1
|
Minervini G. Feature Paper in Oral Physiology and Pathology. Life (Basel) 2024; 14:895. [PMID: 39063647 PMCID: PMC11278310 DOI: 10.3390/life14070895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
In the realm of life sciences, the journal 'Life' has consistently served as a beacon for groundbreaking research and scientific discovery [...].
Collapse
Affiliation(s)
- Giuseppe Minervini
- Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, Tamil Nadu, India;
- Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania “Luigi Vanvitelli”, 80121 Naples, Italy
| |
Collapse
|
2
|
Hu CQ, Hou T, Xiang R, Li X, Li J, Wang TT, Liu WJ, Hou S, Wang D, Zhao QH, Yu XX, Xu M, Liu XK, Chi YJ, Yang JC. PANX1-mediated ATP release confers FAM3A's suppression effects on hepatic gluconeogenesis and lipogenesis. Mil Med Res 2024; 11:41. [PMID: 38937853 PMCID: PMC11210080 DOI: 10.1186/s40779-024-00543-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Extracellular adenosine triphosphate (ATP) is an important signal molecule. In previous studies, intensive research had revealed the crucial roles of family with sequence similarity 3 member A (FAM3A) in controlling hepatic glucolipid metabolism, islet β cell function, adipocyte differentiation, blood pressure, and other biological and pathophysiological processes. Although mitochondrial protein FAM3A plays crucial roles in the regulation of glucolipid metabolism via stimulating ATP release to activate P2 receptor pathways, its mechanism in promoting ATP release in hepatocytes remains unrevealed. METHODS db/db, high-fat diet (HFD)-fed, and global pannexin 1 (PANX1) knockout mice, as well as liver sections of individuals, were used in this study. Adenoviruses and adeno-associated viruses were utilized for in vivo gene overexpression or inhibition. To evaluate the metabolic status in mice, oral glucose tolerance test (OGTT), pyruvate tolerance test (PTT), insulin tolerance test (ITT), and magnetic resonance imaging (MRI) were conducted. Protein-protein interactions were determined by coimmunoprecipitation with mass spectrometry (MS) assays. RESULTS In livers of individuals and mice with steatosis, the expression of ATP-permeable channel PANX1 was increased (P < 0.01). Hepatic PANX1 overexpression ameliorated the dysregulated glucolipid metabolism in obese mice. Mice with hepatic PANX1 knockdown or global PANX1 knockout exhibited disturbed glucolipid metabolism. Restoration of hepatic PANX1 rescued the metabolic disorders of PANX1-deficient mice (P < 0.05). Mechanistically, ATP release is mediated by the PANX1-activated protein kinase B-forkhead box protein O1 (Akt-FOXO1) pathway to inhibit gluconeogenesis via P2Y receptors in hepatocytes. PANX1-mediated ATP release also activated calmodulin (CaM) (P < 0.01), which interacted with c-Jun N-terminal kinase (JNK) to inhibit its activity, thereby deactivating the transcription factor activator protein-1 (AP1) and repressing fatty acid synthase (FAS) expression and lipid synthesis (P < 0.05). FAM3A stimulated the expression of PANX1 via heat shock factor 1 (HSF1) in hepatocytes (P < 0.05). Notably, FAM3A overexpression failed to promote ATP release, inhibit the expression of gluconeogenic and lipogenic genes, and suppress gluconeogenesis and lipid deposition in PANX1-deficient hepatocytes and livers. CONCLUSIONS PANX1-mediated release of ATP plays a crucial role in maintaining hepatic glucolipid homeostasis, and it confers FAM3A's suppressive effects on hepatic gluconeogenesis and lipogenesis.
Collapse
Affiliation(s)
- Cheng-Qing Hu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
- Department of Obstetrics and Gynecology, Peking University Third Hospital/National Clinical Research Center for Obstetrics and Gynecology, Beijing, 100191, China
| | - Tao Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Rui Xiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Xin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Jing Li
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Tian-Tian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Wen-Jun Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Song Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Di Wang
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing, 100044, China
| | - Qing-He Zhao
- Department of Gastroenterology, Peking University People's Hospital, Beijing, 100044, China
| | - Xiao-Xing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Ming Xu
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital/Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Beijing, 100191, China
| | - Xing-Kai Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Centre, the First Hospital of Jilin University, Changchun, 130061, China.
| | - Yu-Jing Chi
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing, 100044, China.
- Department of Gastroenterology, Peking University People's Hospital, Beijing, 100044, China.
| | - Ji-Chun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences/State Key Laboratory of Vascular Homeostasis and Remodeling/Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing, 100191, China.
- Department of Cardiology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
3
|
Salagre D, Navarro-Alarcón M, Villalón-Mir M, Alcázar-Navarrete B, Gómez-Moreno G, Tamimi F, Agil A. Chronic melatonin treatment improves obesity by inducing uncoupling of skeletal muscle SERCA-SLN mediated by CaMKII/AMPK/PGC1α pathway and mitochondrial biogenesis in female and male Zücker diabetic fatty rats. Biomed Pharmacother 2024; 172:116314. [PMID: 38387135 DOI: 10.1016/j.biopha.2024.116314] [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: 12/18/2023] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024] Open
Abstract
Melatonin acute treatment limits obesity of young Zücker diabetic fatty (ZDF) rats by non-shivering thermogenesis (NST). We recently showed melatonin chronically increases the oxidative status of vastus lateralis (VL) in both obese and lean adult male animals. The identification of VL skeletal muscle-based NST by uncoupling of sarcoendoplasmic reticulum Ca2+-ATPase (SERCA)- sarcolipin (SLN) prompted us to investigate whether melatonin is a SERCA-SLN calcium futile cycle uncoupling and mitochondrial biogenesis enhancer. Obese ZDF rats and lean littermates (ZL) of both sexes were subdivided into two subgroups: control (C) and 12 weeks orally melatonin treated (M) (10 mg/kg/day). Compared to the control groups, melatonin decreased the body weight gain and visceral fat in ZDF rats of both sexes. Melatonin treatment in both sex obese rats restored the VL muscle skin temperature and sensitized the thermogenic effect of acute cold exposure. Moreover, melatonin not only raised SLN protein levels in the VL of obese and lean rats of both sexes; also, the SERCA activity. Melatonin treatment increased the SERCA2 expression in obese and lean rats (both sexes), with no effects on SERCA1 expression. Melatonin increased the expression of thermogenic genes and proteins (PGC1-α, PPARγ, and NRF1). Furthermore, melatonin treatment enhanced the expression ratio of P-CaMKII/CaMKII and P-AMPK/AMPK. In addition, it rose mitochondrial biogenesis. These results provided the initial evidence that chronic oral melatonin treatment triggers the CaMKII/AMPK/PGC1α axis by upregulating SERCA2-SLN-mediated NST in ZDF diabetic rats of both sexes. This may further contribute to the body weight control and metabolic benefits of melatonin.
