5351, Bethesda, MD 20892, USA
| | - Shahid Mansoor
- National Institute for Biotechnology and Genetic Engineering College (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Qaiser M Khan
- National Institute for Biotechnology and Genetic Engineering College (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Elaine A Ostrander
- National Human Genome Research Institute, National Institutes of Health, 50 South Drive, Building 51, Room 5351, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Li Q, Liu W, Feng Y, Hou H, Zhang Z, Yu Q, Zhou Y, Luo Q, Luo Y, Ouyang H, Zhang H, Zhu W. Radix Puerariae thomsonii polysaccharide (RPP) improves inflammation and lipid peroxidation in alcohol and high-fat diet mice by regulating gut microbiota. Int J Biol Macromol 2022; 209:858-870. [PMID: 35439478 DOI: 10.1016/j.ijbiomac.2022.04.067] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/12/2022] [Accepted: 04/09/2022] [Indexed: 12/18/2022]
Abstract
Polysaccharides are the important active constituents of Radix Puerariae thomsonii. Numerous studies have shown that polysaccharides can regulate gut microbiota, repair intestinal barrier, and affect the microbiota-intestine-liver axis, thereby showing therapeutic effects on metabolic disorders. In this study, Radix Puerariae thomsonii polysaccharide (RPP) was extracted from Radix Puerariae thomsonii. The average Mw of RPP was determined to be 1.09 × 105 Da and the monosaccharide composition showed it consisted of glucose. The effects and underlying mechanisms of RPP on fatty liver were studied using C57/BL6J mice induced by alcohol and high-fat diet. The results showed that the oral supplementation of RPP could alleviate alcohol and high-fat diet-induced hepatic injury and steatosis. RPP also promoted intestinal barrier integrity and reduced inflammation through NF-κB signaling pathway. RPP could ameliorate the lipid peroxidation by AMPK/NADPH oxidase signaling pathway. Additionally, these improvements might be related to the enrichment of intestinal bacteria Parabacteroides (promote intestinal barrier integrity) and Prevotellaceae UCG 001 (activation of AMPK signaling pathway). These results demonstrated that RPP could improve inflammation and lipid peroxidation in the alcohol and high-fat diet mouse by restoring the intestinal barrier integrity and regulating the gut microbiota. This suggested that RPP was a potential food supplement for the treatment of fatty liver disease.
Collapse
Affiliation(s)
- Qiong Li
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Wenjun Liu
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., No.1899 Meiling Road, Nanchang 330103, PR China
| | - Yulin Feng
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, No. 56 Yangming Road, Nanchang 330006, PR China
| | - Hengwei Hou
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Zhuang Zhang
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Qingqing Yu
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Ying Zhou
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Quan Luo
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China
| | - Yingying Luo
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, No. 56 Yangming Road, Nanchang 330006, PR China
| | - Hui Ouyang
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China; State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, No. 56 Yangming Road, Nanchang 330006, PR China.
| | - Hua Zhang
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China.
| | - Weifeng Zhu
- Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China; Key Laboratory of Modern Preparation of Chinese Medicine of Jiangxi University of Chinese Medicine, No. 818 Yunwan Road, Nanchang 330002, PR China.
| |
Collapse
|
14
|
Relationship Between Myocardial Injury and Expression of PGC-1α and Its Coactivators in Chronic Keshan Disease. Curr Med Sci 2021; 42:85-92. [PMID: 34705216 DOI: 10.1007/s11596-021-2454-7] [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: 02/01/2021] [Accepted: 07/11/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Keshan disease (KD) is a mitochondrial cardiomyopathy. The present study explored the roles of peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1α (PGC-1α), the key regulator of mitochondrial structure and function, and its coactivators in myocardial injury in chronic KD. Furthermore, the usefulness of these molecules in the diagnosis of chronic KD was assessed. METHODS In the present case-control study, 43 patients with chronic KD and 30 healthy individuals living in KD endemic areas were included. The myocardial injury indicators and mRNA expression levels of PGC-1α, nuclear respiratory factor 1 (NRF1), PPARα, and estrogen-related receptor alpha (ERRα) in peripheral blood were examined. RESULTS It was found that the levels of atrial natriuretic peptide, creatine kinase, and lactate dehydrogenase (LDH) were higher in patients with chronic KD, when compared to controls, while the level of bradykinin was lower. Furthermore, the PGC-1α, NRF1 and PPARα mRNA levels were higher in patients with KD. The area under the receiver operating characteristic curve and the optimal diagnostic threshold of LDH was 0.937 and 304.0 U/L, respectively. It is noteworthy that the area under the combined receiver operating characteristic curve was larger, when compared to that for LDH detection alone (Z=2.055, P=0.0399). The area under the curve for the "LDH+PPARα" combination was 0.984, with 96.7% sensitivity and 93.0% specificity. CONCLUSION The combined detection of LDH and the expression of PPARα can be performed to diagnose the chronic KD.
Collapse
|
15
|
Jennings MJ, Hathazi D, Nguyen CDL, Munro B, Münchberg U, Ahrends R, Schenck A, Eidhof I, Freier E, Synofzik M, Horvath R, Roos A. Intracellular Lipid Accumulation and Mitochondrial Dysfunction Accompanies Endoplasmic Reticulum Stress Caused by Loss of the Co-chaperone DNAJC3. Front Cell Dev Biol 2021; 9:710247. [PMID: 34692675 PMCID: PMC8526738 DOI: 10.3389/fcell.2021.710247] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/02/2021] [Indexed: 12/25/2022] Open
Abstract
Recessive mutations in DNAJC3, an endoplasmic reticulum (ER)-resident BiP co-chaperone, have been identified in patients with multisystemic neurodegeneration and diabetes mellitus. To further unravel these pathomechanisms, we employed a non-biased proteomic approach and identified dysregulation of several key cellular pathways, suggesting a pathophysiological interplay of perturbed lipid metabolism, mitochondrial bioenergetics, ER-Golgi function, and amyloid-beta processing. Further functional investigations in fibroblasts of patients with DNAJC3 mutations detected cellular accumulation of lipids and an increased sensitivity to cholesterol stress, which led to activation of the unfolded protein response (UPR), alterations of the ER-Golgi machinery, and a defect of amyloid precursor protein. In line with the results of previous studies, we describe here alterations in mitochondrial morphology and function, as a major contributor to the DNAJC3 pathophysiology. Hence, we propose that the loss of DNAJC3 affects lipid/cholesterol homeostasis, leading to UPR activation, β-amyloid accumulation, and impairment of mitochondrial oxidative phosphorylation.
Collapse
Affiliation(s)
- Matthew J. Jennings
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Denisa Hathazi
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Chi D. L. Nguyen
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Benjamin Munro
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ute Münchberg
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ilse Eidhof
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Erik Freier
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Rita Horvath
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Andreas Roos
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, Children’s Hospital University of Essen, Essen, Germany
| |
Collapse
|
16
|
Anand R, Kondadi AK, Meisterknecht J, Golombek M, Nortmann O, Riedel J, Peifer-Weiß L, Brocke-Ahmadinejad N, Schlütermann D, Stork B, Eichmann TO, Wittig I, Reichert AS. MIC26 and MIC27 cooperate to regulate cardiolipin levels and the landscape of OXPHOS complexes. Life Sci Alliance 2020; 3:e202000711. [PMID: 32788226 PMCID: PMC7425215 DOI: 10.26508/lsa.202000711] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Homologous apolipoproteins of MICOS complex, MIC26 and MIC27, show an antagonistic regulation of their protein levels, making it difficult to deduce their individual functions using a single gene deletion. We obtained single and double knockout (DKO) human cells of MIC26 and MIC27 and found that DKO show more concentric onion-like cristae with loss of CJs than any single deletion indicating overlapping roles in formation of CJs. Using a combination of complexome profiling, STED nanoscopy, and blue-native gel electrophoresis, we found that MIC26 and MIC27 are dispensable for the stability and integration of the remaining MICOS subunits into the complex suggesting that they assemble late into the MICOS complex. MIC26 and MIC27 are cooperatively required for the integrity of respiratory chain (super) complexes (RCs/SC) and the F1Fo-ATP synthase complex and integration of F1 subunits into the monomeric F1Fo-ATP synthase. While cardiolipin was reduced in DKO cells, overexpression of cardiolipin synthase in DKO restores the stability of RCs/SC. Overall, we propose that MIC26 and MIC27 are cooperatively required for global integrity and stability of multimeric OXPHOS complexes by modulating cardiolipin levels.
