1
|
Fidelito G, Todorovski I, Cluse L, Vervoort SJ, Taylor RA, Watt MJ. Lipid-metabolism-focused CRISPR screens identify enzymes of the mevalonate pathway as essential for prostate cancer growth. Cell Rep 2025; 44:115470. [PMID: 40146774 DOI: 10.1016/j.celrep.2025.115470] [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: 10/07/2024] [Revised: 01/22/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Dysregulated lipid metabolism plays an important role in prostate cancer, although the understanding of the essential regulatory processes in tumorigenesis is incomplete. We employ a CRISPR-Cas9 screen using a custom human lipid metabolism knockout library to identify essential genes for prostate cancer survival. Screening in three prostate cancer cell lines reveals 63 shared dependencies, with enrichment in terpenoid backbone synthesis and N-glycan biosynthesis. Independent knockout of key genes of the mevalonate pathway reduces cell proliferation. Further investigation focuses on NUS1, a subunit of cis-prenyltransferase required for dolichol synthesis. NUS1 knockout decreases tumor growth in vivo and viability in patient-derived xenograft (PDX)-derived organoids. Mechanistic studies reveal that loss of NUS1 promotes oxidative stress, lipid peroxidation and ferroptosis sensitivity, endoplasmic reticulum (ER) stress, and G1 cell-cycle arrest, and it dampens androgen receptor (AR) signaling, collectively leading to growth arrest. This study highlights the critical role of the mevalonate-dolichol-N-glycan biosynthesis pathway, particularly NUS1, in prostate cancer survival and growth.
Collapse
Affiliation(s)
- Gio Fidelito
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Izabela Todorovski
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Leonie Cluse
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Stephin J Vervoort
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Renea A Taylor
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Melbourne Urological Research Alliance (MURAL), Monash University, Clayton, VIC 3168, Australia; Cabrini Institute, Cabrini Health, Malvern, VIC 3144, Australia.
| | - Matthew J Watt
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
2
|
Xue C, Zeng P, Gong K, Li Q, Feng Z, Wang M, Chen S, Yang Y, Li J, Zhang S, Yin Z, Liang Y, Yan T, Yu M, Feng K, Zhao D, Yang X, Zhang X, Ma L, Iwakiri Y, Chen L, Tang X, Chen Y, Chen H, Duan Y. Nogo-B inhibition facilitates cholesterol metabolism to reduce hypercholesterolemia. Cell Rep 2024; 43:114691. [PMID: 39235944 DOI: 10.1016/j.celrep.2024.114691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/09/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
Abstract
The strategy of lowering cholesterol levels by promoting cholesterol excretion is still lacking, and few molecular targets act on multiple cholesterol metabolic processes. In this study, we find that Nogo-B deficiency/inhibition simultaneously promotes hepatic uptake of cholesterol and cholesterol excretion. Nogo-B deficiency decreases cholesterol levels by activating ATP-binding cassette transporters (ABCs), apolipoprotein E (ApoE), and low-density lipoprotein receptor (LDLR) expression. We discover that Nogo-B interacts with liver X receptor α (LXRα), and Nogo-B deficiency inhibits ubiquitination degradation of LXRα, thereby enhancing its function on cholesterol excretion. Decreased cellular cholesterol levels further activate SREBP2 and LDLR expression, thereby promoting hepatic uptake of cholesterol. Nogo-B inhibition decreases atherosclerotic plaques and cholesterol levels in mice, and Nogo-B levels are correlated to cholesterol levels in human plasma. In this study, Nogo-B deficiency/inhibition not only promotes hepatic uptake of blood cholesterol but also facilitates cholesterol excretion. This study reports a strategy to lower cholesterol levels by inhibiting Nogo-B expression to promote hepatic cholesterol uptake and cholesterol excretion.
Collapse
Affiliation(s)
- Chao Xue
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Peng Zeng
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Qian Li
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Zian Feng
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Mengyao Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Shasha Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yanfang Yang
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Jiaqi Li
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Zequn Yin
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yingquan Liang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Tengteng Yan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Miao Yu
- Medical College of Soochow University, Suzhou 215031, China
| | - Ke Feng
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Dan Zhao
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xia Zhang
- Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin 301800, China
| | - Likun Ma
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Liang Chen
- College of Life Science, Anhui Medical University, Hefei 230032, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No. 17 People's South Road, Chengdu, Sichuan 610041, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China.
| | - Houzao Chen
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China.
| | - Yajun Duan
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
3
|
Hu R, Long S, Luo M, Tang B, Tan T, Dong W, Wang Q, Zhang J. Hyperglycemia Inhibits Hepatic SHBG Synthesis Through the NGBR-AMPK-HNF4 Pathway in Rats with Polycystic Ovary Syndrome Induced by Letrozole in Combination with a High-Fat Diet. Mol Nutr Food Res 2024; 68:e2300915. [PMID: 38862276 DOI: 10.1002/mnfr.202300915] [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: 12/23/2023] [Revised: 02/25/2024] [Indexed: 06/13/2024]
Abstract
SCOPE Polycystic ovary syndrome (PCOS) is closely related to non-alcoholic fatty liver disease (NAFLD), and sex hormone-binding globulin (SHBG) is a glycoprotein produced by the liver. Hepatic lipogenesis inhibits hepatic SHBG synthesis, which leads to hyperandrogenemia and ovarian dysfunction in PCOS. Therefore, this study aims to characterize the mechanism whereby liver lipogenesis inhibits SHBG synthesis. METHODS AND RESULTS This study establishes a rat model of PCOS complicated by NAFLD using a high-fat diet in combination with letrozole and performs transcriptomic analysis of the liver. Transcriptomic analysis of the liver shows that the expression of neurite growth inhibitor-B receptor (NgBR), hepatocyte nuclear factor 4α (HNF4α), and SHBG is low. Meantime, HepG2 cells are treated with palmitic acid (PA) to model NAFLD in vitro, which causes decreases in the expression of NgBR, HNF4α, and SHBG. However, the expression of HNF4α and SHBG is restored by treatment with the AMP-activated protein kinase (AMPK) agonist AICAR. CONCLUSIONS NgBR regulates the expression of HNF4α by activating the AMPK signaling pathway, thereby affecting the synthesis of SHBG in the liver. Further mechanistic studies regarding the effect of liver fat on NGBR expression are warranted.
Collapse
Affiliation(s)
- Rao Hu
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Shuanglian Long
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Min Luo
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Bowen Tang
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Tao Tan
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Weilei Dong
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qian Wang
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jiaming Zhang
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
4
|
Xiao H, Du X, Tao Z, Jing N, Bao S, Gao W, Dong B, Fang Y. Taurine Inhibits Ferroptosis Mediated by the Crosstalk between Tumor Cells and Tumor-Associated Macrophages in Prostate Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303894. [PMID: 38031260 PMCID: PMC10797466 DOI: 10.1002/advs.202303894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/17/2023] [Indexed: 12/01/2023]
Abstract
Tumor-associated macrophages (TAMs) play an essential role in tumor therapeutic resistance. Although the lethal effect of ferroptosis on tumor cells is well reported, how TAMs inhibit the effect of ferroptosis in tumors has not been clearly defined. In this study, it is demonstrated that TAM-secreted taurine suppresses ferroptosis in prostate cancer (PCa) by activating the Liver X receptor alpha/Stearoyl-Coenzyme A desaturase 1 (LXRα/SCD1) pathway. Blocking taurine intake via inhibition of taurine transporter TauT restores the sensitivity to ferroptosis in tumors. Furthermore, LXRα activates the transcription of both miR-181a-5p and its binding protein FUS to increase the recruitment of miR-181a-5p in tumor-derived extracellular vesicles (EVs). It is observed that macrophages appear to be recipient cells of the miR-181a-5p-enriched EVs. Intake of miR-181a-5p in macrophages promotes their M2 polarization and enhances the taurine export by inhibiting expression of its target gene lats1, which in turn inactivates the hippo pathway and results in a Yes-associated protein (YAP) nuclear translocation for transcriptional activation of both M2 polarization-related genes such as ARG1 and CD163 and the taurine transport gene TauT. Taken together, the findings indicate a reciprocal interaction between PCa cells and TAMs as a positive feedback-loop to repress ferroptosis in PCa, mediated by TAM-secreted taurine and tumor EV-delivered miR-181a-5p.