Collapse
Affiliation(s)
- D Salagre
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, Granada 18016, Spain
| | - M Navarro-Alarcón
- Department of Nutrition and Bromatology, School of Pharmacy, University of Granada, Granada 18071, Spain
| | - M Villalón-Mir
- Department of Nutrition and Bromatology, School of Pharmacy, University of Granada, Granada 18071, Spain
| | - B Alcázar-Navarrete
- CIBERES, Carlos III Health Institute, Madrid, and Pulmonology Unit, Hospital Universitario Virgen de las Nieves, Granada 18014, Spain
| | - G Gómez-Moreno
- Department of Medically Compromised Patients in Dentistry, School of Dentistry, University of Granada, Granada 18011, Spain
| | - F Tamimi
- College of Dental Medicine, QU Health, Qatar University, Doha, Qatar
| | - A Agil
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, Granada 18016, Spain.
| |
Collapse
|
4
|
Liu TT, Xu HH, Liu ZJ, Zhang HP, Zhou HT, Zhu ZX, Wang ZQ, Xue JY, Li Q, Ma Y, You HJ, Luo DL. Downregulated calmodulin expression contributes to endothelial cell impairment in diabetes. Acta Pharmacol Sin 2023; 44:2492-2503. [PMID: 37468692 PMCID: PMC10692162 DOI: 10.1038/s41401-023-01127-1] [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: 02/05/2023] [Accepted: 06/11/2023] [Indexed: 07/21/2023] Open
Abstract
Endothelial dysfunction, a central hallmark of cardiovascular pathogenesis in diabetes mellitus, is characterized by impaired endothelial nitric oxide synthase (eNOS) and NO bioavailability. However, the underlying mechanisms remain unclear. Here in this study, we aimed to identify the role of calmodulin (CaM) in diabetic eNOS dysfunction. Human umbilical vein endothelial cells and murine endothelial progenitor cells (EPCs) treated with high glucose (HG) exhibited downregulated CaM mRNA/protein and vascular endothelial growth factor (VEGF) expression with impeded eNOS phosphorylation and cell migration/tube formation. These perturbations were reduplicated in CALM1-knockdown cells but prevented in CALM1-overexpressing cells. EPCs from type 2 diabetes animals behaved similarly to HG-treated normal EPCs, which could be rescued by CALM1-gene transduction. Consistently, diabetic animals displayed impaired eNOS phosphorylation, endothelium-dependent dilation, and CaM expression in the aorta, as well as deficient physical interaction of CaM and eNOS in the gastrocnemius. Local CALM1 gene delivery into a diabetic mouse ischemic hindlimb improved the blunted limb blood perfusion and gastrocnemius angiogenesis, and foot injuries. Diabetic patients showed insufficient foot microvascular autoregulation, eNOS phosphorylation, and NO production with downregulated CaM expression in the arterial endothelium, and abnormal CALM1 transcription in genome-wide sequencing analysis. Therefore, our findings demonstrated that downregulated CaM expression is responsible for endothelium dysfunction and angiogenesis impairment in diabetes, and provided a novel mechanism and target to protect against diabetic endothelial injury.
Collapse
Affiliation(s)
- Tian-Tian Liu
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Huan-Huan Xu
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Ze-Juan Liu
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - He-Ping Zhang
- Beijing Friendship Hospital, The Affiliated Hospital of Capital Medical University, Beijing, 100065, China
| | - Hai-Tao Zhou
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, and Peaking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Zhi-Xiang Zhu
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, and Peaking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Zhi-Qiang Wang
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Jing-Yi Xue
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Qiang Li
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Yi Ma
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Hong-Jie You
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Da-Li Luo
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
5
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
6
|
Schmidt U, Uluca B, Vokic I, Malik B, Kolbe T, Lassnig C, Holcmann M, Moreno-Viedma V, Robl B, Mühlberger C, Gotthardt D, Sibilia M, Rülicke T, Müller M, Csiszar A. Inducible overexpression of a FAM3C/ILEI transgene has pleiotropic effects with shortened life span, liver fibrosis and anemia in mice. PLoS One 2023; 18:e0286256. [PMID: 37713409 PMCID: PMC10503705 DOI: 10.1371/journal.pone.0286256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/11/2023] [Indexed: 09/17/2023] Open
Abstract
FAM3C/ILEI is an important factor in epithelial-to-mesenchymal transition (EMT) induction, tumor progression and metastasis. Overexpressed in many cancers, elevated ILEI levels and secretion correlate with poor patient survival. Although ILEI's causative role in invasive tumor growth and metastasis has been demonstrated in several cellular tumor models, there are no available transgenic mice to study these effects in the context of a complex organism. Here, we describe the generation and initial characterization of a Tet-ON inducible Fam3c/ILEI transgenic mouse strain. We find that ubiquitous induction of ILEI overexpression (R26-ILEIind) at weaning age leads to a shortened lifespan, reduced body weight and microcytic hypochromic anemia. The anemia was reversible at a young age within a week upon withdrawal of ILEI induction. Vav1-driven overexpression of the ILEIind transgene in all hematopoietic cells (Vav-ILEIind) did not render mice anemic or lower overall fitness, demonstrating that no intrinsic mechanisms of erythroid development were dysregulated by ILEI and that hematopoietic ILEI hyperfunction did not contribute to death. Reduced serum iron levels of R26-ILEIind mice were indicative for a malfunction in iron uptake or homeostasis. Accordingly, the liver, the main organ of iron metabolism, was severely affected in moribund ILEI overexpressing mice: increased alanine transaminase and aspartate aminotransferase levels indicated liver dysfunction, the liver was reduced in size, showed increased apoptosis, reduced cellular iron content, and had a fibrotic phenotype. These data indicate that high ILEI expression in the liver might reduce hepatoprotection and induce liver fibrosis, which leads to liver dysfunction, disturbed iron metabolism and eventually to death. Overall, we show here that the novel Tet-ON inducible Fam3c/ILEI transgenic mouse strain allows tissue specific timely controlled overexpression of ILEI and thus, will serve as a versatile tool to model the effect of elevated ILEI expression in diverse tissue entities and disease conditions, including cancer.