Collapse
Affiliation(s)
- Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Arun Kumar Kondadi
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Jana Meisterknecht
- Functional Proteomics, Sonderforschungsbereich (SFB) 815 Core Unit, Faculty of Medicine, Goethe-University, Frankfurt am Main, Germany
- Cluster of Excellence "Macromolecular Complexes", Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - Mathias Golombek
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Oliver Nortmann
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Julia Riedel
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Leon Peifer-Weiß
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Nahal Brocke-Ahmadinejad
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - David Schlütermann
- Institute of Molecular Medicine I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Björn Stork
- Institute of Molecular Medicine I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| | - Thomas O Eichmann
- Center for Explorative Lipidomics, BioTechMed-Graz, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Ilka Wittig
- Functional Proteomics, Sonderforschungsbereich (SFB) 815 Core Unit, Faculty of Medicine, Goethe-University, Frankfurt am Main, Germany
- Cluster of Excellence "Macromolecular Complexes", Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
| |
Collapse
|
17
|
Sinding MHS, Gopalakrishnan S, Ramos-Madrigal J, de Manuel M, Pitulko VV, Kuderna L, Feuerborn TR, Frantz LAF, Vieira FG, Niemann J, Samaniego Castruita JA, Carøe C, Andersen-Ranberg EU, Jordan PD, Pavlova EY, Nikolskiy PA, Kasparov AK, Ivanova VV, Willerslev E, Skoglund P, Fredholm M, Wennerberg SE, Heide-Jørgensen MP, Dietz R, Sonne C, Meldgaard M, Dalén L, Larson G, Petersen B, Sicheritz-Pontén T, Bachmann L, Wiig Ø, Marques-Bonet T, Hansen AJ, Gilbert MTP. Arctic-adapted dogs emerged at the Pleistocene-Holocene transition. Science 2020; 368:1495-1499. [PMID: 32587022 PMCID: PMC7116267 DOI: 10.1126/science.aaz8599] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/06/2020] [Indexed: 12/18/2022]
Abstract
Although sled dogs are one of the most specialized groups of dogs, their origin and evolution has received much less attention than many other dog groups. We applied a genomic approach to investigate their spatiotemporal emergence by sequencing the genomes of 10 modern Greenland sled dogs, an ~9500-year-old Siberian dog associated with archaeological evidence for sled technology, and an ~33,000-year-old Siberian wolf. We found noteworthy genetic similarity between the ancient dog and modern sled dogs. We detected gene flow from Pleistocene Siberian wolves, but not modern American wolves, to present-day sled dogs. The results indicate that the major ancestry of modern sled dogs traces back to Siberia, where sled dog-specific haplotypes of genes that potentially relate to Arctic adaptation were established by 9500 years ago.
Collapse
Affiliation(s)
- Mikkel-Holger S Sinding
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark.
- Natural History Museum, University of Oslo, Oslo, Norway
- The Qimmeq Project, University of Greenland, Nuussuaq, Greenland
- Greenland Institute of Natural Resources, Nuuk, Greenland
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | | | | | - Marc de Manuel
- Institute of Evolutionary Biology (UPF-CSIC), Barcelona, Spain
| | - Vladimir V Pitulko
- Institute for the History of Material Culture, Russian Academy of Sciences, St. Petersburg, Russia
| | - Lukas Kuderna
- Institute of Evolutionary Biology (UPF-CSIC), Barcelona, Spain
| | - Tatiana R Feuerborn
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- The Qimmeq Project, University of Greenland, Nuussuaq, Greenland
- Department of Bioinformatics and Genetics, Swedish Museum of Natural History, Stockholm, Sweden
- Department of Archaeology and Classical Studies, Stockholm University, Stockholm, Sweden
| | - Laurent A F Frantz
- The Palaeogenomics and Bio-Archaeology Research Network, Research Laboratory for Archaeology and History of Art, University of Oxford, Oxford, UK
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Filipe G Vieira
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Niemann
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- BioArch, Department of Archaeology, University of York, York, UK
| | | | - Christian Carøe
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Emilie U Andersen-Ranberg
- The Qimmeq Project, University of Greenland, Nuussuaq, Greenland
- Department of Clinical Veterinary Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Peter D Jordan
- Arctic Centre and Groningen Institute of Archaeology, University of Groningen, Netherlands
| | - Elena Y Pavlova
- Arctic and Antarctic Research Institute, St. Petersburg, Russia
| | | | - Aleksei K Kasparov
- Institute for the History of Material Culture, Russian Academy of Sciences, St. Petersburg, Russia
| | - Varvara V Ivanova
- VNIIOkeangeologia Research Institute (The All-Russian Research Institute of Geology and Mineral Resources of the World Ocean), St. Petersburg, Russia
| | - Eske Willerslev
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Danish Institute for Advanced Study (D-IAS), University of Southern Denmark, Odense, Denmark
- Department of Zoology, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, University of Cambridge, Cambridge, UK
| | - Pontus Skoglund
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Francis Crick Institute, London, UK
| | - Merete Fredholm
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Sanne Eline Wennerberg
- Ministry of Fisheries, Hunting and Agriculture, Government of Greenland, Nuuk, Greenland
| | | | - Rune Dietz
- Department of Bioscience, Arctic Research Centre, Aarhus University, Roskilde, Denmark
| | - Christian Sonne
- The Qimmeq Project, University of Greenland, Nuussuaq, Greenland
- Department of Bioscience, Arctic Research Centre, Aarhus University, Roskilde, Denmark
- Henan Province Engineering Research Center for Biomass Value-added Products, School of Forestry, Henan Agricultural University, Zhengzhou, Henan, China
| | - Morten Meldgaard
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- The Qimmeq Project, University of Greenland, Nuussuaq, Greenland
| | - Love Dalén
- Department of Bioinformatics and Genetics, Swedish Museum of Natural History, Stockholm, Sweden
- Centre for Palaeogenetics, Stockholm, Sweden
| | - Greger Larson
- The Palaeogenomics and Bio-Archaeology Research Network, Research Laboratory for Archaeology and History of Art, University of Oxford, Oxford, UK
| | - Bent Petersen
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Centre of Excellence for Omics-Driven Computational Biodiscovery (COMBio), Faculty of Applied Sciences, AIMST University, Kedah, Malaysia
| | - Thomas Sicheritz-Pontén
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Centre of Excellence for Omics-Driven Computational Biodiscovery (COMBio), Faculty of Applied Sciences, AIMST University, Kedah, Malaysia
| | - Lutz Bachmann
- Natural History Museum, University of Oslo, Oslo, Norway
| | - Øystein Wiig
- Natural History Museum, University of Oslo, Oslo, Norway
| | - Tomas Marques-Bonet
- Institute of Evolutionary Biology (UPF-CSIC), Barcelona, Spain.
- Catalan Institution of Research and Advanced Studies, Barcelona, Spain
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
- Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anders J Hansen
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark.
- The Qimmeq Project, University of Greenland, Nuussuaq, Greenland
| | - M Thomas P Gilbert
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark.
- University Museum, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
18
|
Szostaczuk N, van Schothorst EM, Sánchez J, Priego T, Palou M, Bekkenkamp-Grovenstein M, Faustmann G, Obermayer-Pietsch B, Tiran B, Roob JM, Winklhofer-Roob BM, Keijer J, Palou A, Picó C. Identification of blood cell transcriptome-based biomarkers in adulthood predictive of increased risk to develop metabolic disorders using early life intervention rat models. FASEB J 2020; 34:9003-9017. [PMID: 32474969 DOI: 10.1096/fj.202000071rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/20/2022]
Abstract
Calorie restriction during gestation in rats has long-lasting adverse effects in the offspring. It induces metabolic syndrome-related alterations, which are partially reversed by leptin supplementation during lactation. We employed these conditions to identify transcript-based nutrient sensitive biomarkers in peripheral blood mononuclear cells (PBMCs) predictive of later adverse metabolic health. The best candidate was validated in humans. Transcriptome analysis of PBMCs from adult male Wistar rats of three experimental groups was performed: offspring of control dams (CON), and offspring of 20% calorie-restricted dams during gestation without (CR) and with leptin supplementation throughout lactation (CR-LEP). The expression of 401 genes was affected by gestational calorie restriction and reversed by leptin. The changes preceded metabolic syndrome-related phenotypic alterations. Of these genes, Npc1 mRNA levels were lower in CR vs CON, and normalized to CON in CR-LEP. In humans, NPC1 mRNA levels in peripheral blood cells (PBCs) were decreased in subjects with mildly impaired metabolic health compared to healthy subjects. Therefore, a set of potential transcript-based biomarkers indicative of a predisposition to metabolic syndrome-related alterations were identified, including NPC1, which was validated in humans. Low NPC1 transcript levels in PBCs are a candidate biomarker of increased risk for impaired metabolic health in humans.