Collapse
Affiliation(s)
- Huixiang Xiao
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterDepartment of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
| | - Xinxing Du
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
| | - Zhenkeke Tao
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterDepartment of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
| | - Nan Jing
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Shijia Bao
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterDepartment of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
| | - Wei‐Qiang Gao
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterDepartment of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Baijun Dong
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
| | - Yu‐Xiang Fang
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterDepartment of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
| |
Collapse
|
5
|
Chen Y, Li Q, Zhao S, Sun L, Yin Z, Wang X, Li X, Iwakiri Y, Han J, Duan Y. Berberine protects mice against type 2 diabetes by promoting PPARγ-FGF21-GLUT2-regulated insulin sensitivity and glucose/lipid homeostasis. Biochem Pharmacol 2023; 218:115928. [PMID: 37979703 DOI: 10.1016/j.bcp.2023.115928] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/20/2023]
Abstract
Type 2 diabetes (T2D) is a chronic, burdensome disease that is characterized by disordered insulin sensitivity and disturbed glucose/lipid homeostasis. Berberine (BBR) has multiple therapeutic actions on T2D, including regulation of glucose and lipid metabolism, improvement of insulin sensitivity and energy expenditure. Recently, the function of BBR on fibroblast growth factor 21 (FGF21) has been identified. However, if BBR ameliorates T2D through FGF21, the underlying mechanisms remain unknown. Herein, we used T2D wild type (WT) and FGF21 global knockout (FKO) mice [mouse T2D model: established by high-fat diet (HFD) feeding plus streptozotocin (STZ) injection], and hepatocyte-specific peroxisome proliferator activated receptor γ (PPARγ) deficient (PPARγHepKO) mice, and cultured human liver carcinoma cells line, HepG2 cells, to characterize the role of BBR in glucose/lipid metabolism and insulin sensitivity. We found that BBR activated FGF21 expression by up-regulating PPARγ expression at the cellular level. Meanwhile, BBR ameliorated glucosamine hydrochloride (Glcn)-induced insulin resistance and increased glucose transporter 2 (GLUT2) expression in a PPARγ/FGF21-dependent manner. In T2D mice, BBR up-regulated the expression of PPARγ, FGF21 and GLUT2 in the liver, and GLUT2 in the pancreas. BBR also reversed T2D-induced insulin resistance, liver lipid accumulation, and damage in liver and pancreas. However, FGF21 deficiency diminished these effects of BBR on diabetic mice. Altogether, our study demonstrates that the therapeutic effects of BBR on T2D were partly accomplished by activating PPARγ-FGF21-GLUT2 signaling pathway. The discovery of this new pathway provides a deeper understanding of the mechanism of BBR for T2D treatment.
Collapse
Affiliation(s)
- Yi Chen
- Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Qi Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Shiwei Zhao
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Lei Sun
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Zequn Yin
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaolin Wang
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xiaoju Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Jihong Han
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
6
|
Singh AK, Chaube B, Citrin KM, Fowler JW, Lee S, Catarino J, Knight J, Lowery S, Shree S, Boutagy N, Ruz-Maldonado I, Harry K, Shanabrough M, Ross TT, Malaker S, Suárez Y, Fernández-Hernando C, Grabinska K, Sessa WC. Loss of cis-PTase function in the liver promotes a highly penetrant form of fatty liver disease that rapidly transitions to hepatocellular carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566870. [PMID: 38014178 PMCID: PMC10680637 DOI: 10.1101/2023.11.13.566870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Obesity-linked fatty liver is a significant risk factor for hepatocellular carcinoma (HCC)1,2; however, the molecular mechanisms underlying the transition from non-alcoholic fatty liver disease (NAFLD) to HCC remains unclear. The present study explores the role of the endoplasmic reticulum (ER)-associated protein NgBR, an essential component of the cis-prenyltransferases (cis-PTase) enzyme3, in chronic liver disease. Here we show that genetic depletion of NgBR in hepatocytes of mice (N-LKO) intensifies triacylglycerol (TAG) accumulation, inflammatory responses, ER/oxidative stress, and liver fibrosis, ultimately resulting in HCC development with 100% penetrance after four months on a high-fat diet. Comprehensive genomic and single cell transcriptomic atlas from affected livers provides a detailed molecular analysis of the transition from liver pathophysiology to HCC development. Importantly, pharmacological inhibition of diacylglycerol acyltransferase-2 (DGAT2), a key enzyme in hepatic TAG synthesis, abrogates diet-induced liver damage and HCC burden in N-LKO mice. Overall, our findings establish NgBR/cis-PTase as a critical suppressor of NAFLD-HCC conversion and suggests that DGAT2 inhibition may serve as a promising therapeutic approach to delay HCC formation in patients with advanced non-alcoholic steatohepatitis (NASH).
Collapse
Affiliation(s)
- Abhishek K. Singh
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Balkrishna Chaube
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Kathryn M Citrin
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Joseph Wayne Fowler
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Sungwoon Lee
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Jonatas Catarino
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - James Knight
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Sarah Lowery
- Chemistry Research Building, Yale University, New Haven, CT, USA
| | - Sonal Shree
- Molecular Biophysics & Biochemistry, Yale University, New Haven, CT, USA
| | - Nabil Boutagy
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Inmaculada Ruz-Maldonado
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Kathy Harry
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Marya Shanabrough
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Stacy Malaker
- Chemistry Research Building, Yale University, New Haven, CT, USA
| | - Yajaira Suárez
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Carlos Fernández-Hernando
- Department of Comparative Medicine, Yale Center for Molecular and Systems Metabolism and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Kariona Grabinska
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - William C. Sessa
- Department of Pharmacology, and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| |
Collapse
|
7
|
Slieker RC, Donnelly LA, Akalestou E, Lopez-Noriega L, Melhem R, Güneş A, Abou Azar F, Efanov A, Georgiadou E, Muniangi-Muhitu H, Sheikh M, Giordano GN, Åkerlund M, Ahlqvist E, Ali A, Banasik K, Brunak S, Barovic M, Bouland GA, Burdet F, Canouil M, Dragan I, Elders PJM, Fernandez C, Festa A, Fitipaldi H, Froguel P, Gudmundsdottir V, Gudnason V, Gerl MJ, van der Heijden AA, Jennings LL, Hansen MK, Kim M, Leclerc I, Klose C, Kuznetsov D, Mansour Aly D, Mehl F, Marek D, Melander O, Niknejad A, Ottosson F, Pavo I, Duffin K, Syed SK, Shaw JL, Cabrera O, Pullen TJ, Simons K, Solimena M, Suvitaival T, Wretlind A, Rossing P, Lyssenko V, Legido Quigley C, Groop L, Thorens B, Franks PW, Lim GE, Estall J, Ibberson M, Beulens JWJ, 't Hart LM, Pearson ER, Rutter GA. Identification of biomarkers for glycaemic deterioration in type 2 diabetes. Nat Commun 2023; 14:2533. [PMID: 37137910 PMCID: PMC10156700 DOI: 10.1038/s41467-023-38148-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 04/18/2023] [Indexed: 05/05/2023] Open
Abstract
We identify biomarkers for disease progression in three type 2 diabetes cohorts encompassing 2,973 individuals across three molecular classes, metabolites, lipids and proteins. Homocitrulline, isoleucine and 2-aminoadipic acid, eight triacylglycerol species, and lowered sphingomyelin 42:2;2 levels are predictive of faster progression towards insulin requirement. Of ~1,300 proteins examined in two cohorts, levels of GDF15/MIC-1, IL-18Ra, CRELD1, NogoR, FAS, and ENPP7 are associated with faster progression, whilst SMAC/DIABLO, SPOCK1 and HEMK2 predict lower progression rates. In an external replication, proteins and lipids are associated with diabetes incidence and prevalence. NogoR/RTN4R injection improved glucose tolerance in high fat-fed male mice but impaired it in male db/db mice. High NogoR levels led to islet cell apoptosis, and IL-18R antagonised inflammatory IL-18 signalling towards nuclear factor kappa-B in vitro. This comprehensive, multi-disciplinary approach thus identifies biomarkers with potential prognostic utility, provides evidence for possible disease mechanisms, and identifies potential therapeutic avenues to slow diabetes progression.