Collapse
Affiliation(s)
- Ulrike Schmidt
- IMP—Research Institute of Molecular Pathology, Vienna, Austria
| | - Betül Uluca
- IMP—Research Institute of Molecular Pathology, Vienna, Austria
| | - Iva Vokic
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Barizah Malik
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Kolbe
- Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria
- Department IFA-Tulln, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Caroline Lassnig
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Martin Holcmann
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | | | - Bernhard Robl
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Carina Mühlberger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dagmar Gotthardt
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Maria Sibilia
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Rülicke
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Agnes Csiszar
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Hu Y, Li J, Li X, Wang D, Xiang R, Liu W, Hou S, Zhao Q, Yu X, Xu M, Zhao D, Li T, Chi Y, Yang J. Hepatocyte-secreted FAM3D ameliorates hepatic steatosis by activating FPR1-hnRNP U-GR-SCAD pathway to enhance lipid oxidation. Metabolism 2023:155661. [PMID: 37454871 DOI: 10.1016/j.metabol.2023.155661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide; however, the underlying mechanisms remain poorly understood. FAM3D is a member of the FAM3 family; however, its role in hepatic glycolipid metabolism remains unknown. Serum FAM3D levels are positively correlated with fasting blood glucose levels in patients with diabetes. Hepatocytes express and secrete FAM3D, and its expression is increased in steatotic human and mouse livers. Hepatic FAM3D overexpression ameliorated hyperglycemia and steatosis in obese mice, whereas FAM3D-deficient mice exhibited exaggerated hyperglycemia and steatosis after high-fat diet (HFD)-feeding. In cultured hepatocytes, FAM3D overexpression or recombinant FAM3D protein (rFAM3D) treatment reduced gluconeogenesis and lipid deposition, which were blocked by anti-FAM3D antibodies or inhibition of its receptor, formyl peptide receptor 1 (FPR1). FPR1 overexpression suppressed gluconeogenesis and reduced lipid deposition in wild hepatocytes but not in FAM3D-deficient hepatocytes. The addition of rFAM3D restored FPR1's inhibitory effects on gluconeogenesis and lipid deposition in FAM3D-deficient hepatocytes. Hepatic FPR1 overexpression ameliorated hyperglycemia and steatosis in obese mice. RNA sequencing and DNA pull-down revealed that the FAM3D-FPR1 axis upregulated the expression of heterogeneous nuclear ribonucleoprotein U (hnRNP U), which recruits the glucocorticoid receptor (GR) to the promoter region of the short-chain acyl-CoA dehydrogenase (SCAD) gene, promoting its transcription to enhance lipid oxidation. Moreover, FAM3D-FPR1 axis also activates calmodulin-Akt pathway to suppress gluconeogenesis in hepatocytes. In conclusion, hepatocyte-secreted FAM3D activated the FPR1-hnRNP U-GR-SCAD pathway to enhance lipid oxidation in hepatocytes. Under obesity conditions, increased hepatic FAM3D expression is a compensatory mechanism against dysregulated glucose and lipid metabolism.
Collapse
Affiliation(s)
- Yuntao Hu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Jing Li
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100027, China
| | - Xin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Di Wang
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, China
| | - Rui Xiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Wenjun Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Song Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Qinghe Zhao
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
| | - Xiaoxing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Ming Xu
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Beijing 100191, China
| | - Dong Zhao
- Department of Endocrinology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China
| | - Tao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing 100044, China
| | - Yujing Chi
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, China; Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China.
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China; Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
8
|
Malik B, Vokic I, Mohr T, Poppelaars M, Holcmann M, Novoszel P, Timelthaler G, Lendl T, Krauss D, Elling U, Mildner M, Penninger JM, Petzelbauer P, Sibilia M, Csiszar A. FAM3C/ILEI protein is elevated in psoriatic lesions and triggers psoriasiform hyperproliferation in mice. EMBO Mol Med 2023; 15:e16758. [PMID: 37226685 PMCID: PMC10331587 DOI: 10.15252/emmm.202216758] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023] Open
Abstract
FAM3C/ILEI is an important cytokine for tumor progression and metastasis. However, its involvement in inflammation remains elusive. Here, we show that ILEI protein is highly expressed in psoriatic lesions. Inducible keratinocyte-specific ILEI overexpression in mice (K5-ILEIind ) recapitulates many aspects of psoriasis following TPA challenge, primarily manifested by impaired epidermal differentiation and increased neutrophil recruitment. Mechanistically, ILEI triggers Erk and Akt signaling, which then activates STAT3 via Ser727 phosphorylation. Keratinocyte-specific ILEI deletion ameliorates TPA-induced skin inflammation. A transcriptomic ILEI signature obtained from the K5-ILEIind model shows enrichment in several signaling pathways also found in psoriasis and identifies urokinase as a targetable enzyme to counteract ILEI activity. Pharmacological inhibition of urokinase in TPA-induced K5-ILEIind mice results in significant improvement of psoriasiform symptoms by reducing ILEI secretion. The ILEI signature distinguishes psoriasis from healthy skin with uPA ranking among the top "separator" genes. Our study identifies ILEI as a key driver in psoriasis, indicates the relevance of ILEI-regulated genes for disease manifestation, and shows the clinical impact of ILEI and urokinase as novel potential therapeutic targets in psoriasis.