Collapse
Affiliation(s)
- Nara Szostaczuk
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), University of the Balearic Islands, Palma de Mallorca, Spain
| | | | - Juana Sánchez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), University of the Balearic Islands, Palma de Mallorca, Spain.,Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - Teresa Priego
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), University of the Balearic Islands, Palma de Mallorca, Spain
| | - Mariona Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), University of the Balearic Islands, Palma de Mallorca, Spain.,Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain
| | | | - Gernot Faustmann
- Human Nutrition & Metabolism Research and Training Center, Institute of Molecular Biosciences, Karl-Franzens University of Graz, Graz, Austria.,Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Barbara Obermayer-Pietsch
- Division of Endocrinology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Beate Tiran
- Clinical Institute of Medical and Clinical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Johannes M Roob
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Brigitte M Winklhofer-Roob
- Human Nutrition & Metabolism Research and Training Center, Institute of Molecular Biosciences, Karl-Franzens University of Graz, Graz, Austria
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), University of the Balearic Islands, Palma de Mallorca, Spain.,Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics and Obesity), CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), University of the Balearic Islands, Palma de Mallorca, Spain.,Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain
| |
Collapse
|
19
|
Chitu V, Biundo F, Shlager GGL, Park ES, Wang P, Gulinello ME, Gokhan Ş, Ketchum HC, Saha K, DeTure MA, Dickson DW, Wszolek ZK, Zheng D, Croxford AL, Becher B, Sun D, Mehler MF, Stanley ER. Microglial Homeostasis Requires Balanced CSF-1/CSF-2 Receptor Signaling. Cell Rep 2020; 30:3004-3019.e5. [PMID: 32130903 PMCID: PMC7370656 DOI: 10.1016/j.celrep.2020.02.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 12/18/2019] [Accepted: 02/06/2020] [Indexed: 02/08/2023] Open
Abstract
CSF-1R haploinsufficiency causes adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). Previous studies in the Csf1r+/- mouse model of ALSP hypothesized a central role of elevated cerebral Csf2 expression. Here, we show that monoallelic deletion of Csf2 rescues most behavioral deficits and histopathological changes in Csf1r+/- mice by preventing microgliosis and eliminating most microglial transcriptomic alterations, including those indicative of oxidative stress and demyelination. We also show elevation of Csf2 transcripts and of several CSF-2 downstream targets in the brains of ALSP patients, demonstrating that the mechanisms identified in the mouse model are functional in humans. Our data provide insights into the mechanisms underlying ALSP. Because increased CSF2 levels and decreased microglial Csf1r expression have also been reported in Alzheimer's disease and multiple sclerosis, we suggest that the unbalanced CSF-1R/CSF-2 signaling we describe in the present study may contribute to the pathogenesis of other neurodegenerative conditions.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Gabriel G L Shlager
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Eun S Park
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maria E Gulinello
- Behavioral Core Facility, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Şölen Gokhan
- Institute for Brain Disorders and Neural Regeneration, Departments of Neurology, Neuroscience, and Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Harmony C Ketchum
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kusumika Saha
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Michael A DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Deyou Zheng
- The Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, and Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich 8057, Switzerland
| | - Daqian Sun
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Mark F Mehler
- Institute for Brain Disorders and Neural Regeneration, Departments of Neurology, Neuroscience, and Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
20
|
Fan J, Campioli E, Sottas C, Zirkin B, Papadopoulos V. Amhr2-Cre-Mediated Global Tspo Knockout. J Endocr Soc 2020; 4:bvaa001. [PMID: 32099945 PMCID: PMC7031085 DOI: 10.1210/jendso/bvaa001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/09/2020] [Indexed: 12/27/2022] Open
Abstract
Although the role of translocator protein (TSPO) in cholesterol transport in steroid-synthesizing cells has been studied extensively, recent studies of TSPO genetic depletion have questioned its role. Amhr2-Cre mice have been used to generate Leydig cell-specific Tspo conditional knockout (cKO) mice. Using the same Cre line, we were unable to generate Tspo cKO mice possibly because of genetic linkage between Tspo and Amhr2 and coexpression of Amhr2-Cre and Tspo in early embryonic development. We found that Amhr2-Cre is expressed during preimplantation stages, resulting in global heterozygous mice (gHE; Amhr2-Cre+/-,Tspo -/+). Two gHE mice were crossed, generating Amhr2-Cre-mediated Tspo global knockout (gKO; Tspo -/-) mice. We found that 33.3% of blastocysts at E3.5 to E4.5 showed normal morphology, whereas 66.7% showed delayed development, which correlates with the expected Mendelian proportions of Tspo +/+ (25%), Tspo -/- (25%), and Tspo +/- (50%) genotypes from crossing 2 Tspo -/+ mice. Adult Tspo gKO mice exhibited disturbances in neutral lipid homeostasis and reduced intratesticular and circulating testosterone levels, but no change in circulating basal corticosterone levels. RNA-sequencing data from mouse adrenal glands and lungs revealed transcriptome changes in response to the loss of TSPO, including changes in several cholesterol-binding and transfer proteins. This study demonstrates that Amhr2-Cre can be used to produce Tspo gKO mice instead of cKO, and can serve as a new global "Cre deleter." Moreover, our results show that Tspo deletion causes delayed preimplantation embryonic development, alters neutral lipid storage and steroidogenesis, and leads to transcriptome changes that may reflect compensatory mechanisms in response to the loss of function of TSPO.
Collapse
Affiliation(s)
- Jinjiang Fan
- The Research Institute of the McGill University Health Centre
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Enrico Campioli
- The Research Institute of the McGill University Health Centre
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Chantal Sottas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, US
| | - Barry Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, US
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, US
| |
Collapse
|
21
|
Abstract
The apolipoproteins are well known for their roles in both health and disease, as components of plasma lipoprotein particles, such as high-density lipoprotein (HDL), low-density lipoprotein (LDL), very-low-density lipoprotein (VLDL), chylomicrons, and metabolic, vascular- and inflammation-related disorders, such as cardiovascular disease, atherosclerosis, metabolic syndrome, and diabetes. Increasingly, their roles in neurovascular and neurodegenerative disorders are also being elucidated. They play major roles in lipid and cholesterol transport between blood and organs and are, therefore, critical to maintenance and homeostasis of the lipidome, with apolipoprotein-lipid interactions, including cholesterol, fatty acids, triglycerides, phospholipids, and isoprostanes. Further, they have important pleiotropic roles related to aging and longevity, which are largely managed through their many structural variants, including multiple isoforms, and a diversity of post-translational modifications. Consequently, tools for the characterization and accurate quantification of apolipoproteins, including their diverse array of variant forms, are required to understand their salutary and disease related roles. In this chapter we outline three distinct quantitative approaches suitable for targeting apolipoproteins: (1) multiplex immunoassays, (2) mass spectrometric immunoassay, and (3) multiple reaction monitoring, mass spectrometric quantification. We also discuss management of pre-analytical and experimental design variables.
Collapse
|
22
|
Eramo MJ, Lisnyak V, Formosa LE, Ryan MT. The ‘mitochondrial contact site and cristae organising system’ (MICOS) in health and human disease. J Biochem 2019; 167:243-255. [DOI: 10.1093/jb/mvz111] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022] Open
Abstract
AbstractThe ‘mitochondrial contact site and cristae organising system’ (MICOS) is an essential protein complex that promotes the formation, maintenance and stability of mitochondrial cristae. As such, loss of core MICOS components disrupts cristae structure and impairs mitochondrial function. Aberrant mitochondrial cristae morphology and diminished mitochondrial function is a pathological hallmark observed across many human diseases such as neurodegenerative conditions, obesity and diabetes mellitus, cardiomyopathy, and in muscular dystrophies and myopathies. While mitochondrial abnormalities are often an associated secondary effect to the pathological disease process, a direct role for the MICOS in health and human disease is emerging. This review describes the role of MICOS in the maintenance of mitochondrial architecture and summarizes both the direct and associated roles of the MICOS in human disease.