Collapse
Affiliation(s)
- Roderick C Slieker
- Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUMC, Amsterdam, the Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Louise A Donnelly
- Population Health & Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Elina Akalestou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Livia Lopez-Noriega
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Rana Melhem
- CHUM Research Centre and University of Montreal, Montreal, QC, Canada
| | - Ayşim Güneş
- IRCM and University of Montreal, Montreal, QC, Canada
| | | | - Alexander Efanov
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, US
| | - Eleni Georgiadou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Hermine Muniangi-Muhitu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Mahsa Sheikh
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Mikael Åkerlund
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Emma Ahlqvist
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ashfaq Ali
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark
| | - Marko Barovic
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Medical Faculty, Dresden, Germany
| | - Gerard A Bouland
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frédéric Burdet
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mickaël Canouil
- INSERM U1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille University Hospital, Lille, F-59000, France
| | - Iulian Dragan
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Petra J M Elders
- Department of General Practice and Elderly Care Medicine, Amsterdam Public Health Research Institute, Amsterdam UMC-location VUmc, Amsterdam, the Netherlands
| | | | - Andreas Festa
- Eli Lilly Regional Operations GmbH, Vienna, Austria
- 1st Medical Department, LK Stockerau, Niederösterreich, Austria
| | - Hugo Fitipaldi
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Phillippe Froguel
- INSERM U1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille University Hospital, Lille, F-59000, France
- Division of Systems Biology, Department of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Valborg Gudmundsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Icelandic Heart Association, Kopavogur, Iceland
| | | | - Amber A van der Heijden
- Department of General Practice and Elderly Care Medicine, Amsterdam Public Health Research Institute, Amsterdam UMC-location VUmc, Amsterdam, the Netherlands
| | - Lori L Jennings
- Novartis Institutes for Biomedical Research, Cambridge, MA, 02139, USA
| | - Michael K Hansen
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, Spring House, PA, USA
| | - Min Kim
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicines, King's College London, London, UK
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- CHUM Research Centre and University of Montreal, Montreal, QC, Canada
| | | | - Dmitry Kuznetsov
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Florence Mehl
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Diana Marek
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Anne Niknejad
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Filip Ottosson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Imre Pavo
- Eli Lilly Regional Operations GmbH, Vienna, Austria
| | - Kevin Duffin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, US
| | - Samreen K Syed
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, US
| | - Janice L Shaw
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, US
| | - Over Cabrera
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, US
| | - Timothy J Pullen
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Diabetes, Guy's Campus King's College London, London, UK
| | | | - Michele Solimena
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Medical Faculty, Dresden, Germany
- Molecular Diabetology, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | | | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Valeriya Lyssenko
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, Bergen, Norway
- Genomics, Diabetes and Endocrinology Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Cristina Legido Quigley
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicines, King's College London, London, UK
| | - Leif Groop
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Finnish Institute of Molecular Medicine, Helsinki University, Helsinki, Finland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Paul W Franks
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - Gareth E Lim
- CHUM Research Centre and University of Montreal, Montreal, QC, Canada
| | | | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Joline W J Beulens
- Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUMC, Amsterdam, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Leen M 't Hart
- Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUMC, Amsterdam, the Netherlands.
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands.
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Ewan R Pearson
- Population Health & Genomics, School of Medicine, University of Dundee, Dundee, UK.
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
- CHUM Research Centre and University of Montreal, Montreal, QC, Canada.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
8
|
Huang H, Guo S, Chen Y, Liu Y, Jin J, Liang Y, Fan L, Xiang R. Increased RTN3 phenocopies nonalcoholic fatty liver disease by inhibiting the AMPK-IDH2 pathway. MedComm (Beijing) 2023; 4:e226. [PMID: 36925557 PMCID: PMC10013133 DOI: 10.1002/mco2.226] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 03/16/2023] Open
Abstract
Reticulon 3 (RTN3), an endoplasmic reticulum protein, is crucial in neurodegenerative and kidney diseases. However, the role of RTN3 in liver tissues has not been described. Here, we employed public datasets, patients, and several animal models to explore the role of RTN3 in nonalcoholic fatty liver disease (NAFLD). The underlying mechanisms were studied in primary hepatocytes and L02 cells in vitro. We found an increased expression of RTN3 in NAFLD patients, high-fat diet mice, and oxidized low-density lipoprotein-treated L02 cells. The RTN3 transgenic mice exhibited the phenotypes of fatty liver and lipid accumulation. Single-cell RNA sequencing analysis indicated that increased RTN3 might induce mitochondrial dysfunction. We further showed this in primary hepatocytes, the L02 cell line, and the Caenorhabditis elegans strain. Mechanistically, RTN3 regulated these events through its interactions with glucose-regulated protein 78 (GRP78), which further inhibited the adenosine 5 monophosphate-activated protein kinase (AMPK)-isocitrate dehydrogenase 2 (IDH2) pathway. In the end, knockout of RTN3 relieved fatty liver and mitochondrial dysfunction. Our study indicated that RTN3 was important in NAFLD and lipid catabolism and that an increase in RTN3 in the liver might be a risk factor for nonalcoholic steatohepatitis and NAFLD.
Collapse
Affiliation(s)
- Hao Huang
- Department of NephrologyXiangya HospitalCentral South UniversityChangshaChina
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaChina
- Hunan Key Laboratory of Animal Models for Human DiseasesSchool of Life SciencesCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Shuai Guo
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaChina
| | - Ya‐Qin Chen
- Department of CardiologySecond Xiangya HospitalCentral South UniversityChangshaChina
| | - Yu‐Xing Liu
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaChina
| | - Jie‐Yuan Jin
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaChina
| | - Yun Liang
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaChina
| | - Liang‐Liang Fan
- Department of NephrologyXiangya HospitalCentral South UniversityChangshaChina
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaChina
- Hunan Key Laboratory of Animal Models for Human DiseasesSchool of Life SciencesCentral South UniversityChangshaChina
| | - Rong Xiang
- Department of NephrologyXiangya HospitalCentral South UniversityChangshaChina
- Department of Cell BiologySchool of Life SciencesCentral South UniversityChangshaChina
- Hunan Key Laboratory of Animal Models for Human DiseasesSchool of Life SciencesCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
- Department of CardiologySecond Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
9
|
Role of Neurite Outgrowth Inhibitor B Receptor in hepatic steatosis. Acta Histochem 2022; 124:151977. [DOI: 10.1016/j.acthis.2022.151977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022]
|
10
|
Li H, Liu L, Chen HY, Yan X, Li RL, Lan J, Xue KY, Li X, Zhuo CL, Lin L, Li LY, Wu Z, Zhang D, Wang XM, Huang WJ, Wang Y, Jiang W, Zhou L. Mogrol suppresses lung cancer cell growth by activating AMPK-dependent autophagic death and inducing p53-dependent cell cycle arrest and apoptosis. Toxicol Appl Pharmacol 2022; 444:116037. [PMID: 35489526 DOI: 10.1016/j.taap.2022.116037] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/23/2022] [Accepted: 04/22/2022] [Indexed: 02/08/2023]
Abstract
Lung carcinoma is the leading cause of cancer-related death worldwide. Chemotherapy remains the cornerstone of lung cancer treatment. Unfortunately, most types of cancer will develop resistance to chemotherapies over the time. One of the efforts to prevent the chemotherapy resistance is to find alternative chemotherapy drugs. Mogrol has been found to have antitumor activity. However, little is known about the pharmacological mechanisms underlying the suppression of mogrol on lung cancers. In this study, we observed that mogrol exposure significantly reduced the tumor volume and weight in tumor-bearing nude mice without obvious effect on body weight and cardiac function. Mogrol also significantly inhibited the proliferation and migration of lung cancer cells, including non-small-cell lung carcinoma cells, A549, H1299, H1975 and SK-MES-1 cells, with no obvious effect on control human bronchial epithelial cells (HBE). Further studies revealed that mogrol stirred excessive autophagy and autophagic flux, and finally, autophagic cell death, in lung cancer cells, which could be attenuated by autophagy inhibitors, 3-MA and chloroquine. Furthermore, mogrol significantly activated AMPK to induce autophagy and autophagic cell death, which could be abrogated by Compound C, an AMPK inhibitor. In addition, mogrol induced a significant increase in p53 activity in lung cancer cells, accompanied with cell cycle arrest and apoptosis, which could be weakened by p53 silence. Our results indicated that mogrol effectively suppressed lung cancer cells in vivo and in vitro by inducing the excessive autophagy and autophagic cell death via activating AMPK signaling pathway, as well as cell cycle arrest and apoptosis via activating p53 pathway.