Collapse
Affiliation(s)
- Barizah Malik
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
- Present address:
School of Biochemistry and Biotechnology, Quaid‐e‐Azam CampusUniversity of the PunjabLahorePakistan
| | - Iva Vokic
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Thomas Mohr
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
- Department of Analytical Chemistry, Faculty of ChemistryUniversity of ViennaViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University ViennaViennaAustria
| | - Marle Poppelaars
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Martin Holcmann
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Philipp Novoszel
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Gerald Timelthaler
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Thomas Lendl
- Research Institute of Molecular PathologyViennaAustria
| | - Dana Krauss
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Ulrich Elling
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA)ViennaAustria
| | - Michael Mildner
- Department of DermatologyMedical University of ViennaViennaAustria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA)ViennaAustria
- Department of Medical Genetics, Life Science InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | | | - Maria Sibilia
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| | - Agnes Csiszar
- Center for Cancer ResearchMedical University of Vienna, Comprehensive Cancer CenterViennaAustria
| |
Collapse
|
9
|
Moser C, Gosselé KA, Balaz M, Balazova L, Horvath C, Künzle P, Okreglicka KM, Li F, Blüher M, Stierstorfer B, Hess E, Lamla T, Hamilton B, Klein H, Neubauer H, Wolfrum C, Wolfrum S. FAM3D: A gut secreted protein and its potential in the regulation of glucose metabolism. Peptides 2023:171047. [PMID: 37328068 DOI: 10.1016/j.peptides.2023.171047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023]
Abstract
The number of diabetic patients is rising globally and concomitantly so do the diabetes associated complications. The gut secretes a variety of proteins to control blood glucose levels and/or food intake. As the drug class of GLP-1 agonists is based on a gut secreted peptide and the positive metabolic effects of bariatric surgery are at least partially mediated by gut peptides, we were interested in other gut secreted proteins which have yet to be explored. In this respect we identified the gut secreted protein FAM3D by analyzing sequencing data from L- and epithelial cells of VSG and sham operated as well as chow and HFD fed mice. FAM3D was overexpressed in diet induced obese mice via an adeno-associated virus (AAV), which resulted in a significant improvement of fasting blood glucose levels, glucose tolerance and insulin sensitivity. The liver lipid deposition was reduced, and the steatosis morphology was improved. Hyperinsulinemic clamps indicated that FAM3D is a global insulin sensitizer and increases glucose uptake into various tissues. In conclusion, the current study demonstrated that FAM3D controls blood glucose levels by acting as an insulin sensitizing protein and improves hepatic lipid deposition.
Collapse
Affiliation(s)
- Caroline Moser
- Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland
| | - Katherine A Gosselé
- Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland
| | - Miroslav Balaz
- Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland
| | - Lucia Balazova
- Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland
| | - Carla Horvath
- Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland
| | - Patricia Künzle
- Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland
| | - Katarzyna Maria Okreglicka
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| | - Fengqi Li
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| | - Matthias Blüher
- Medical Department III (Endocrinology, Nephrology and Rheumatology), University of Leipzig, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Birgit Stierstorfer
- Cardiometabolic Diseases Research Department, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach/Riss, Germany
| | - Eva Hess
- Cardiometabolic Diseases Research Department, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach/Riss, Germany
| | - Thorsten Lamla
- Cardiometabolic Diseases Research Department, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach/Riss, Germany
| | - Bradford Hamilton
- Cardiometabolic Diseases Research Department, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach/Riss, Germany
| | - Holger Klein
- Global Computational Biology and Digital Sciences Department, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach/Riss, Germany
| | - Heike Neubauer
- Cardiometabolic Diseases Research Department, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach/Riss, Germany
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland.
| | - Susanne Wolfrum
- Laboratory of Organic Chemistry, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
10
|
Li Y, Lin S, Xu X, Jin W, Su Y, Yuan F, Zhang Y, Li Z, Zhou Y, Zhu L, Zhang L. Skeletal muscle HSF1 prevents insulin resistance by improving glucose utilization. FASEB J 2022; 36:e22667. [PMID: 36421020 DOI: 10.1096/fj.202201160rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022]
Abstract
The regulation of muscle glucose utilization has significant potential for the treatment of type 2 diabetes mellitus (T2DM) and obesity. Heat shock factor 1 (HSF1) is involved in cellular metabolism and regulation of muscle metabolism. However, it is unclear how HSF1 regulates muscle glucose metabolism. In the present study, the development of obesity in mice was associated with HSF1 downregulation. Serum samples and muscle biopsies were obtained from obese and healthy humans. Fasting glucose and insulin levels and the homeostasis model assessment of insulin resistance value showed that obesity was associated with insulin resistance. The skeletal muscle level of HSF1 was decreased in obese and ob/ob mice. HSF1 was selectively over-expressed in the skeletal muscles of high fat diet (HFD)-fed mice. Muscle HSF1 over-expression successfully triggered glycolytic-to-oxidative myofiber switch and increased fatty acid metabolism and insulin sensitivity in the skeletal muscles of HFD-fed mice. Moreover, HSF1 improved energy expenditure and blocked muscle accumulation of triglycerides in HFD-fed mice. Consequently, muscle HSF1 mitigated the impaired muscle insulin signaling and insulin resistance in HFD-fed mice. In conclusion, T2DM and obesity in HFD-fed mice may be treated with selective HSF1-directed programming of exercise-like effects in skeletal muscle. These findings may aid the development of a new therapeutic approach for obesity and T2DM.
Collapse
Affiliation(s)
- Yun Li
- Department of Pediatric Laboratory, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Shibo Lin
- Department of Bariatric and Metabolic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xu Xu
- Department of Pediatrics, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Weilai Jin
- Department of Pediatric Laboratory, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Yinglin Su
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Fuqiang Yuan
- Department of Pediatric Laboratory, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Yiting Zhang
- Department of Pediatric Laboratory, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Zhengying Li
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Yahui Zhou
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Lihong Zhu
- Department of Pediatrics, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Le Zhang
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
11
|
Nakano M, Imamura R, Sugi T, Nishimura M. Human FAM3C restores memory-based thermotaxis of Caenorhabditis elegans famp-1/m70.4 loss-of-function mutants. PNAS NEXUS 2022; 1:pgac242. [PMID: 36712359 PMCID: PMC9802357 DOI: 10.1093/pnasnexus/pgac242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 10/21/2022] [Indexed: 06/18/2023]
Abstract
The family with sequence similarity 3 (FAM3) superfamily represents a distinct class of signaling molecules that share a characteristic structural feature. Mammalian FAM3 member C (FAM3C) is abundantly expressed in neuronal cells and released from the synaptic vesicle to the extracellular milieu in an activity-dependent manner. However, the neural function of FAM3C has yet to be fully clarified. We found that the protein sequence of human FAM3C is similar to that of the N-terminal tandem domains of Caenorhabditis elegans FAMP-1 (formerly named M70.4), which has been recognized as a tentative ortholog of mammalian FAM3 members or protein-O-mannose β-1,2-N-acetylglucosaminyltransferase 1 (POMGnT1). Missense mutations in the N-terminal domain, named Fam3L2, caused defects in memory-based thermotaxis but not in chemotaxis behaviors; these defects could be restored by AFD neuron-specific exogenous expression of a polypeptide corresponding to the Fam3L2 domain but not that corresponding to the Fam3L1. Moreover, human FAM3C could also rescue defective thermotaxis behavior in famp-1 mutant worms. An in vitro assay revealed that the Fam3L2 and FAM3C can bind with carbohydrates, similar to the stem domain of POMGnT1. The athermotactic mutations in the Fam3L2 domain caused a partial loss-of-function of FAMP-1, whereas the C-terminal truncation mutations led to more severe neural dysfunction that reduced locomotor activity. Overall, we show that the Fam3L2 domain-dependent function of FAMP-1 in AFD neurons is required for the thermotaxis migration of C. elegans and that human FAM3C can act as a substitute for the Fam3L2 domain in thermotaxis behaviors.