Collapse
Affiliation(s)
- Matthew J Eramo
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, 23 Innovation Walk, Monash University, 3800 Melbourne, Victoria, Australia
| | - Valerie Lisnyak
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, 23 Innovation Walk, Monash University, 3800 Melbourne, Victoria, Australia
| | - Luke E Formosa
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, 23 Innovation Walk, Monash University, 3800 Melbourne, Victoria, Australia
| | - Michael T Ryan
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, 23 Innovation Walk, Monash University, 3800 Melbourne, Victoria, Australia
| |
Collapse
|
23
|
Montgomery MK. Mitochondrial Dysfunction and Diabetes: Is Mitochondrial Transfer a Friend or Foe? BIOLOGY 2019; 8:E33. [PMID: 31083560 PMCID: PMC6627584 DOI: 10.3390/biology8020033] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/21/2018] [Accepted: 12/20/2018] [Indexed: 01/01/2023]
Abstract
Obesity, insulin resistance and type 2 diabetes are accompanied by a variety of systemic and tissue-specific metabolic defects, including inflammation, oxidative and endoplasmic reticulum stress, lipotoxicity, and mitochondrial dysfunction. Over the past 30 years, association studies and genetic manipulations, as well as lifestyle and pharmacological invention studies, have reported contrasting findings on the presence or physiological importance of mitochondrial dysfunction in the context of obesity and insulin resistance. It is still unclear if targeting mitochondrial function is a feasible therapeutic approach for the treatment of insulin resistance and glucose homeostasis. Interestingly, recent studies suggest that intact mitochondria, mitochondrial DNA, or other mitochondrial factors (proteins, lipids, miRNA) are found in the circulation, and that metabolic tissues secrete exosomes containing mitochondrial cargo. While this phenomenon has been investigated primarily in the context of cancer and a variety of inflammatory states, little is known about the importance of exosomal mitochondrial transfer in obesity and diabetes. We will discuss recent evidence suggesting that (1) tissues with mitochondrial dysfunction shed their mitochondria within exosomes, and that these exosomes impair the recipient's cell metabolic status, and that on the other hand, (2) physiologically healthy tissues can shed mitochondria to improve the metabolic status of recipient cells. In this context the determination of whether mitochondrial transfer in obesity and diabetes is a friend or foe requires further studies.
Collapse
Affiliation(s)
- Magdalene K Montgomery
- Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne 3010, Australia.
| |
Collapse
|
24
|
Montasser ME, O’Hare EA, Wang X, Howard AD, McFarland R, Perry JA, Ryan KA, Rice K, Jaquish CE, Shuldiner AR, Miller M, Mitchell BD, Zaghloul NA, Chang YPC. An APOO Pseudogene on Chromosome 5q Is Associated With Low-Density Lipoprotein Cholesterol Levels. Circulation 2018; 138:1343-1355. [PMID: 29593015 PMCID: PMC6162188 DOI: 10.1161/circulationaha.118.034016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 03/19/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Elevated levels of low-density lipoprotein cholesterol (LDL-C) are a major risk factor for cardiovascular disease via its contribution to the development and progression of atherosclerotic lesions. Although the genetic basis of LDL-C has been studied extensively, currently known genetic variants account for only ≈20% of the variation in LDL-C levels. METHODS Through an array-based association analysis in 1102 Amish subjects, we identified a variant strongly associated with LDL-C levels. Using a combination of genetic analyses, zebrafish models, and in vitro experiments, we sought to identify the causal gene driving this association. RESULTS We identified a founder haplotype associated with a 15 mg/dL increase in LDL-C on chromosome 5. After recombination mapping, the associated region contained 8 candidate genes. Using a zebrafish model to evaluate the relevance of these genes to cholesterol metabolism, we found that expression of the transcribed pseudogene, APOOP1, increased LDL-C and vascular plaque formation. CONCLUSIONS Based on these data, we propose that APOOP1 regulates levels of LDL-C in humans, thus identifying a novel mechanism of lipid homeostasis.
Collapse
Affiliation(s)
- May E. Montasser
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Elizabeth A. O’Hare
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Xiaochun Wang
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Alicia D. Howard
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Rebecca McFarland
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - James A. Perry
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Kathleen A. Ryan
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Kenneth Rice
- Dept of Biostatistics, University of Washington, Seattle, WA
| | | | - Alan R. Shuldiner
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Michael Miller
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Braxton D. Mitchell
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Norann A. Zaghloul
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Yen-Pei C. Chang
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
25
|
Liang Y, Lang AL, Zhang J, Chen J, Wang K, Chen L, Beier JI, Qian Y, Cai L. Exposure to Vinyl Chloride and Its Influence on Western Diet-Induced Cardiac Remodeling. Chem Res Toxicol 2018; 31:482-493. [PMID: 29727174 PMCID: PMC6167925 DOI: 10.1021/acs.chemrestox.8b00043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obesity, usually caused by high fat diets (HFD), is a major public health issue worldwide, causing obesity associated cardiomyopathy. Moreover, the environmental toxicant vinyl chloride (VC) can exacerbate HFD-induced fatty liver disease. However, whether VC serves to enhance obesity-associated cardiomyopathy remains unclear. This study aims to investigate the interaction of western diet (WD) containing relatively low fat (42%) with VC on cardiac remodeling and its underling mechanisms. Adult male C57BL/6J mice were exposed to WD coinhalation of low-dose VC (<1 ppm/d) for 12 weeks. Results showed that WD feeding for 12 weeks caused slight cardiac systolic dysfunction without significant hypertrophy or fibrosis, even with VC. Nevertheless, WD upregulated NF-κB function and expression of IL-1β and PAI-1, while VC showed no significant impact on these effects. In contrast, WD together with VC significantly increased the expression of CHOP and TGF-β1, key markers for endoplasmic reticulum stress and profibrotic cytokine, respectively. In summary, exposure to low-dose of environmental toxicant VC while a WD is consumed for a relatively short time does not have significant impact on cardiac remodeling except for a mild systolic dysfunction of the heart.
Collapse
Affiliation(s)
- Yaqin Liang
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Anna L. Lang
- Department of Pharmacology and Toxicology, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- Department of Hepatobiology and Toxicology Program, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Jian Zhang
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- The Center of Cardiovascular Disorders, The First Hospital of the Jilin University, Changchun 130021, China
| | - Jing Chen
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Kai Wang
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Liya Chen
- Department of Pharmacology and Toxicology, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- Department of Hepatobiology and Toxicology Program, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Juliane I. Beier
- Department of Pharmacology and Toxicology, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- Department of Hepatobiology and Toxicology Program, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
| | - Yan Qian
- Department of Pediatrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky 40292, United States
- The Center of Cardiovascular Disorders, The First Hospital of the Jilin University, Changchun 130021, China
| |
Collapse
|
26
|
Weijler AM, Schmidinger B, Kapiotis S, Laggner H, Hermann M. Oleic acid induces the novel apolipoprotein O and reduces mitochondrial membrane potential in chicken and human hepatoma cells. Biochimie 2018; 147:136-142. [PMID: 29432786 DOI: 10.1016/j.biochi.2018.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 02/05/2018] [Indexed: 12/28/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is marked by hepatic fat accumulation and reflects a spectrum of chronic liver diseases associated with obesity, impaired insulin sensitivity and dyslipidemia. Apolipoprotein O (ApoO) is a new member of the plasma apolipoprotein family that may play a role in lipid metabolism and electron transport activity of the mitochondrium. However, its physiological functions have not been elucidated yet. Based on our previous data in a non-mammalian experimental system [1], we hypothesized that hepatic expression of ApoO is tightly linked not only to diet-induced hepatosteatosis, but also to increased lipoprotein-production induced by, e.g., hormones and oxidative stress. To gain insight into a mammalian experimental system, we compared the effects of lipid loading on ApoO regulation in chicken hepatoma LMH cells with those in the human hepatoma cell line HepG2. Incubation of the cells with BSA-complexed oleic acid (OA-Alb) induced triglyceride accumulation, but did not affect cell viability. qPCR using specific primer pairs and Western blot analysis with in-house produced rabbit anti-ApoO antisera demonstrated significant increase in ApoO transcript and protein levels in both cell lines. ROS formation due to OA-Alb treatment was only slightly altered in LMH cells, indicating an intact antioxidant defense system of the cells. Oxidative stress applied by addition of H2O2 revealed induction of ApoO transcript and protein level in the same or even higher extent as monitored in the presence of OA-Alb. Upon treatment with estrogen for 24 h quantitative analysis of ApoO transcript and protein revealed increases of ApoO expression supporting the assumption that estrogen affects lipoprotein metabolism at various points. Furthermore, both cell lines showed a significant decrease of the mitochondrial membrane potential upon incubation with OA-Alb. Therefore, we assume that our findings support a role of ApoO as an effector of compromised mitochondrial function that likely accompanies the onset of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Anna M Weijler
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Barbara Schmidinger
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Stylianos Kapiotis
- The Central Laboratory, Hospital of the Divine Redeemer, Vienna, Austria
| | - Hilde Laggner
- Department of Medical Chemistry and Pathobiochemistry, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Marcela Hermann
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
27
|
Tian F, Wu CL, Yu BL, Liu L, Hu JR. Apolipoprotein O expression in mouse liver enhances hepatic lipid accumulation by impairing mitochondrial function. Biochem Biophys Res Commun 2017. [PMID: 28647361 DOI: 10.1016/j.bbrc.2017.06.128] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Apolipoprotein O (ApoO) was recently observed in the cellular mitochondrial inner membrane, which plays a role in mitochondrial function and is associated with myocardiopathy. Empirical information on the physiological functions of apoO is therefore limited. In this study, we aimed to elucidate the effect of apoO on hepatic fatty acid metabolism. An adenoviral vector expressing hApoO was constructed and introduced into chow diet and high-fat diet induced mice and the L02 human hepatoma cell line. High levels of hApoO mRNA and protein were detected in the liver, and the expression of lipid metabolism genes was significantly altered compared with negative controls. The liver function indices (serum ALT and AST) were clearly elevated, and the ultrastructure of cellular mitochondria was distinctly altered in the liver after apoO overexpression. Further, mitochondrial membrane potential decreased with hApoO treatment in L02 cells. These results establish a link between apoO and lipid accumulation and could suggest a new pathway for regulating non-alcoholic fatty liver disease progression.