Collapse
Affiliation(s)
- He Li
- School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, PR China; Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Linling Liu
- School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, PR China; Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Hong-Ying Chen
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xin Yan
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Ru-Li Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Jie Lan
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Kun-Yue Xue
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xue Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Cai-Li Zhuo
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Lan Lin
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Ling-Yu Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Zhuang Wu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Die Zhang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xue-Mei Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Wen-Jing Huang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Yingling Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Wei Jiang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China.
| | - Liming Zhou
- School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
11
|
Ma J, Zeng P, Liu L, Zhu M, Zheng J, Wang C, Zhao X, Hu W, Yang X, Duan Y, Han J, Miao QR, Chen Y. Peroxisome Proliferator-Activated Receptor-Gamma Reduces ER Stress and Inflammation via Targeting NGBR Expression. Front Pharmacol 2022; 12:817784. [PMID: 35111067 PMCID: PMC8801792 DOI: 10.3389/fphar.2021.817784] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/27/2021] [Indexed: 01/01/2023] Open
Abstract
Increased Nogo-B receptor (NGBR) expression in the liver improves insulin sensitivity by reducing endoplasmic reticulum stress (ER stress) and activating the AMPK pathway, although it remains elusive the mechanisms by which NGBR is induced. In this study, we found that PPARγ ligands (rosiglitazone or pioglitazone) increased NGBR expression in hepatic cells and HUVECs. Furthermore, promoter analysis defined two PPREs (PPARγ-responsive elements) in the promoter region of NGBR, which was further confirmed by the ChIP assay. In vivo, using liver-specific PPARγ deficient (PPARγLKO) mice, we identified the key role of PPARγ expression in pioglitazone-induced NGBR expression. Meanwhile, the basal level of ER stress and inflammation was slightly increased by NGBR knockdown. However, the inhibitory effect of rosiglitazone on inflammation was abolished while rosiglitazone-inhibited ER stress was weakened by NGBR knockdown. Taken together, these findings show that NGBR is a previously unrecognized target of PPARγ activation and plays an essential role in PPARγ-reduced ER stress and inflammation.
Collapse
Affiliation(s)
- Jialing Ma
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Peng Zeng
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Lipei Liu
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Mengmeng Zhu
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Juan Zheng
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Chengyi Wang
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaokang Zhao
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Wenquan Hu
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Xiaoxiao Yang
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yajun Duan
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.,College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Qing R Miao
- Diabetes and Obesity Research Center, New York University Long Island School of Medicine, New York, NY, United States
| | - Yuanli Chen
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| |
Collapse
|
12
|
Wang D, Yin Z, Han L, Zhang M, Li H, Yang X, Chen Y, Zhang S, Han J, Duan Y. Ascorbic acid inhibits transcriptional activities of LXRα to ameliorate lipid metabolism disorder. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
13
|
Sfikas G, Psallas M, Koumaras C, Imprialos K, Perdikakis E, Doumas M, Giouleme O, Karagiannis A, Athyros VG. Prevalence, Diagnosis, and Treatment with 3 Different Statins of Non-alcoholic Fatty Liver Disease/Non-alcoholic Steatohepatitis in Military Personnel. Do Genetics Play a Role? Curr Vasc Pharmacol 2021; 19:572-581. [DOI: 10.2174/1570161118666201015152921] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/18/2022]
Abstract
Background:
Non-alcoholic fatty liver disease (NAFLD) and its severe form, non-alcoholic
steatohepatitis (NASH), are major health problems worldwide. Genetics may play a role in
the pathogenesis of NAFLD/NASH.
Aim:
To investigate the prevalence of NAFLD/NASH in 5,400 military personnel and evaluate
the effect of treatment with 3 statins on NAFLD/NASH using 2 non-invasive scores [NAFLD Activity
Score (NAS); Fibrosis-4 score (FIB-4)].
Methods:
During the mandatory annual medical check-up, military personnel underwent a clinical
and laboratory evaluation. Participants with NAFLD/NASH were randomized into 4 groups
(n=151 each): diet-exercise, atorvastatin, rosuvastatin, or pitavastatin for 1 year (i.e., until the next
routine evaluation).
Results:
From all the participants, 613 had NAFLD/NASH (prevalence 11.3 vs 39.8% in the general
population, p<0.001), and a total of 604 consented to participate in the study. After a year of
treatment, the diet-exercise group showed no significant changes in both scores (NAS 4.98 baseline
vs. 5.62, p=0.07; FIB-4 3.42 vs. 3.52, p=0.7). For the atorvastatin group, both scores were reduced
(NAS 4.97 vs 1.95, p<0.001, FIB-4 3.56 vs 0.83, p<0.001), for rosuvastatin (NAS 5.55 vs
1.81, p<0.001, FIB-4 3.61 vs 0.79, p<0.001), and for pitavastatin (NAS 4.89 vs 1.99, p<0.001,
FIB-4 3.78 vs 0.87, p<0.001).
Conclusions:
Atorvastatin, rosuvastatin, and pitavastatin have a beneficial and safe effect in
NAFLD/NASH patients as recorded by the improvement in the NAS (representing NAFLD activity)
and FIB-4 (representing liver fibrosis) scores. Since both those with and without NAFLD/-
NASH shared several baseline characteristics, genetics may play a role in the pathogenesis of
NAFLD/NASH and its treatment with statins.
Collapse
Affiliation(s)
- Georgios Sfikas
- 1st Department of Internal Medicine, 424 General Military Training Hospital, Thessaloniki, Greece
| | - Michael Psallas
- 1st Department of Internal Medicine, 424 General Military Training Hospital, Thessaloniki, Greece
| | - Charalambos Koumaras
- 1st Department of Internal Medicine, 424 General Military Training Hospital, Thessaloniki, Greece
| | - Konstantinos Imprialos
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University School of Medicine, Thessaloniki, Greece
| | - Evangelos Perdikakis
- Department of Ultrasonography, 424 General Military Training Hospital, Thessaloniki, Greece
| | - Michael Doumas
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University School of Medicine, Thessaloniki, Greece
| | - Olga Giouleme
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University School of Medicine, Thessaloniki, Greece
| | - Asterios Karagiannis
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University School of Medicine, Thessaloniki, Greece
| | - Vasilios G. Athyros
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University School of Medicine, Thessaloniki, Greece
| |
Collapse
|
14
|
Feng K, Liu Y, Sun J, Zhao C, Duan Y, Wang W, Yan K, Yan X, Sun H, Hu Y, Han J. Compound Danshen Dripping Pill inhibits doxorubicin or isoproterenol-induced cardiotoxicity. Biomed Pharmacother 2021; 138:111531. [PMID: 34311530 DOI: 10.1016/j.biopha.2021.111531] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is the advanced heart disease with high morbidity and mortality. Compound DanShen Dripping Pill (CDDP) is a widely used Traditional Chinese Medicine for cardiovascular disease treatment. Herein, we investigated if CDDP can protect mice against doxorubicin (DOX) or isoprenaline (ISO)-induced HF. After 3 days feeding of normal chow containing CDDP, mice were started DOX or ISO treatment for 4 weeks or 18 days. At the end of treatment, mice were conducted electrocardiogram and echocardiographic test. Blood and heart samples were determined biochemical parameters, myocardial structure and expression of the related molecules. CDDP normalized DOX/ISO-induced heart weight changes, HF parameters and fibrogenesis. The DOX/ISO-impaired left ventricular ejection fraction and fractional shortening were restored by CDDP. Mechanistically, CDDP blocked DOX/ISO-inhibited expression of antioxidant enzymes and DOX/ISO-induced expression of pro-fibrotic molecules, inflammation and cell apoptosis. Additional DOX/ISO-impaired targets in cardiac function but protected by CDDP were identified by RNAseq, qRT-PCR and Western blot. In addition, CDDP protected cardiomyocytes against oxygen-glucose deprivation-induced injuries. Taken together, our study shows that CDDP can protect against myocardial injuries in different models, suggesting its potential application for HF treatment.