Collapse
Affiliation(s)
- Masaki Nakano
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta-Tsukinowa, Otsu, Shiga 520-2192, Japan
| | - Ryuki Imamura
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-0046, Japan
| | | | | |
Collapse
|
12
|
Bosi E, Marselli L, Suleiman M, Tesi M, De Luca C, Del Guerra S, Cnop M, Eizirik D, Marchetti P. A single-cell human islet interactome atlas identifies disrupted autocrine and paracrine communications in type 2 diabetes. NAR Genom Bioinform 2022; 4:lqac084. [DOI: 10.1093/nargab/lqac084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 10/04/2022] [Accepted: 10/29/2022] [Indexed: 11/19/2022] Open
Abstract
Abstract
A sensible control of hormone secretion from pancreatic islets requires concerted inter-cellular communications, but a comprehensive picture of the whole islet interactome is presently missing. Single-cell transcriptomics allows to overcome this and we used here a single-cell dataset from type 2 diabetic (T2D) and non-diabetic (ND) donors to leverage islet interaction networks. The single-cell dataset contains 3046 cells classified in 7 cell types. The interactions across cell types in T2D and ND were obtained and resulting networks analysed to identify high-centrality genes and altered interactions in T2D. The T2D interactome displayed a higher number of interactions (10 787) than ND (9707); 1289 interactions involved beta cells (1147 in ND). High-centrality genes included EGFR, FGFR1 and FGFR2, important for cell survival and proliferation. In conclusion, this analysis represents the first in silico model of the human islet interactome, enabling the identification of signatures potentially relevant for T2D pathophysiology.
Collapse
Affiliation(s)
- Emanuele Bosi
- Department of Experimental and Clinical Medicine, Pancreatic islets laboratory, University of Pisa , Pisa , Italy
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa , Genoa , Italy
| | - Lorella Marselli
- Department of Experimental and Clinical Medicine, Pancreatic islets laboratory, University of Pisa , Pisa , Italy
| | - Mara Suleiman
- Department of Experimental and Clinical Medicine, Pancreatic islets laboratory, University of Pisa , Pisa , Italy
| | - Marta Tesi
- Department of Experimental and Clinical Medicine, Pancreatic islets laboratory, University of Pisa , Pisa , Italy
| | - Carmela De Luca
- Department of Experimental and Clinical Medicine, Pancreatic islets laboratory, University of Pisa , Pisa , Italy
| | - Silvia Del Guerra
- Department of Experimental and Clinical Medicine, Pancreatic islets laboratory, University of Pisa , Pisa , Italy
| | - Miriam Cnop
- ULB Center for Diabetes Research , Université Libre de Bruxelles, Brussels , Belgium
- Division of Endocrinology, Erasmus Hospital , Université Libre de Bruxelles, Brussels , Belgium
| | - Decio L Eizirik
- ULB Center for Diabetes Research , Université Libre de Bruxelles, Brussels , Belgium
| | - Piero Marchetti
- Department of Experimental and Clinical Medicine, Pancreatic islets laboratory, University of Pisa , Pisa , Italy
| |
Collapse
|
13
|
Ruozi G, Bortolotti F, Mura A, Tomczyk M, Falcione A, Martinelli V, Vodret S, Braga L, Dal Ferro M, Cannatà A, Zentilin L, Sinagra G, Zacchigna S, Giacca M. Cardioprotective factors against myocardial infarction selected in vivo from an AAV secretome library. Sci Transl Med 2022; 14:eabo0699. [PMID: 36044596 DOI: 10.1126/scitranslmed.abo0699] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Therapies for patients with myocardial infarction and heart failure are urgently needed, in light of the breadth of these conditions and lack of curative treatments. To systematically identify previously unidentified cardioactive biologicals in an unbiased manner in vivo, we developed cardiac FunSel, a method for the systematic, functional selection of effective factors using a library of 1198 barcoded adeno-associated virus (AAV) vectors encoding for the mouse secretome. By pooled vector injection into the heart, this library was screened to functionally select for factors that confer cardioprotection against myocardial infarction. After two rounds of iterative selection in mice, cardiac FunSel identified three proteins [chordin-like 1 (Chrdl1), family with sequence similarity 3 member C (Fam3c), and Fam3b] that preserve cardiomyocyte viability, sustain cardiac function, and prevent pathological remodeling. In particular, Chrdl1 exerted its protective activity by binding and inhibiting extracellular bone morphogenetic protein 4 (BMP4), which resulted in protection against cardiomyocyte death and induction of autophagy in cardiomyocytes after myocardial infarction. Chrdl1 also inhibited fibrosis and maladaptive cardiac remodeling by binding transforming growth factor-β (TGF-β) and preventing cardiac fibroblast differentiation into myofibroblasts. Production of secreted and circulating Chrdl1, Fam3c, and Fam3b from the liver also protected the heart from myocardial infarction, thus supporting the use of the three proteins as recombinant factors. Together, these findings disclose a powerful method for the in vivo, unbiased selection of tissue-protective factors and describe potential cardiac therapeutics.