Collapse
Affiliation(s)
- Feng Tian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Chen-Lu Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Bi-Lian Yu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Ling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jia-Rui Hu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
28
|
Kurowski BG, Treble-Barna A, Pitzer AJ, Wade SL, Martin LJ, Chima RS, Jegga A. Applying Systems Biology Methodology To Identify Genetic Factors Possibly Associated with Recovery after Traumatic Brain Injury. J Neurotrauma 2017; 34:2280-2290. [PMID: 28301983 DOI: 10.1089/neu.2016.4856] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality worldwide. It is linked with a number of medical, neurological, cognitive, and behavioral sequelae. The influence of genetic factors on the biology and related recovery after TBI is poorly understood. Studies that seek to elucidate the impact of genetic influences on neurorecovery after TBI will lead to better individualization of prognosis and inform development of novel treatments, which are considerably lacking. Current genetic studies related to TBI have focused on specific candidate genes. The objectives of this study were to use a system biology-based approach to identify biologic processes over-represented with genetic variants previously implicated in clinical outcomes after TBI and identify unique genes potentially related to recovery after TBI. After performing a systematic review to identify genes in the literature associated with clinical outcomes, we used the genes identified to perform a systems biology-based integrative computational analysis to ascertain the interactions between molecular components and to develop models for regulation and function of genes involved in TBI recovery. The analysis identified over-representation of genetic variants primarily in two biologic processes: response to injury (cell proliferation, cell death, inflammatory response, and cellular metabolism) and neurocognitive and behavioral reserve (brain development, cognition, and behavior). Overall, this study demonstrates the use of a systems biology-based approach to identify unique/novel genes or sets of genes important to the recovery process. Findings from this systems biology-based approach provide additional insight into the potential impact of genetic variants on the underlying complex biological processes important to TBI recovery and may inform the development of empirical genetic-related studies for TBI. Future studies that combine systems biology methodology and genomic, proteomic, and epigenetic approaches are needed in TBI.
Collapse
Affiliation(s)
- Brad G Kurowski
- 1 Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Amery Treble-Barna
- 2 Division of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Alexis J Pitzer
- 3 Department of Psychology, Xavier University , Cincinnati, Ohio
| | - Shari L Wade
- 1 Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Lisa J Martin
- 1 Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Ranjit S Chima
- 1 Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Anil Jegga
- 1 Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine , Cincinnati, Ohio
| |
Collapse
|
29
|
Abstract
The heart utilizes large amounts of fatty acids as energy providing substrates. The physiological balance of lipid uptake and oxidation prevents accumulation of excess lipids. Several processes that affect cardiac function, including ischemia, obesity, diabetes mellitus, sepsis, and most forms of heart failure lead to altered fatty acid oxidation and often also to the accumulation of lipids. There is now mounting evidence associating certain species of these lipids with cardiac lipotoxicity and subsequent myocardial dysfunction. Experimental and clinical data are discussed and paths to reduction of toxic lipids as a means to improve cardiac function are suggested.
Collapse
Affiliation(s)
- P Christian Schulze
- From the Divisions of Cardiology, Friedrich-Schiller-University Jena, Germany, and Columbia University, New York, NY (P.C.S.); Metabolic Biology Laboratory, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.D.); and Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY (I.J.G.).
| | - Konstantinos Drosatos
- From the Divisions of Cardiology, Friedrich-Schiller-University Jena, Germany, and Columbia University, New York, NY (P.C.S.); Metabolic Biology Laboratory, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.D.); and Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY (I.J.G.)
| | - Ira J Goldberg
- From the Divisions of Cardiology, Friedrich-Schiller-University Jena, Germany, and Columbia University, New York, NY (P.C.S.); Metabolic Biology Laboratory, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.D.); and Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY (I.J.G.)
| |
Collapse
|
30
|
Ong SB, Hausenloy DJ. Mitochondrial Dynamics as a Therapeutic Target for Treating Cardiac Diseases. Handb Exp Pharmacol 2017; 240:251-279. [PMID: 27844171 DOI: 10.1007/164_2016_7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mitochondria are dynamic in nature and are able to shift their morphology between elongated interconnected mitochondrial networks and a fragmented disconnected arrangement by the processes of mitochondrial fusion and fission, respectively. Changes in mitochondrial morphology are regulated by the mitochondrial fusion proteins - mitofusins 1 and 2 (Mfn1 and 2), and optic atrophy 1 (Opa1) as well as the mitochondrial fission proteins - dynamin-related peptide 1 (Drp1) and fission protein 1 (Fis1). Despite having a unique spatial arrangement, cardiac mitochondria have been implicated in a variety of disorders including ischemia-reperfusion injury (IRI), heart failure, diabetes, and pulmonary hypertension. In this chapter, we review the influence of mitochondrial dynamics in these cardiac disorders as well as their potential as therapeutic targets in tackling cardiovascular disease.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Cardiovascular and Metabolic Disorders (CVMD) Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore.
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders (CVMD) Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore
- The Hatter Cardiovascular Institute, University College London Hospitals and Medical School, London, UK
| |
Collapse
|
31
|
White CR, Datta G, Giordano S. High-Density Lipoprotein Regulation of Mitochondrial Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:407-429. [PMID: 28551800 DOI: 10.1007/978-3-319-55330-6_22] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lipoproteins play a key role in regulating plasma and tissue levels of cholesterol. Apolipoprotein B (apoB)-containing lipoproteins, including chylomicrons, very-low density lipoprotein (VLDL) and low-density lipoprotein (LDL), serve as carriers of triglycerides and cholesterol and deliver these metabolites to peripheral tissues. In contrast, high-density lipoprotein (HDL) mediates Reverse Cholesterol Transport (RCT), a process by which excess cholesterol is removed from the periphery and taken up by hepatocytes where it is metabolized and excreted. Anti-atherogenic properties of HDL have been largely ascribed to apoA-I, the major protein component of the lipoprotein particle. The inflammatory response associated with atherosclerosis and ischemia-reperfusion (I-R) injury has been linked to the development of mitochondrial dysfunction. Under these conditions, an increase in reactive oxygen species (ROS) formation induces damage to mitochondrial structural elements, leading to a reduction in ATP synthesis and initiation of the apoptotic program. Recent studies suggest that HDL-associated apoA-I and lysosphingolipids attenuate mitochondrial injury by multiple mechanisms, including the suppression of ROS formation and induction of autophagy. Other apolipoproteins, however, present in lower abundance in HDL particles may exert opposing effects on mitochondrial function. This chapter examines the role of HDL-associated apolipoproteins and lipids in the regulation of mitochondrial function and bioenergetics.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Geeta Datta
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Samantha Giordano
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
32
|
Anand R, Strecker V, Urbach J, Wittig I, Reichert AS. Mic13 Is Essential for Formation of Crista Junctions in Mammalian Cells. PLoS One 2016; 11:e0160258. [PMID: 27479602 PMCID: PMC4968808 DOI: 10.1371/journal.pone.0160258] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/15/2016] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial cristae are connected to the inner boundary membrane via crista junctions which are implicated in the regulation of oxidative phosphorylation, apoptosis, and import of lipids and proteins. The MICOS complex determines formation of crista junctions. We performed complexome profiling and identified Mic13, also termed Qil1, as a subunit of the MICOS complex. We show that MIC13 is an inner membrane protein physically interacting with MIC60, a central subunit of the MICOS complex. Using the CRISPR/Cas method we generated the first cell line deleted for MIC13. These knockout cells show a complete loss of crista junctions demonstrating that MIC13 is strictly required for the formation of crista junctions. MIC13 is required for the assembly of MIC10, MIC26, and MIC27 into the MICOS complex. However, it is not needed for the formation of the MIC60/MIC19/MIC25 subcomplex suggesting that the latter is not sufficient for crista junction formation. MIC13 is also dispensable for assembly of respiratory chain complexes and for maintaining mitochondrial network morphology. Still, lack of MIC13 resulted in a moderate reduction of mitochondrial respiration. In summary, we show that MIC13 has a fundamental role in crista junction formation and that assembly of respiratory chain supercomplexes is independent of mitochondrial cristae shape.