Collapse
Affiliation(s)
- Ke Feng
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yuxin Liu
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Jia Sun
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
| | - Chunlai Zhao
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Wenjia Wang
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
| | - Kaijing Yan
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China; The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China; Tasly Pharmaceutical Group Co., Ltd, Tianjin, China
| | - Xijun Yan
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China; The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China; Tasly Pharmaceutical Group Co., Ltd, Tianjin, China
| | - He Sun
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China; The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China; Tasly Pharmaceutical Group Co., Ltd, Tianjin, China
| | - Yunhui Hu
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China.
| | - Jihong Han
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China.
| |
Collapse
|
15
|
Chen Y, Hu W, Li Q, Zhao S, Zhao D, Zhang S, Wei Z, Yang X, Chen Y, Li X, Liao C, Han J, Miao QR, Duan Y. NGBR is required to ameliorate type 2 diabetes in mice by enhancing insulin sensitivity. J Biol Chem 2021; 296:100624. [PMID: 33812996 PMCID: PMC8111265 DOI: 10.1016/j.jbc.2021.100624] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/11/2021] [Accepted: 03/31/2021] [Indexed: 02/07/2023] Open
Abstract
The reduction of insulin resistance or improvement of insulin sensitivity is the most effective treatment for type 2 diabetes (T2D). We previously reported that Nogo-B receptor (NGBR), encoded by the NUS1 gene, is required for attenuating hepatic lipogenesis by blocking nuclear translocation of liver X receptor alpha, suggesting its important role in regulating hepatic lipid metabolism. Herein, we demonstrate that NGBR expression was decreased in the liver of obesity-associated T2D patients and db/db mice. NGBR knockout in mouse hepatocytes resulted in increased blood glucose, insulin resistance, and beta-cell loss. High-fat diet (HFD)/streptozotocin (STZ)-treated mice presented the T2D phenotype by showing increased nonesterified fatty acid (NEFA) and triglyceride (TG) in the liver and plasma and increased insulin resistance and beta-cell loss. AAV-mediated NGBR overexpression in the liver reduced NEFA and TG in the liver and circulation and improved liver functions. Consequently, HFD/STZ-treated mice with hepatic NGBR overexpression had increased insulin sensitivity and reduced beta-cell loss. Mechanistically, NGBR overexpression restored insulin signaling of AMPKα1-dependent phosphorylation of AKT and GSK3β. NGBR overexpression also reduced expression of endoplasmic reticulum stress-associated genes in the liver and skeletal muscle to improve insulin sensitivity. Together, our results reveal that NGBR is required to ameliorate T2D in mice, providing new insight into the role of hepatic NGBR in insulin sensitivity and T2D treatment.
Collapse
Affiliation(s)
- Yi Chen
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Wenquan Hu
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Diabetes and Obesity Research Center, New York University Long Island School of Medicine, Mineola, New York, USA
| | - Qi Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Shiwei Zhao
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Dan Zhao
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Shuang Zhang
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Zhuo Wei
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xiaoxiao Yang
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoju Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Chenzhong Liao
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Qing Robert Miao
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Diabetes and Obesity Research Center, New York University Long Island School of Medicine, Mineola, New York, USA.
| | - Yajun Duan
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
16
|
Wang P, Mu X, Zhao H, Li Y, Wang L, Wolfe V, Cui SN, Wang X, Peng T, Zingarelli B, Wang C, Fan GC. Administration of GDF3 Into Septic Mice Improves Survival via Enhancing LXRα-Mediated Macrophage Phagocytosis. Front Immunol 2021; 12:647070. [PMID: 33679812 PMCID: PMC7925632 DOI: 10.3389/fimmu.2021.647070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/26/2021] [Indexed: 12/26/2022] Open
Abstract
The defective eradication of invading pathogens is a major cause of death in sepsis. As professional phagocytic cells, macrophages actively engulf/kill microorganisms and play essential roles in innate immune response against pathogens. Growth differentiation factor 3 (GDF3) was previously implicated as an important modulator of inflammatory response upon acute sterile injury. In this study, administration of recombinant GDF3 protein (rGDF3) either before or after CLP surgery remarkably improved mouse survival, along with significant reductions in bacterial load, plasma pro-inflammatory cytokine levels, and organ damage. Notably, our in vitro experiments revealed that rGDF3 treatment substantially promoted macrophage phagocytosis and intracellular killing of bacteria in a dose-dependent manner. Mechanistically, RNA-seq analysis results showed that CD5L, known to be regulated by liver X receptor α (LXRα), was the most significantly upregulated gene in rGDF3-treated macrophages. Furthermore, we observed that rGDF3 could promote LXRα nuclear translocation and thereby, augmented phagocytosis activity in macrophages, which was similar as LXRα agonist GW3965 did. By contrast, pre-treating macrophages with LXRα antagonist GSK2033 abolished beneficial effects of rGDF3 in macrophages. In addition, rGDF3 treatment failed to enhance bacteria uptake and killing in LXRα-knockout (KO) macrophages. Taken together, these results uncover that GDF3 may represent a novel mediator for controlling bacterial infection.
Collapse
Affiliation(s)
- Peng Wang
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Xingjiang Mu
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Hongyan Zhao
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Department of Critical Care Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yutian Li
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Lu Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Vivian Wolfe
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Shu-Nan Cui
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Department of Anesthesiology, Beijing Cancer Hospital, Peking University School of Oncology, Beijing, China
| | - Xiaohong Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Tianqing Peng
- The Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada
| | - Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Chunting Wang
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
17
|
Zhang S, Guo F, Yu M, Yang X, Yao Z, Li Q, Wei Z, Feng K, Zeng P, Zhao D, Li X, Zhu Y, Miao QR, Iwakiri Y, Chen Y, Han J, Duan Y. Reduced Nogo expression inhibits diet-induced metabolic disorders by regulating ChREBP and insulin activity. J Hepatol 2020; 73:1482-1495. [PMID: 32738448 DOI: 10.1016/j.jhep.2020.07.034] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/26/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND & AIMS Chronic overconsumption of a high-carbohydrate diet leads to steatosis and its associated metabolic disorder and, eventually, to non-alcoholic fatty liver disease. Carbohydrate-responsive element binding protein (ChREBP) and insulin regulate de novo lipogenesis from glucose. Herein, we studied the effect of reticulon-4 (Nogo) expression on diet-induced metabolic disorders in mice. METHODS Nogo-deficient (Nogo-/-) and littermate control [wild-type (WT)] mice were fed a high-glucose or high-fructose diet (HGD/HFrD) to induce metabolic disorders. The effects of Nogo small interfering (si) RNA (siRNA) on HFrD-induced metabolic disorders were investigated in C57BL/6J mice. RESULTS HGD/HFrD induced steatosis and its associated metabolic disorders in WT mice by activating ChREBP and impairing insulin sensitivity. They also activated Nogo-B expression, which in turn inhibited insulin activity. In response to HGD/HFrD feeding, Nogo deficiency enhanced insulin sensitivity and energy metabolism to reduce the expression of ChREBP and lipogenic molecules, activated AMP-activated catalytic subunit α, peroxisome proliferator activated receptor α and fibroblast growth factor 21, and reduced endoplasmic reticulum (ER) stress and inflammation, thereby blocking HGD/HFrD-induced hepatic lipid accumulation, insulin resistance and other metabolic disorders. Injection of Nogo siRNA protected C57BL/6J mice against HFrD-induced metabolic disorders by ameliorating insulin sensitivity, ChREBP activity, ER stress and inflammation. CONCLUSIONS Our study identified Nogo as an important mediator of insulin sensitivity and ChREBP activity. Reduction of Nogo expression is a potential strategy for the treatment of high-carbohydrate diet-induced metabolic complications. LAY SUMMARY Nogo deficiency blocks high-carbohydrate diet-induced glucose intolerance and insulin resistance, while increasing glucose/lipid utilisation and energy expenditure. Thus, reduction of Nogo expression protects against high-carbohydrate diet-induced body-weight gain, hepatic lipid accumulation and the associated metabolic disorders, indicating that approaches inhibiting Nogo expression can be applied for the treatment of diseases associated with metabolic disorders.
Collapse
Affiliation(s)
- Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China; College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Fangling Guo
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Miao Yu
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Zhi Yao
- Tianjin Medical University, Tianjin, China
| | - Qi Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Zhuo Wei
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Ke Feng
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Peng Zeng
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Dan Zhao
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xiaoju Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yan Zhu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Robert Miao
- Winthrop Hospital Diabetes and Obesity Research Center, New York University, New York, NY, USA
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China; College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China.