Collapse
Affiliation(s)
- Giulia Ruozi
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Francesca Bortolotti
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,Cardiovascular Department, ASUGI, 34149 Trieste, Italy
| | - Antonio Mura
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Mateusz Tomczyk
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, UK
| | - Antonella Falcione
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Valentina Martinelli
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Simone Vodret
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Luca Braga
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, UK
| | | | - Antonio Cannatà
- Cardiovascular Department, ASUGI, 34149 Trieste, Italy.,British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, UK
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, ASUGI, 34149 Trieste, Italy.,Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Serena Zacchigna
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34139 Trieste, Italy.,British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, UK.,Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| |
Collapse
|
14
|
Yao M, Fu L, Liu X, Zheng D. In-Silico Multi-Omics Analysis of the Functional Significance of Calmodulin 1 in Multiple Cancers. Front Genet 2022; 12:793508. [PMID: 35096010 PMCID: PMC8790318 DOI: 10.3389/fgene.2021.793508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/23/2021] [Indexed: 01/14/2023] Open
Abstract
Aberrant activation of calmodulin 1 (CALM1) has been reported in human cancers. However, comprehensive understanding of the role of CALM1 in most cancer types has remained unclear. We systematically analyzed the expression landscape, DNA methylation, gene alteration, immune infiltration, clinical relevance, and molecular pathway of CALM1 in multiple cancers using various online tools, including The Cancer Genome Atlas, cBioPortal and the Human Protein Atlas databases. Kaplan–Meier and receiver operating characteristic (ROC) curves were plotted to explore the prognostic and diagnostic potential of CALM1 expression. Multivariate analyses were used to evaluate whether the CALM1 expression could be an independent risk factor. A nomogram predicting the overall survival (OS) of patients was developed, evaluated, and compared with the traditional Tumor-Node-Metastasis (TNM) model using decision curve analysis. R language was employed as the main tool for analysis and visualization. Results revealed CALM1 to be highly expressed in most cancers, its expression being regulated by DNA methylation in multiple cancers. CALM1 had a low mutation frequency (within 3%) and was associated with immune infiltration. We observed a substantial positive correlation between CALM1 expression and macrophage and neutrophil infiltration levels in multiple cancers. Different mutational forms of CALM1 hampered immune cell infiltration. Additionally, CALM1 expression had high diagnostic and prognostic potential. Multivariate analyses revealed CALM1 expression to be an independent risk factor for OS. Therefore, our newly developed nomogram had a higher clinical value than the TNM model. The concordance index, calibration curve, and time-dependent ROC curves of the nomogram exhibited excellent performance in terms of predicting the survival rate of patients. Moreover, elevated CALM1 expression contributes to the activation of cancer-related pathways, such as the WNT and MAPK pathways. Overall, our findings improved our understanding of the function of CALM1 in human cancers.
Collapse
Affiliation(s)
- Maolin Yao
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Lanyi Fu
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Xuedong Liu
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Dong Zheng
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| |
Collapse
|
15
|
Zhu Y, Pu Z, Wang G, Li Y, Wang Y, Li N, Peng F. FAM3C: an emerging biomarker and potential therapeutic target for cancer. Biomark Med 2021; 15:373-384. [PMID: 33666514 DOI: 10.2217/bmm-2020-0179] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
FAM3C is a member of the FAM3 family. Recently, overexpression of FAM3C has been reported in numerous types of cancer, including breast and colon cancer. Increasing evidence suggests that elevated FAM3C and its altered subcellular localization are closely associated with tumor formation, invasion, metastasis and poor survival. Moreover, FAM3C has been found to be the regulator of various proteins that associate with cancer, including Ras, STAT3, TGF-β and LIFR. This review summarizes the current knowledge regarding FAM3C, including its structure, expression patterns, regulation, physiological roles and regulatory functions in various malignancies. These findings highlight the importance of FAM3C in cancer development and provide evidence that FAM3C is a novel biomarker and potential therapeutic target for various cancers.
Collapse
Affiliation(s)
- Yuanyuan Zhu
- Department of Blood Transfusion, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China.,NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
| | - Zhangya Pu
- Department of Infectious Diseases & Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
| | - Guoqiang Wang
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
| | - Yubin Li
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
| | - Yinmiao Wang
- Department of Blood Transfusion, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China.,NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
| | - Ning Li
- Department of Blood Transfusion, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
| | - Fang Peng
- Department of Blood Transfusion, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China.,NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
| |
Collapse
|
16
|
Jurivich DA, Manocha GD, Trivedi R, Lizakowski M, Rakoczy S, Brown-Borg H. Multifactorial Attenuation of the Murine Heat Shock Response With Age. J Gerontol A Biol Sci Med Sci 2021; 75:1846-1852. [PMID: 31612204 DOI: 10.1093/gerona/glz204] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Indexed: 01/08/2023] Open
Abstract
Age-dependent perturbation of the cellular stress response affects proteostasis and other key functions relevant to cellular action and survival. Central to age-related changes in the stress response is loss of heat shock factor 1 (HSF1)-DNA binding and transactivation properties. This report elucidates how age alters different checkpoints of HSF1 activation related to posttranslational modification and protein interactions. When comparing liver extracts from middle aged (12 M) and old (24 M) mice, significant differences are found in HSF1 phosphorylation and acetylation. HSF1 protein levels and messenger RNA decline with age, but its protein levels are stress-inducible and exempt from age-dependent changes. This surprising adaptive change in the stress response has additional implications for aging and chronic physiological stress that might explain an age-dependent dichotomy of HSF1 protein levels that are low in neurodegeneration and elevated in cancer.