Collapse
Affiliation(s)
- Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University, Medical Faculty, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Valentina Strecker
- Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Jennifer Urbach
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University, Medical Faculty, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Ilka Wittig
- Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Goethe-University, Frankfurt am Main, Germany
- Cluster of Excellence “Macromolecular Complexes”, Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Andreas S. Reichert
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University, Medical Faculty, Universitätsstr. 1, 40225, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
33
|
Nuebel E, Manganas P, Tokatlidis K. Orphan proteins of unknown function in the mitochondrial intermembrane space proteome: New pathways and metabolic cross-talk. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2613-2623. [PMID: 27425144 PMCID: PMC5404111 DOI: 10.1016/j.bbamcr.2016.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/07/2016] [Accepted: 07/12/2016] [Indexed: 12/14/2022]
Abstract
The mitochondrial intermembrane space (IMS) is involved in protein transport, lipid homeostasis and metal ion exchange, while further acting in signalling pathways such as apoptosis. Regulation of these processes involves protein modifications, as well as stress-induced import or release of proteins and other signalling molecules. Even though the IMS is the smallest sub-compartment of mitochondria, its redox state seems to be tightly regulated. However, the way in which this compartment participates in the cross-talk between the multiple organelles and the cytosol is far from understood. Here we focus on newly identified IMS proteins that may represent future challenges in mitochondrial research. We present an overview of the import pathways, the recently discovered new components of the IMS proteome and how these relate to key aspects of cell signalling and progress made in stem cell and cancer research. A brief overview of the classic mitochondrial import pathways is featured Recent studies assigning a number of new proteins to the mitochondrial IMS are discussed Analysis of the expanded IMS proteomes can provide insights into organelle cross-talk and signalling pathways
Collapse
Affiliation(s)
- Esther Nuebel
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Phanee Manganas
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Kostas Tokatlidis
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK.
| |
Collapse
|
34
|
Kozjak-Pavlovic V. The MICOS complex of human mitochondria. Cell Tissue Res 2016; 367:83-93. [PMID: 27245231 DOI: 10.1007/s00441-016-2433-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/09/2016] [Indexed: 11/25/2022]
Abstract
Mitochondria are organelles of endosymbiotic origin, surrounded by two membranes. The inner membrane forms invaginations called cristae that enhance its surface and are important for mitochondrial function. A recently described mitochondrial contact site and cristae organizing system (MICOS) in the inner mitochondrial membrane is crucial for the formation and maintenance of cristae structure. The MICOS complex in human mitochondria exhibits specificities and greater complexity in comparison to the yeast system. Many subunits of this complex have been previously described, but several others and their function remain to be explored. This review will summarize our present knowledge about the human MICOS complex and its constituents, while discussing the future research perspectives in this exciting and important field.
Collapse
Affiliation(s)
- Vera Kozjak-Pavlovic
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, 97074, Würzburg, Germany.
| |
Collapse
|
35
|
Schmidinger B, Weijler AM, Schneider WJ, Hermann M. Hepatosteatosis and estrogen increase apolipoprotein O production in the chicken. Biochimie 2016; 127:37-43. [PMID: 27126072 DOI: 10.1016/j.biochi.2016.04.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/21/2016] [Indexed: 10/21/2022]
Abstract
Apolipoprotein O (ApoO) is a recently discovered plasma apolipoprotein that may also play a role in the mitochondrial inner membrane. Possibly due to this complexity, its physiological functions have not been elucidated yet. To gain insight from a non-mammalian experimental system, we have investigated the regulation of ApoO levels in an alternative, well-suited model for studies on lipid metabolism, the chicken. qPCR using specific primer pairs and Western blot analysis with our rabbit anti-chicken ApoO antiserum demonstrated ApoO in the liver of chickens fed a control or a fat-enriched diet, as well as in 2 chicken hepatoma cell lines, LMH cells and the estrogen-responsive LMH-2A cells, under conditions of lipid loading by incubation with BSA-complexed oleic acid. Induced triglyceride accumulation in both the liver and the hepatic cells was associated with significantly increased levels of ApoO mRNA and protein. Furthermore, upon treatment for 24 h with estrogen of the estrogen receptor-expressing LMH-2A cells, quantitative analysis of ApoO transcripts and Western blotting revealed increases of ApoO expression. Finally, upon a single administration of estrogen to roosters that leads to hyperlipidemia, higher hepatic levels of both ApoO transcript and protein were observed within 24 h. Based on these data, we propose that hepatic expression of ApoO is tightly linked not only to diet-induced hepatosteatosis, but also to increased lipoprotein-production induced by, e.g., hormones. The findings support a role of ApoO as an effector of compromised mitochondrial function that likely accompanies the onset of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Barbara Schmidinger
- Max F. Perutz Laboratories, Department of Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Anna M Weijler
- Max F. Perutz Laboratories, Department of Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Wolfgang J Schneider
- Max F. Perutz Laboratories, Department of Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Marcela Hermann
- Max F. Perutz Laboratories, Department of Medical Biochemistry, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
36
|
O'Connell GC, Nichols C, Guo G, Croston TL, Thapa D, Hollander JM, Pistilli EE. IL-15Rα deficiency in skeletal muscle alters respiratory function and the proteome of mitochondrial subpopulations independent of changes to the mitochondrial genome. Mitochondrion 2015; 25:87-97. [PMID: 26458787 DOI: 10.1016/j.mito.2015.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/24/2015] [Accepted: 10/05/2015] [Indexed: 10/22/2022]
Abstract
Interleukin-15 receptor alpha knockout (IL15RαKO) mice exhibit a greater skeletal muscle mitochondrial density with an altered mitochondrial morphology. However, the mechanism and functional impact of these changes have not been determined. In this study, we characterized the functional, proteomic, and genomic alterations in mitochondrial subpopulations isolated from the skeletal muscles of IL15RαKO mice and B6129 background control mice. State 3 respiration was greater in interfibrillar mitochondria and whole muscle ATP levels were greater in IL15RαKO mice supporting the increases in respiration rate. However, the state 3/state 4 ratio was lower, suggesting some degree of respiratory uncoupling. Proteomic analyses identified several markers independently in mitochondrial subpopulations that are associated with these functional alterations. Next Generation Sequencing of mtDNA revealed a high degree of similarity between the mitochondrial genomes of IL15RαKO mice and controls in terms of copy number, consensus coding and the presence of minor alleles, suggesting that the functional and proteomic alterations we observed occurred independent of alterations to the mitochondrial genome. These data provide additional evidence to implicate IL-15Rα as a regulator of skeletal muscle phenotypes through effects on the mitochondrion, and suggest these effects are driven by alterations to the mitochondrial proteome.
Collapse
Affiliation(s)
| | | | - Ge Guo
- Division of Exercise Physiology, United States
| | | | | | - John M Hollander
- Division of Exercise Physiology, United States; Center for Cardiovascular and Respiratory Sciences, United States
| | - Emidio E Pistilli
- Division of Exercise Physiology, United States; Center for Cardiovascular and Respiratory Sciences, United States; Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, United States.
| |
Collapse
|
37
|
Tuck E, Estabel J, Oellrich A, Maguire AK, Adissu HA, Souter L, Siragher E, Lillistone C, Green AL, Wardle-Jones H, Carragher DM, Karp NA, Smedley D, Adams NC, Bussell JN, Adams DJ, Ramírez-Solis R, Steel KP, Galli A, White JK. A gene expression resource generated by genome-wide lacZ profiling in the mouse. Dis Model Mech 2015; 8:1467-78. [PMID: 26398943 PMCID: PMC4631787 DOI: 10.1242/dmm.021238] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/10/2015] [Indexed: 01/26/2023] Open
Abstract
Knowledge of the expression profile of a gene is a critical piece of information required to build an understanding of the normal and essential functions of that gene and any role it may play in the development or progression of disease. High-throughput, large-scale efforts are on-going internationally to characterise reporter-tagged knockout mouse lines. As part of that effort, we report an open access adult mouse expression resource, in which the expression profile of 424 genes has been assessed in up to 47 different organs, tissues and sub-structures using a lacZ reporter gene. Many specific and informative expression patterns were noted. Expression was most commonly observed in the testis and brain and was most restricted in white adipose tissue and mammary gland. Over half of the assessed genes presented with an absent or localised expression pattern (categorised as 0-10 positive structures). A link between complexity of expression profile and viability of homozygous null animals was observed; inactivation of genes expressed in ≥ 21 structures was more likely to result in reduced viability by postnatal day 14 compared with more restricted expression profiles. For validation purposes, this mouse expression resource was compared with Bgee, a federated composite of RNA-based expression data sets. Strong agreement was observed, indicating a high degree of specificity in our data. Furthermore, there were 1207 observations of expression of a particular gene in an anatomical structure where Bgee had no data, indicating a large amount of novelty in our data set. Examples of expression data corroborating and extending genotype-phenotype associations and supporting disease gene candidacy are presented to demonstrate the potential of this powerful resource.