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
18
|
De Giorgi M, Jarrett KE, Burton JC, Doerfler AM, Hurley A, Li A, Hsu RH, Furgurson M, Patel KR, Han J, Borchers CH, Lagor WR. Depletion of essential isoprenoids and ER stress induction following acute liver-specific deletion of HMG-CoA reductase. J Lipid Res 2020; 61:1675-1686. [PMID: 33109681 PMCID: PMC7707164 DOI: 10.1194/jlr.ra120001006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
HMG-CoA reductase (Hmgcr) is the rate-limiting enzyme in the mevalonate pathway and is inhibited by statins. In addition to cholesterol, Hmgcr activity is also required for synthesizing nonsterol isoprenoids, such as dolichol, ubiquinone, and farnesylated and geranylgeranylated proteins. Here, we investigated the effects of Hmgcr inhibition on nonsterol isoprenoids in the liver. We have generated new genetic models to acutely delete genes in the mevalonate pathway in the liver using AAV-mediated delivery of Cre-recombinase (AAV-Cre) or CRISPR/Cas9 (AAV-CRISPR). The genetic deletion of Hmgcr by AAV-Cre resulted in extensive hepatocyte apoptosis and compensatory liver regeneration. At the biochemical level, we observed decreased levels of sterols and depletion of the nonsterol isoprenoids, dolichol and ubiquinone. At the cellular level, Hmgcr-null hepatocytes showed ER stress and impaired N-glycosylation. We further hypothesized that the depletion of dolichol, essential for N-glycosylation, could be responsible for ER stress. Using AAV-CRISPR, we somatically disrupted dehydrodolichyl diphosphate synthase subunit (Dhdds), encoding a branch point enzyme required for dolichol biosynthesis. Dhdds-null livers showed ER stress and impaired N-glycosylation, along with apoptosis and regeneration. Finally, the combined deletion of Hmgcr and Dhdds synergistically exacerbated hepatocyte ER stress. Our data show a critical role for mevalonate-derived dolichol in the liver and suggest that dolichol depletion is at least partially responsible for ER stress and apoptosis upon potent Hmgcr inhibition.
Collapse
Affiliation(s)
- Marco De Giorgi
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Kelsey E Jarrett
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA; Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Jason C Burton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA; Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Alexandria M Doerfler
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Ayrea Hurley
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Ang Li
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Rachel H Hsu
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Mia Furgurson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Kalyani R Patel
- Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Jun Han
- Genome British Columbia Proteomics Centre, University of Victoria, Victoria, British Columbia, Canada
| | - Christoph H Borchers
- Proteomics Centre, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, Quebec, Canada; Department of Data Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Skolkovo Innovation Center, Moscow, Russia
| | - William R Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
19
|
Zhang R, Tang BS, Guo JF. Research advances on neurite outgrowth inhibitor B receptor. J Cell Mol Med 2020; 24:7697-7705. [PMID: 32542927 PMCID: PMC7348171 DOI: 10.1111/jcmm.15391] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 12/24/2022] Open
Abstract
Neurite outgrowth inhibitor‐B (Nogo‐B) is a membrane protein which is extensively expressed in multiple organs, especially in endothelial cells and vascular smooth muscle cells of blood vessels and belongs to the reticulon protein family. Notably, its specific receptor, Nogo‐B receptor (NgBR), encoded by NUS1, has been implicated in many crucial cellular processes, such as cholesterol trafficking, lipid metabolism, dolichol synthesis, protein N‐glycosylation, vascular remodelling, angiogenesis, tumorigenesis and neurodevelopment. In recent years, accumulating studies have demonstrated the statistically significant changes of NgBR expression levels in human diseases, including Niemann‐Pick type C disease, fatty liver, congenital disorders of glycosylation, persistent pulmonary hypertension of the newborn, invasive ductal breast carcinoma, malignant melanoma, non‐small cell lung carcinoma, paediatric epilepsy and Parkinson's disease. Besides, both the in vitro and in vivo studies have shown that NgBR overexpression or knockdown contribute to the alteration of various pathophysiological processes. Thus, there is a broad development potential in therapeutic strategies by modifying the expression levels of NgBR.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing, China
| | - Ji-Feng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| |
Collapse
|
20
|
Tian Y, Yang B, Qiu W, Hao Y, Zhang Z, Yang B, Li N, Cheng S, Lin Z, Rui YC, Cheung OKW, Yang W, Wu WKK, Cheung YS, Lai PBS, Luo J, Sung JJY, Chen R, Wang HY, Cheng ASL, Yang P. ER-residential Nogo-B accelerates NAFLD-associated HCC mediated by metabolic reprogramming of oxLDL lipophagy. Nat Commun 2019; 10:3391. [PMID: 31358770 PMCID: PMC6662851 DOI: 10.1038/s41467-019-11274-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 06/18/2019] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome that elevates the risk of hepatocellular carcinoma (HCC). Although alteration of lipid metabolism has been increasingly recognized as a hallmark of cancer cells, the deregulated metabolic modulation of HCC cells in the NAFLD progression remains obscure. Here, we discovers an endoplasmic reticulum-residential protein, Nogo-B, as a highly expressed metabolic modulator in both murine and human NAFLD-associated HCCs, which accelerates high-fat, high-carbohydrate diet-induced metabolic dysfunction and tumorigenicity. Mechanistically, CD36-mediated oxLDL uptake triggers CEBPβ expression to directly upregulate Nogo-B, which interacts with ATG5 to promote lipophagy leading to lysophosphatidic acid-enhanced YAP oncogenic activity. This CD36-Nogo-B-YAP pathway consequently reprograms oxLDL metabolism and induces carcinogenetic signaling for NAFLD-associated HCCs. Targeting the Nogo-B pathway may represent a therapeutic strategy for HCC arising from the metabolic syndrome.
Collapse
Affiliation(s)
- Yuan Tian
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Bin Yang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Weinan Qiu
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yajing Hao
- University of Chinese Academy of Sciences, 100049, Beijing, China.,Key Laboratory of RNA Biology of CAS, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Zhenxing Zhang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Bo Yang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Nan Li
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Shuqun Cheng
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Zhangjun Lin
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yao-Cheng Rui
- Department of Pharmacology and School of Pharmacy, Second Military Medical University, 200433, Shanghai, China
| | - Otto K W Cheung
- School of Biomedical Sciences, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Weiqin Yang
- School of Biomedical Sciences, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - William K K Wu
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, 999077, Hong Kong, China.,Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Yue-Sun Cheung
- Department of Surgery, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Paul B S Lai
- Department of Surgery, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Jianjun Luo
- Key Laboratory of RNA Biology of CAS, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Joseph J Y Sung
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, 999077, Hong Kong, China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Runsheng Chen
- Key Laboratory of RNA Biology of CAS, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Hong-Yang Wang
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 200433, Shanghai, China.,National Center for Liver Cancer, Second Military Medical University, 200433, Shanghai, China
| | - Alfred S L Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, 999077, Hong Kong, China. .,State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, 999077, Hong Kong, China.
| | - Pengyuan Yang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China. .,University of Chinese Academy of Sciences, 100049, Beijing, China. .,National Center for Liver Cancer, Second Military Medical University, 200433, Shanghai, China.
| |
Collapse
|
21
|
Wang X, Zheng Y, Wu H, Tian S, Wu M, Luo P, Zhang F, Fang H, Li H, Xia Z. Transplantation of HUVECs with genetically modified Nogo-B accelerates wound-healing in nude mice. Am J Transl Res 2019; 11:2866-2876. [PMID: 31217860 PMCID: PMC6556635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/08/2019] [Indexed: 06/09/2023]
Abstract
Wound repair is an intractable problem in clinic, with limited treatment options. Previous studies have demonstrated the therapeutic potential of Nogo-B in tissue repairs. However, the therapeutic effect of Nogo-B in HUVEC is still unknown. In this study, we examined the benefit of genetically modified Human Umbilical Vein Endothelial Cells (HUVECs) and found that down regulation of Nogo-B significantly promoted secretion of growth factors involving in wound healing and greatly boosted the proliferation, migration of fibroblasts and epithelial cells. Moreover, using an excisional wound splinting model, we showed that injection of exogenous HUVEC-siNogo-B around the wound significantly enhanced angiogenesis and wound healing in nude mice. Thus, our data suggests that genetically modified HUVECs support microenvironment suitable for wound healing and systemically demonstrates the beneficial effect of HUVECs in wound healing.