Collapse
Affiliation(s)
- Donald A Jurivich
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Gunjan D Manocha
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Rachana Trivedi
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Mary Lizakowski
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Sharlene Rakoczy
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Holly Brown-Borg
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| |
Collapse
|
17
|
Song Q, Song J, Li C, Wang Y, Qi L, Wang H. Genetic variants in the FAM3C gene are associated with lipid traits in Chinese children. Pediatr Res 2021; 89:673-678. [PMID: 32316026 DOI: 10.1038/s41390-020-0897-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/12/2020] [Accepted: 04/01/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Previous studies have related FAM3C gene with childhood bone health, and the regulation of lipid metabolism in hepatocytes. The present case-control study aimed to analyze the association of FAM3C genetic variants with overweight/obesity and lipid traits among Chinese children. METHODS Two genetic variants (rs7776725 and rs7793554) within the FAM3C gene were genotyped in 3305 Chinese children aged 6-18 years. RESULTS In the whole study population, the T-allele of rs7776725 and A-allele of rs7793554 within the FAM3C gene were associated with 40.2% (95% CI: 11.6-76.1%; P = 0.004) and 29.1% (6.9-56.0%; P = 0.008) increased risk of dyslipidemia, higher triglyceride (P = 0.014 and P = 0.001) and lower HDL-C (P = 0.015 and P = 0.003). In addition, we found that rs7776725 interacted with sex on dyslipidemia (Pfor interaction = 0.004), and sex-stratified analyses showed that it was significantly associated with dyslipidemia only in girls (P = 8.78 × 10-5). The variant also showed nominally significant interactions with sex on total cholesterol and LDL-C (Pfor interaction = 0.012 and 0.008). CONCLUSION We found that FAM3C genetic variants were associated with dyslipidemia and lipid traits among Chinese children. In addition, we found significant gene-by-sex interactions. Our findings provided evidence supporting the role of FAM3C gene in regulating lipid metabolism in humans. IMPACT FAM3C genetic variants were associated with dyslipidemia and lipid traits among Chinese children. In addition, we found significant gene-by-sex interactions. FAM3C/rs7776725 was associated with dyslipidemia and lipid traits only in girls. Our findings provided evidence supporting the role of FAM3C gene in regulating lipid metabolism in humans.
Collapse
Affiliation(s)
- Qiying Song
- Department of Maternal and Child Health, School of Public Health, Peking University, 100191, Beijing, China
| | - Jieyun Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, 100191, Beijing, China
| | - Chenxiong Li
- Department of Maternal and Child Health, School of Public Health, Peking University, 100191, Beijing, China
| | - Yang Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, 100191, Beijing, China
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Haijun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, 100191, Beijing, China.
| |
Collapse
|
18
|
Nakano M, Mitsuishi Y, Liu L, Watanabe N, Hibino E, Hata S, Saito T, Saido TC, Murayama S, Kasuga K, Ikeuchi T, Suzuki T, Nishimura M. Extracellular Release of ILEI/FAM3C and Amyloid-β Is Associated with the Activation of Distinct Synapse Subpopulations. J Alzheimers Dis 2021; 80:159-174. [PMID: 33492290 DOI: 10.3233/jad-201174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Brain amyloid-β (Aβ) peptide is released into the interstitial fluid (ISF) in a neuronal activity-dependent manner, and Aβ deposition in Alzheimer's disease (AD) is linked to baseline neuronal activity. Although the intrinsic mechanism for Aβ generation remains to be elucidated, interleukin-like epithelial-mesenchymal transition inducer (ILEI) is a candidate for an endogenous Aβ suppressor. OBJECTIVE This study aimed to access the mechanism underlying ILEI secretion and its effect on Aβ production in the brain. METHODS ILEI and Aβ levels in the cerebral cortex were monitored using a newly developed ILEI-specific ELISA and in vivo microdialysis in mutant human Aβ precursor protein-knockin mice. ILEI levels in autopsied brains and cerebrospinal fluid (CSF) were measured using ELISA. RESULTS Extracellular release of ILEI and Aβ was dependent on neuronal activation and specifically on tetanus toxin-sensitive exocytosis of synaptic vesicles. However, simultaneous monitoring of extracellular ILEI and Aβ revealed that a spontaneous fluctuation of ILEI levels appeared to inversely mirror that of Aβ levels. Selective activation and inhibition of synaptic receptors differentially altered these levels. The evoked activation of AMPA-type receptors resulted in opposing changes to ILEI and Aβ levels. Brain ILEI levels were selectively decreased in AD. CSF ILEI concentration correlated with that of Aβ and were reduced in AD and mild cognitive impairment. CONCLUSION ILEI and Aβ are released from distinct subpopulations of synaptic terminals in an activity-dependent manner, and ILEI negatively regulates Aβ production in specific synapse types. CSF ILEI might represent a surrogate marker for the accumulation of brain Aβ.
Collapse
Affiliation(s)
- Masaki Nakano
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Yachiyo Mitsuishi
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Lei Liu
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Naoki Watanabe
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Emi Hibino
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan.,Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan.,Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| | - Shigeo Murayama
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Hokkaido, Japan
| | - Masaki Nishimura
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
19
|
Zappaterra M, Zambonelli P, Schivazappa C, Simoncini N, Virgili R, Stefanon B, Davoli R. Investigating the Features of PDO Green Hams during Salting: Insights for New Markers and Genomic Regions in Commercial Hybrid Pigs. Animals (Basel) 2021; 11:E68. [PMID: 33401485 PMCID: PMC7823679 DOI: 10.3390/ani11010068] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022] Open
Abstract
Protected Designation of Origin (PDO) dry-cured hams production is greatly dependent on raw meat quality. This study was performed to identify genetic markers associated with the quality of dry-cured ham. Carcass traits of 229 heavy pigs belonging to three commercial genetic lines were registered (weight, EUROP classification). Phenotypic traits (Semimembranosus muscle ultimate pH, ham weight and lean meat content, adsorbed salt) of the corresponding thighs, undergone PDO ham process in three different plants, were measured, using a fast and non-invasive technology. Green ham weight and lean meat percentage influenced the estimated salt content and the weight loss during salting, even if the processing plant greatly affected the variability of the measured ham traits. The genomic data were obtained with the GeneSeek Genomic Profiler (GGP) 70k HD Porcine Array, using the slaughter day and the sex of the animals in the statistical analyses. The phenotypic traits were associated with the genotypes through GenAbel software. The results showed that 18 SNPs located on nine porcine chromosomes were found to be associated with nine phenotypic traits, mainly related to ham weight loss during salting. New associations were found between markers in the genes Neural Precursor Cell Expressed Developmentally Down-Regulated 9 (NEDD9, SSC7), T-Cell Lymphoma Invasion and Metastasis 2 (TIAM2, SSC1), and the ham quality traits. After validation, these SNPs may be useful to improve the quality of thighs for the production of PDO dry-cured hams.