Collapse
Affiliation(s)
- Elizabeth Tuck
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Jeanne Estabel
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Anika Oellrich
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Hibret A Adissu
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, 25 Orde Street, Toronto, Canada M5T 3H7
| | - Luke Souter
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Emma Siragher
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Angela L Green
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | | | | | - Natasha A Karp
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Damian Smedley
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Niels C Adams
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | | | - James N Bussell
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - David J Adams
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Karen P Steel
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Antonella Galli
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | | |
Collapse
|
38
|
Koob S, Barrera M, Anand R, Reichert AS. The non-glycosylated isoform of MIC26 is a constituent of the mammalian MICOS complex and promotes formation of crista junctions. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:1551-63. [PMID: 25764979 DOI: 10.1016/j.bbamcr.2015.03.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/13/2015] [Accepted: 03/02/2015] [Indexed: 01/05/2023]
Abstract
Mitochondrial membrane architecture is important for organelle function. Alterations thereof are linked to a number of human disorders including diabetes and cardiomyopathy. The MICOS complex was recently reported to be a central player determining cristae structure and formation of crista junctions. Here we investigated the functional role of MIC26, a lipoprotein formerly termed APOO. Its levels are increased in diabetic heart tissue and in blood plasma of patients suffering from acute coronary syndrome. We demonstrate that human MIC26 exists in three distinct forms: (1) a glycosylated and secreted 55kDa protein, (2) an ER/Golgi-resident form thereof, and (3) a non-glycosylated 22kDa mitochondrial protein. The latter isoform spans the mitochondrial inner membrane and physically interacts with several MICOS complex subunits such as MIC60, MIC27, and MIC10. We further demonstrate that MIC26 and MIC27, a homologous protein formerly termed APOOL, regulate their levels in an antagonistic manner. Both proteins are positively correlated with the levels of MIC10 as well as tafazzin, an enzyme required for cardiolipin remodeling. Overexpression of MIC26 induced fragmentation of mitochondria, promoted ROS formation and resulted in impaired mitochondrial respiration. Downregulation of MIC26 induced a decrease in mitochondrial oxygen consumption, whereas mitochondrial network morphology and ROS levels remained unaffected. MIC26 depletion led to alterations in mitochondrial ultrastructure and caused a significant reduction in the number of crista junctions. In summary, we show that the human apolipoprotein MIC26 is a bona fide subunit of the MICOS complex and that MIC26 is linked to cardiolipin metabolism and promotes crista junction formation.
Collapse
Affiliation(s)
- Sebastian Koob
- Mitochondrial Biology, Buchmann Institute of Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Miguel Barrera
- Mitochondrial Biology, Buchmann Institute of Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University, Medical Faculty, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Andreas S Reichert
- Mitochondrial Biology, Buchmann Institute of Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany; Institute of Biochemistry and Molecular Biology I, Heinrich Heine University, Medical Faculty, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
39
|
Sung MM, Hamza SM, Dyck JRB. Myocardial metabolism in diabetic cardiomyopathy: potential therapeutic targets. Antioxid Redox Signal 2015; 22:1606-30. [PMID: 25808033 DOI: 10.1089/ars.2015.6305] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Cardiovascular complications in diabetes are particularly serious and represent the primary cause of morbidity and mortality in diabetic patients. Despite early observations of cardiac dysfunction in diabetic humans, cardiomyopathy unique to diabetes has only recently been recognized. RECENT ADVANCES Research has focused on understanding the pathogenic mechanisms underlying the initiation and development of diabetic cardiomyopathy. Emerging data highlight the importance of altered mitochondrial function as a major contributor to cardiac dysfunction in diabetes. Mitochondrial dysfunction occurs by several mechanisms involving altered cardiac substrate metabolism, lipotoxicity, impaired cardiac insulin and glucose homeostasis, impaired cellular and mitochondrial calcium handling, oxidative stress, and mitochondrial uncoupling. CRITICAL ISSUES Currently, treatment is not specifically tailored for diabetic patients with cardiac dysfunction. Given the multifactorial development and progression of diabetic cardiomyopathy, traditional treatments such as anti-diabetic agents, as well as cellular and mitochondrial fatty acid uptake inhibitors aimed at shifting the balance of cardiac metabolism from utilizing fat to glucose may not adequately target all aspects of this condition. Thus, an alternative treatment such as resveratrol, which targets multiple facets of diabetes, may represent a safe and promising supplement to currently recommended clinical therapy and lifestyle changes. FUTURE DIRECTIONS Elucidation of the mechanisms underlying the initiation and progression of diabetic cardiomyopathy is essential for development of effective and targeted treatment strategies. Of particular interest is the investigation of alternative therapies such as resveratrol, which can function as both preventative and mitigating agents in the management of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Miranda M Sung
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| | - Shereen M Hamza
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| | - Jason R B Dyck
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| |
Collapse
|
40
|
Roul D, Recchia FA. Metabolic alterations induce oxidative stress in diabetic and failing hearts: different pathways, same outcome. Antioxid Redox Signal 2015; 22:1502-14. [PMID: 25836025 PMCID: PMC4449624 DOI: 10.1089/ars.2015.6311] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Several authors have proposed a link between altered cardiac energy substrate metabolism and reactive oxygen species (ROS) generation. A cogent evidence of this association has been found in diabetic cardiomyopathy (dCM); however, experimental findings in animal models of heart failure (HF) and in human myocardium also seem to support the coexistence of the two alterations in HF. CRITICAL ISSUES Two important questions remain open: whether pathological changes in metabolism play an important role in enhancing oxidative stress and whether there is a common pathway linking altered substrate utilization and activation of ROS-generating enzymes, independently of the underlying cardiac pathology. In this regard, the comparison between dCM and HF is intriguing, in that these pathological conditions display very different cardiac metabolic phenotypes. RECENT ADVANCES Our literature review on this topic indicates that a vast body of knowledge is now available documenting the relationship between the metabolism of energy substrates and ROS generation in dCM. In some cases, biochemical mechanisms have been identified. On the other hand, only a few and relatively recent studies have explored this phenomenon in HF and their conclusions are not consistent. FUTURE DIRECTIONS Better methods of investigation, especially in vivo, will be necessary to test whether the metabolic fate of certain substrates is causally linked to ROS production. If successful, these studies will place a new emphasis on the potential clinical relevance of metabolic modulators, which might indirectly mitigate cardiac oxidative stress in dCM, HF, and, possibly, in other pathological conditions.