Collapse
Affiliation(s)
- Xingtong Wang
- Department of Burns and Plastic Surgery, Sixth Medical Center of General Hospital, The People’s Liberation ArmyBeijing 100046, China
| | - Yongjun Zheng
- Burns Center of Changhai Hospital, The Second Military Medical UniversityShanghai 200433, China
| | - Haibin Wu
- Department of Burns and Plastic Surgery, General Hospital of Central Theater Command, The People’s Liberation ArmyWuhan 430070, China
| | - Song Tian
- Burns Center of Changhai Hospital, The Second Military Medical UniversityShanghai 200433, China
| | - Minjuan Wu
- Department of Histology and Embryology, The Second Military Medical UniversityShanghai 200433, China
| | - Pengfei Luo
- Burns Center of Changhai Hospital, The Second Military Medical UniversityShanghai 200433, China
| | - Fang Zhang
- Burns Center of Changhai Hospital, The Second Military Medical UniversityShanghai 200433, China
| | - He Fang
- Burns Center of Changhai Hospital, The Second Military Medical UniversityShanghai 200433, China
| | - Hengyu Li
- The Fourth Department of General Surgery, Changhai Hospital, Second Military Medical UniversityShanghai 200433, China
| | - Zhaofan Xia
- Burns Center of Changhai Hospital, The Second Military Medical UniversityShanghai 200433, China
| |
Collapse
|
22
|
Liu T, Wang X, Hu W, Fang Z, Jin Y, Fang X, Miao QR. Epigenetically Down-Regulated Acetyltransferase PCAF Increases the Resistance of Colorectal Cancer to 5-Fluorouracil. Neoplasia 2019; 21:557-570. [PMID: 31042625 PMCID: PMC6488821 DOI: 10.1016/j.neo.2019.03.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/22/2022] Open
Abstract
Only 10%–20% of colorectal cancer (CRC) patients observe effective responses to 5-fluorouracil (5-FU) based chemo-treatment. We used real-time PCR array and Western blot analysis to examine the expression alteration of acetyltransferases and deacetylases in 5-FU resistant CRC cell lines as compared to their respective parental CRC cell lines. Unlike other acetyltransferases and deacetylases, we found that the expression of acetyltransferase P300/CBP-associated factor (PCAF) is consistently decreased in three 5-FU resistant CRC cell lines. Similarly, knockdown of PCAF in HCT116 CRC parental cell line also increases the resistance to 5-FU and attenuates 5-FU-induced apoptosis. Mechanistically, we demonstrated that increased binding of trimethylated histone H3K27 in the promoter region of PCAF attenuated its transcription in 5-FU resistant HCT116/5-FU cells. Decreased PCAF impairs the acetylation of p53 and attenuates the p53-dependent transcription of p21, which results in the increased cyclin D1 and phosphorylation of Retinoblastoma 1. Conversely, overexpression of PCAF in CRC cell lines increases p21 and their susceptibility to 5-FU in vitro and in vivo. However, knockdown of p21 abolishes the beneficial effects of PCAF overexpression on increasing the sensitivity of HCT116/5-FU cells to 5-FU. Also, the reduced intensity of PCAF immunostaining was observed in the precancerous lesion, and microarray data from the public database further demonstrated the association between PCAF down-regulation and poor survival outcome. Our data suggest that PCAF-mediated p53 acetylation is an essential regulatory mechanism for increasing the susceptibility of CRC to 5-FU.
Collapse
Affiliation(s)
- Tong Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, 126 Xiantai Street, Changchun, Jilin 130033, China; Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; Divisions of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA
| | - Xiang Wang
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; Divisions of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; New York University Winthrop Hospital, Mineola, NY 11501
| | - Wenquan Hu
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; Divisions of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; New York University Winthrop Hospital, Mineola, NY 11501
| | - Zhi Fang
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; Divisions of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; New York University Winthrop Hospital, Mineola, NY 11501
| | - Ying Jin
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; Divisions of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, 126 Xiantai Street, Changchun, Jilin 130033, China.
| | - Qing Robert Miao
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; Divisions of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA; New York University Winthrop Hospital, Mineola, NY 11501.
| |
Collapse
|
23
|
Cai H, Saiyin H, Liu X, Han D, Ji G, Qin B, Zuo J, Shen S, Yu W, Wu J, Wu Y, Yu L. Nogo-B promotes tumor angiogenesis and provides a potential therapeutic target in hepatocellular carcinoma. Mol Oncol 2018; 12:2042-2054. [PMID: 30019429 PMCID: PMC6275258 DOI: 10.1002/1878-0261.12358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/27/2018] [Accepted: 07/01/2018] [Indexed: 12/11/2022] Open
Abstract
Tumor angiogenesis is one of the hallmarks of cancer as well as an attractive target for cancer therapy. Characterization of novel pathways that act in parallel with the VEGF/VEGFR axis to promote tumor angiogenesis may provide insights into novel anti-angiogenic therapeutic targets. We found that the expression level of Nogo-B is positively correlated with tumor vessel density in hepatocellular carcinoma (HCC). While Nogo-B depletion inhibited tumor angiogenesis, Nogo-B overexpression promoted tumor angiogenesis in a tumor xenograft subcutaneous model of the human HCC cell line. Mechanically, Nogo-B regulates tumor angiogenesis based on its association with integrin αv β3 and activation of focal adhesion kinase. Moreover, Nogo-B antibody successfully abolished the function of Nogo-B in tumor angiogenesis in vitro and in vivo. Collectively, our results strongly suggest that Nogo-B is an important tumor angiogenic factor and blocking Nogo-B selectively inhibits tumor angiogenesis.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Adhesion
- Cell Line, Tumor
- Female
- Gene Knockdown Techniques
- Humans
- Liver Neoplasms/blood supply
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Mice, Inbred C57BL
- Mice, Nude
- Molecular Targeted Therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Nogo Proteins/analysis
- Nogo Proteins/antagonists & inhibitors
- Nogo Proteins/genetics
- Nogo Proteins/metabolism
Collapse
Affiliation(s)
- Hao Cai
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
| | - Hexige Saiyin
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
| | - Xing Liu
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
- Present address:
Shanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Dingding Han
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
- Present address:
CAS Key Laboratory of Computational Biology320 Yueyang RoadShanghai200031China
| | - Guoqing Ji
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
| | - Bo Qin
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
| | - Jie Zuo
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
| | - Suqin Shen
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
| | - Wenbo Yu
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
| | - Jiaxue Wu
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
- Department of Liver SurgeryLiver Cancer InstituteZhongshan HospitalKey Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationFudan UniversityShanghaiChina
| | - Yanhua Wu
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
| | - Long Yu
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsSchool of Life SciencesFudan UniversityShanghaiChina
| |
Collapse
|
24
|
Ma C, Zhang W, Yang X, Liu Y, Liu L, Feng K, Zhang X, Yang S, Sun L, Yu M, Yang J, Li X, Hu W, Miao RQ, Zhu Y, Li L, Han J, Chen Y, Duan Y. Functional interplay between liver X receptor and AMP-activated protein kinase α inhibits atherosclerosis in apolipoprotein E-deficient mice - a new anti-atherogenic strategy. Br J Pharmacol 2018; 175:1486-1503. [PMID: 29394501 DOI: 10.1111/bph.14156] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE The liver X receptor (LXR) agonist T317 reduces atherosclerosis but induces fatty liver. Metformin activates energy metabolism by activating AMPKα. In this study, we determined if interactions between metformin and T317 could inhibit atherosclerosis without activation of hepatic lipogenesis. EXPERIMENTAL APPROACH Apolipoprotein E-deficient mice were treated with T317, metformin or both agents, in a high-fat diet for 16 weeks. Then, samples of aorta, liver, macrophage and serum were collected to determine atherosclerotic lesions, fatty liver, lipid profiles and expression of related proteins. Techniques used included immunohistochemistry, histology, qRT-PCR and Western blot. KEY RESULTS T317 inhibited en face and aortic root sinus lesions, and the inhibition was further enhanced by addition of metformin. Co-treatment with metformin and T317 increased lesion stability, by increasing collagen content, and reducing necrotic cores and calcification. Formation of macrophages/foam cells and their accumulation in arterial wall were inhibited by the co-treatment, which was accompanied by increased ABCA1/ABCG1 expression, reduced monocyte adhesion and apparent local proliferation of macrophages. Metformin blocked T317-induced fatty liver by inhibiting T317-induced hepatic LXRα nuclear translocation and expression of lipogenic genes and by activating AMPKα. Moreover, co-treatment with T317 and metformin improved triglyceride metabolism by inducing expression of adipose triglyceride lipase, hormone-sensitive lipase, PPARα and carnitine acetyltransferase and by inhibiting acyl-CoA:diacylglycerol acyltransferase 1 expression. CONCLUSIONS AND IMPLICATIONS Co-treatment with T317 and metformin inhibited the development of atherosclerosis without activation of lipogenesis, suggesting that combined treatment with T317 and metformin may be a novel approach to inhibition of atherosclerosis.