Collapse
Affiliation(s)
- David Roul
- 1Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Fabio A Recchia
- 1Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania.,2Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
41
|
Zhang Y, Bao M, Dai M, Wang X, He W, Tan T, Lin D, Wang W, Wen Y, Zhang R. Cardiospecific CD36 suppression by lentivirus-mediated RNA interference prevents cardiac hypertrophy and systolic dysfunction in high-fat-diet induced obese mice. Cardiovasc Diabetol 2015; 14:69. [PMID: 26036798 PMCID: PMC4464858 DOI: 10.1186/s12933-015-0234-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/26/2015] [Indexed: 01/12/2023] Open
Abstract
Background Fatty acid (FA) catabolism abnormality has been proved to play an important role in obesity-related cardiomyopathy. We hypothesized that cardiospecific suppression of CD36, the predominant membrane FA transporter, would protect against obesity-related cardiomyopathy. Methods Four-wk-old male C57BL/6 J mice were fed with either high-fat-diet (HFD) or control-normal-diet for 2 wk. Then they were subjected to intramyocardial injection with recombinant lentiviral vectors containing short hairpin RNAs to selectively downregulate the expression of either cardiac CD36 or irrelevant gene by RNA interference. After a 10-wk continuation of the diet, biochemical, functional, morphological, histological, metabolic and molecular profiles were assessed. Results HFD administration elicited obesity, cardiac hypertrophy and systolic dysfunction accompanied with elevated serum levels of blood urea nitrogen (BUN), creatinine, fasting serum glucose (FSG), total cholesterol (TC) and triglyceride. Additionally, HFD consumption promoted lipid accumulation and reactive oxygen species (ROS) generation in the cardiomyocytes. Cardiospecific CD36 inhibition protected against HFD induced cardiac remodeling by decreasing heart/body weight ratio, increasing left ventricular (LV) ejection fraction and fractional shortening as well as normalizing LV diameter, without influencing body weight gain. Inhibition of cardiac CD36 also mitigated obesity induced alteration in BUN, creatinine and triglyceride, but had no effect on FSG or TC. Moreover, cardiospecific CD36 deficiency corrected myocardial lipid overaccumulation and intracellular ROS overproduction that were induced by HFD feeding. Conclusions Cardiospecific CD36 inhibition protects against the aggravation of cardiac functional and morphological changes associated with HFD induced obesity. CD36 represents a potential therapeutic target for obesity cardiomyopathy.
Collapse
Affiliation(s)
- Yijie Zhang
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Central Laboratory of Renmin Hospital, Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Mingwei Bao
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Mingyan Dai
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Xin Wang
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Wenbo He
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Tuantuan Tan
- Department of Ultrasonography, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Dandan Lin
- Department of Oncology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Wei Wang
- Department of Thoracic Surgery, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Ying Wen
- Department of Cardiology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China. .,Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| | - Rui Zhang
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Peoples Republic of China.
| |
Collapse
|
42
|
Hu WS, Ting WJ, Chiang WD, Pai P, Yeh YL, Chang CH, Lin WT, Huang CY. The Heart Protection Effect of Alcalase Potato Protein Hydrolysate Is through IGF1R-PI3K-Akt Compensatory Reactivation in Aging Rats on High Fat Diets. Int J Mol Sci 2015; 16:10158-72. [PMID: 25950762 PMCID: PMC4463638 DOI: 10.3390/ijms160510158] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 12/27/2022] Open
Abstract
The prevalence of obesity is high in older adults. Alcalase potato protein hydrolysate (APPH), a nutraceutical food, might have greater benefits and be more economical than hypolipidemic drugs. In this study, serum lipid profiles and heart protective effects were evaluated in high fat diet (HFD) induced hyperlipidemia in aging rats treated with APPH (15, 45 and 75 mg/kg/day) and probucol (500 mg/kg/day). APPH treatments reduced serum triacylglycerol (TG), total cholesterol (TC), and low density lipoprotein (LDL) levels to the normal levels expressed in the control group. Additionally, the IGF1R-PI3K-Akt survival pathway was reactivated, and Fas-FADD (Fas-associated death domain) induced apoptosis was inhibited by APPH treatments (15 and 45 mg/kg/day) in HFD aging rat hearts. APPH (75 mg/kg/day) rather than probucol (500 mg/kg/day) treatment could reduce serum lipids without affecting HDL expression. The heart protective effect of APPH in aging rats with hyperlipidemia was through lowering serum lipids and enhancing the activation of the compensatory IGF1R-PI3K-Akt survival pathway.
Collapse
Affiliation(s)
- Wei-Syun Hu
- PhD Program for Aging, China Medical University, Taichung 40402, Taiwan.
| | - Wei-Jen Ting
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Wen-Dee Chiang
- Department of Food Science, College of Agriculture, Tunghai University, Taichung 40704, Taiwan.
| | - Peiying Pai
- Division of Cardiology, China Medical University Hospital, Taichung 40402, Taiwan.
| | - Yu-Lan Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua 50006, Taiwan.
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan.
| | - Chung-Ho Chang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan Town 35053, Taiwan.
| | - Wan-Teng Lin
- Department of Hospitality Management, College of Agriculture, Tunghai University, Taichung 40704, Taiwan.
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung 40402, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|
43
|
Chauhan S, Mandell MA, Deretic V. IRGM governs the core autophagy machinery to conduct antimicrobial defense. Mol Cell 2015; 58:507-21. [PMID: 25891078 DOI: 10.1016/j.molcel.2015.03.020] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 02/19/2015] [Accepted: 03/12/2015] [Indexed: 01/21/2023]
Abstract
IRGM, encoded by a uniquely human gene conferring risk for inflammatory diseases, affects autophagy through an unknown mechanism. Here, we show how IRGM controls autophagy. IRGM interacts with ULK1 and Beclin 1 and promotes their co-assembly thus governing the formation of autophagy initiation complexes. We further show that IRGM interacts with pattern recognition receptors including NOD2. IRGM, NOD2, and ATG16L1, all of which are Crohn's disease risk factors, form a molecular complex to modulate autophagic responses to microbial products. NOD2 enhances K63-linked polyubiquitination of IRGM, which is required for interactions of IRGM with the core autophagy factors and for microbial clearance. Thus, IRGM plays a direct role in organizing the core autophagy machinery to endow it with antimicrobial and anti-inflammatory functions.
Collapse
Affiliation(s)
- Santosh Chauhan
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA.
| | - Michael A Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA.
| |
Collapse
|
44
|
Ott C, Dorsch E, Fraunholz M, Straub S, Kozjak-Pavlovic V. Detailed analysis of the human mitochondrial contact site complex indicate a hierarchy of subunits. PLoS One 2015; 10:e0120213. [PMID: 25781180 PMCID: PMC4363703 DOI: 10.1371/journal.pone.0120213] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 01/20/2015] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial inner membrane folds into cristae, which significantly increase its surface and are important for mitochondrial function. The stability of cristae depends on the mitochondrial contact site (MICOS) complex. In human mitochondria, the inner membrane MICOS complex interacts with the outer membrane sorting and assembly machinery (SAM) complex, to form the mitochondrial intermembrane space bridging complex (MIB). We have created knockdown cell lines of most of the MICOS and MIB components and have used them to study the importance of the individual subunits for the cristae formation and complex stability. We show that the most important subunits of the MIB complex in human mitochondria are Mic60/Mitofilin, Mic19/CHCHD3 and an outer membrane component Sam50. We provide additional proof that ApoO indeed is a subunit of the MICOS and MIB complexes and propose the name Mic23 for this protein. According to our results, Mic25/CHCHD6, Mic27/ApoOL and Mic23/ApoO appear to be periphery subunits of the MICOS complex, because their depletion does not affect cristae morphology or stability of other components.
Collapse
Affiliation(s)
- Christine Ott
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Eva Dorsch
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Martin Fraunholz
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Sebastian Straub
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Vera Kozjak-Pavlovic
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, Würzburg, Germany
| |
Collapse
|
45
|
Rouet P, Harmancey R, Turkieh A, Caubère C, Barutaut M, Koukoui F, Dambrin C, Galinier M, Smih F. [A matter of fat: APOO regulates mitochondrial function in the heart]. Med Sci (Paris) 2015; 31:31-4. [PMID: 25658728 DOI: 10.1051/medsci/20153101010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Philippe Rouet
- Inserm U1048, équipe 7, obésité et insuffisance cardiaque : approches moléculaires et cliniques, BP 84225, 1, avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| | - Romain Harmancey
- Inserm U1048, équipe 7, obésité et insuffisance cardiaque : approches moléculaires et cliniques, BP 84225, 1, avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| | - Annie Turkieh
- Inserm U1048, équipe 7, obésité et insuffisance cardiaque : approches moléculaires et cliniques, BP 84225, 1, avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| | - Céline Caubère
- Inserm U1048, équipe 7, obésité et insuffisance cardiaque : approches moléculaires et cliniques, BP 84225, 1, avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| | - Manon Barutaut
- Inserm U1048, équipe 7, obésité et insuffisance cardiaque : approches moléculaires et cliniques, BP 84225, 1, avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| | - François Koukoui
- Inserm U1048, équipe 7, obésité et insuffisance cardiaque : approches moléculaires et cliniques, BP 84225, 1, avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| | - Camille Dambrin
- Inserm U1048, équipe 7, obésité et insuffisance cardiaque : approches moléculaires et cliniques, BP 84225, 1, avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| | - Michel Galinier
- Inserm U1048, équipe 7, obésité et insuffisance cardiaque : approches moléculaires et cliniques, BP 84225, 1, avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| | - Fatima Smih
- Inserm U1048, équipe 7, obésité et insuffisance cardiaque : approches moléculaires et cliniques, BP 84225, 1, avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| |
Collapse
|