Collapse
Affiliation(s)
- Chuanrui Ma
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China.,College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Wenwen Zhang
- Research Institute of Obstetrics and Gynecology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Xiaoxiao Yang
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China
| | - Ying Liu
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Lipei Liu
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Ke Feng
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xiaomeng Zhang
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Shu Yang
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Lei Sun
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Miao Yu
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Jie Yang
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xiaoju Li
- College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Wenquan Hu
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert Q Miao
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yan Zhu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Luyuan Li
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Jihong Han
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China.,College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China.,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yuanli Chen
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China.,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yajun Duan
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China.,College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| |
Collapse
|
25
|
Liu Y, Wei Z, Ma X, Yang X, Chen Y, Sun L, Ma C, Miao QR, Hajjar DP, Han J, Duan Y. 25-Hydroxycholesterol activates the expression of cholesterol 25-hydroxylase in an LXR-dependent mechanism. J Lipid Res 2018; 59:439-451. [PMID: 29298812 DOI: 10.1194/jlr.m080440] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/06/2017] [Indexed: 12/13/2022] Open
Abstract
Cholesterol 25-hydroxylase (CH25H) catalyzes the production of 25-hydroxycholesterol (25-HC), an oxysterol that can play an important role in different biological processes. However, the mechanisms regulating CH25H expression have not been fully elucidated. In this study, we determined that CH25H is highly expressed in mouse liver and peritoneal macrophages. We identified several liver X receptor (LXR) response elements (LXREs) in the human CH25H promoter. In HepG2 cells, activation of LXR by 25-HC or other oxysterols and synthetic ligands [T0901317 (T317) and GW3965] induced CH25H protein expression, which was associated with increased CH25H mRNA expression. 25-HC or T317 activated CH25H transcription in an LXRE-dependent manner. Thus, high-expressing LXRα or LXRβ activated CH25H expression, and the activation was further enhanced by LXR ligands. In contrast, inhibition of LXRα/β expression attenuated 25-HC or T317-induced CH25H expression. Deficiency of interferon γ expression reduced, but did not block, LXR ligand-induced hepatic CH25H expression. Activation of LXR also substantially induced macrophage CH25H expression. In vivo, administration of GW3965 to mice increased CH25H expression in both liver and peritoneal macrophages. Taken together, our study demonstrates that 25-HC can activate CH25H expression in an LXR-dependent manner, which may be an important mechanism to exert the biological actions of 25-HC.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China.,Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China
| | - Zhuo Wei
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xingzhe Ma
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Xiaoxiao Yang
- Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Lei Sun
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Chuanrui Ma
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Qing R Miao
- Departments of Surgery and Pathology, Medical College of Wisconsin, Milwaukee, WI
| | - David P Hajjar
- Department of Pathology, Weill Cornell Medical College of Cornell University, New York, NY
| | - Jihong Han
- Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China .,College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yajun Duan
- Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China .,College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
26
|
Zhang W, Yang X, Chen Y, Hu W, Liu L, Zhang X, Liu M, Sun L, Liu Y, Yu M, Li X, Li L, Zhu Y, Miao QR, Han J, Duan Y. Activation of hepatic Nogo-B receptor expression-A new anti-liver steatosis mechanism of statins. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:177-190. [PMID: 29217477 DOI: 10.1016/j.bbalip.2017.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/27/2017] [Accepted: 12/01/2017] [Indexed: 12/20/2022]
Abstract
Deficiency of hepatic Nogo-B receptor (NgBR) expression activates liver X receptor α (LXRα) in an adenosine monophosphate-activated protein kinase α (AMPKα)-dependent manner, thereby inducing severe hepatic lipid accumulation and hypertriglyceridemia. Statins have been demonstrated non-cholesterol lowering effects including anti-nonalcoholic fatty liver disease (NAFLD). Herein, we investigated if the anti-NAFLD function of statins depends on activation of NgBR expression. In vivo, atorvastatin protected apoE deficient or NgBR floxed, but not hepatic NgBR deficient mice, against Western diet (WD)-increased triglyceride levels in liver and serum. In vitro, statins reduced lipid accumulation in nonsilencing small hairpin RNA-transfected (shNSi), but not in NgBR small hairpin RNA-transfected (shNgBRi) HepG2 cells. Inhibition of cellular lipid accumulation by atorvastatin is related to activation of AMPKα, and inactivation of LXRα and lipogenic genes. Statin also inhibited expression of oxysterol producing enzymes. Associated with changes of hepatic lipid levels by WD or atorvastatin, NgBR expression was inversely regulated. At cellular levels, statins increased NgBR mRNA and protein expression, and NgBR protein stability. In contrast to reduced cellular cholesterol levels by statin or β-cyclodextrin, increased cellular cholesterol levels decreased NgBR expression suggesting cholesterol or its synthesis intermediates inhibit NgBR expression. Indeed, mevalonate, geranylgeraniol or geranylgeranyl pyrophosphate, but not farnesyl pyrophosphate or farnesol, blocked atorvastatin-induced NgBR expression. Furthermore, we determined that induction of hepatic NgBR expression by atorvastatin mainly depended on inactivation of extracellular signal-regulated kinases 1/2 (ERK1/2) and protein kinase B (Akt). Taken together, our study demonstrates that statins inhibit NAFLD mainly through activation of NgBR expression.
Collapse
Affiliation(s)
- Wenwen Zhang
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China; Research Institute of Obstetrics and Gynecology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Xiaoxiao Yang
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China; Key Laboratory of Immuno Microenvironment and Disease, Ministry of Education, Tianjin Medical University, Tianjin, China; State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Wenquan Hu
- Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lipei Liu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaomeng Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Mengyang Liu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Lei Sun
- College of Life Sciences, Nankai University, Tianjin, China
| | - Ying Liu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Miao Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoju Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Luyuan Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yan Zhu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | | | - Jihong Han
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China; State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; College of Life Sciences, Nankai University, Tianjin, China.
| | - Yajun Duan
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China; State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
27
|
Long SL, Li YK, Xie YJ, Long ZF, Shi JF, Mo ZC. Neurite Outgrowth Inhibitor B Receptor: A Versatile Receptor with Multiple Functions and Actions. DNA Cell Biol 2017; 36:1142-1150. [PMID: 29058484 DOI: 10.1089/dna.2017.3813] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Members of the reticulon protein family are predominantly distributed within the endoplasmic reticulum. The neurite outgrowth inhibitor (Nogo) has three subtypes, including Nogo-A (200 kDa), Nogo-B (55 kDa), and Nogo-C (25 kDa). Nogo-A and Nogo-C are potent Nogos that are predominantly expressed in the central nervous system. Nogo-B, the splice variant of reticulon-4, is expressed widely in multiple human organ systems, including the liver, lung, kidney, blood vessels, and inflammatory cells. Moreover, the Nogo-B receptor (NgBR) can interact with Nogo-B and can independently affect nervous system regeneration, the chemotaxis of endothelial cells, proliferation, and apoptosis. In recent years, it has been demonstrated that NgBR plays an important role in human pathophysiological processes, including lipid metabolism, angiogenesis, N-glycosylation, cell apoptosis, chemoresistance in human hepatocellular carcinoma, and epithelial-mesenchymal transition. The pathophysiologic effects of NgBR have garnered increased attention, and the detection and enhancement of NgBR expression may be a novel approach to monitor the development and to improve the prognosis of relevant human clinical diseases.
Collapse
Affiliation(s)
- Shuang-Lian Long
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Yu-Kun Li
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Yuan-Jie Xie
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Zhi-Feng Long
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Jin-Feng Shi
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| | - Zhong-Cheng Mo
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China , Hengyang, China
| |
Collapse
|