Collapse
Affiliation(s)
- Martina Zappaterra
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Viale Fanin 46, I-40127 Bologna, Italy;
| | - Paolo Zambonelli
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Viale Fanin 46, I-40127 Bologna, Italy;
| | - Cristina Schivazappa
- Stazione Sperimentale per l’Industria delle Conserve Alimentari (SSICA), Viale Faustino Tanara 31/A, I-43121 Parma, Italy; (C.S.); (N.S.); (R.V.)
| | - Nicoletta Simoncini
- Stazione Sperimentale per l’Industria delle Conserve Alimentari (SSICA), Viale Faustino Tanara 31/A, I-43121 Parma, Italy; (C.S.); (N.S.); (R.V.)
| | - Roberta Virgili
- Stazione Sperimentale per l’Industria delle Conserve Alimentari (SSICA), Viale Faustino Tanara 31/A, I-43121 Parma, Italy; (C.S.); (N.S.); (R.V.)
| | - Bruno Stefanon
- Department of Agrifood, Environmental and Animal Science, University of Udine, Via delle Scienze 208, I-33100 Udine, Italy;
| | - Roberta Davoli
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Viale Fanin 46, I-40127 Bologna, Italy;
| |
Collapse
|
20
|
Gagné-Ouellet V, Breton E, Thibeault K, Fortin CA, Desgagné V, Girard Tremblay É, Cardenas A, Guérin R, Perron P, Hivert MF, Bouchard L. Placental Epigenome-Wide Association Study Identified Loci Associated with Childhood Adiposity at 3 Years of Age. Int J Mol Sci 2020; 21:ijms21197201. [PMID: 33003475 PMCID: PMC7582906 DOI: 10.3390/ijms21197201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/22/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to identify placental DNA methylation (DNAm) variations associated with adiposity at 3 years of age. We quantified placental DNAm using the Infinium MethylationEPIC BeadChips. We assessed associations between DNAm at single-CpGs and skinfold thickness using robust linear regression models adjusted for gestational age, child's sex, age at follow-up and cellular heterogeneity. We sought replication of DNAm association with child adiposity in an independent cohort. We quantified placental mRNA levels for annotated gene using qRT-PCR and tested for correlation with DNAm. Lower DNAm at cg22593959 and cg22436429 was associated with higher adiposity (β = -1.18, q = 0.002 and β = -0.82, q = 0.04). The cg22593959 is located in an intergenic region (chr7q31.3), whereas cg22436429 is within the TFAP2E gene (1p34.3). DNAm at cg22593959 and cg22436429 was correlated with mRNA levels at FAM3C (rs = -0.279, p = 0.005) and TFAP2E (rs = 0.216, p = 0.03). In an independent cohort, the association between placental DNAm at cg22593959 and childhood adiposity was of similar strength and direction (β = -3.8 ± 4.1, p = 0.36), yet non-significant. Four genomic regions were also associated with skinfold thickness within FMN1, MAGI2, SKAP2 and BMPR1B genes. We identified placental epigenetic variations associated with adiposity at 3 years of age suggesting that childhood fat accretion patterns might be established during fetal life.
Collapse
Affiliation(s)
- Valérie Gagné-Ouellet
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Edith Breton
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Kathrine Thibeault
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Carol-Ann Fortin
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Véronique Desgagné
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
- Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean—Hôpital Universitaire de Chicoutimi, Saguenay, QC G7H 5H6, Canada
| | - Élise Girard Tremblay
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
| | - Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720-7360, USA;
| | - Renée Guérin
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
- Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean—Hôpital Universitaire de Chicoutimi, Saguenay, QC G7H 5H6, Canada
| | - Patrice Perron
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (P.P.); (M.-F.H.)
- Centre de Recherche du CHUS, Sherbrooke, QC J1H 5N4, Canada
| | - Marie-France Hivert
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (P.P.); (M.-F.H.)
- Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Luigi Bouchard
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (V.G.-O.); (E.B.); (K.T.); (C.-A.F.); (V.D.); (É.G.T.); (R.G.)
- Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean—Hôpital Universitaire de Chicoutimi, Saguenay, QC G7H 5H6, Canada
- Centre de Recherche du CHUS, Sherbrooke, QC J1H 5N4, Canada
- Correspondence:
| |
Collapse
|
21
|
Chen Z, Liu X, Luo Y, Wang J, Meng Y, Sun L, Chang Y, Cui Q, Yang J. Repurposing Doxepin to Ameliorate Steatosis and Hyperglycemia by Activating FAM3A Signaling Pathway. Diabetes 2020; 69:1126-1139. [PMID: 32312868 PMCID: PMC7243289 DOI: 10.2337/db19-1038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/13/2020] [Indexed: 01/07/2023]
Abstract
Mitochondrial protein FAM3A suppresses hepatic gluconeogenesis and lipogenesis. This study aimed to screen drug(s) that activates FAM3A expression and evaluate its effect(s) on hyperglycemia and steatosis. Drug-repurposing methodology predicted that antidepressive drug doxepin was among the drugs that potentially activated FAM3A expression. Doxepin was further validated to stimulate the translocation of transcription factor HNF4α from the cytoplasm into the nucleus, where it promoted FAM3A transcription to enhance ATP synthesis, suppress gluconeogenesis, and reduce lipid deposition in hepatocytes. HNF4α antagonism or FAM3A deficiency blunted doxepin-induced suppression on gluconeogenesis and lipid deposition in hepatocytes. Doxepin administration attenuated hyperglycemia, steatosis, and obesity in obese diabetic mice with upregulated FAM3A expression in liver and brown adipose tissues (BAT). Notably, doxepin failed to correct dysregulated glucose and lipid metabolism in FAM3A-deficient mice fed on high-fat diet. Doxepin's effects on ATP production, Akt activation, gluconeogenesis, and lipogenesis repression were also blunted in FAM3A-deficient mouse livers. In conclusion, FAM3A is a therapeutic target for diabetes and steatosis. Antidepressive drug doxepin activates FAM3A signaling pathways in liver and BAT to improve hyperglycemia and steatosis of obese diabetic mice. Doxepin might be preferentially recommended as an antidepressive drug in potential treatment of patients with diabetes complicated with depression.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, China
| | - Xiangyang Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Yanjin Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Junpei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Yuhong Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Yongsheng Chang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Qinghua Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
- Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| |
Collapse
|
22
|
Saxena S, Mathur P, Shukla V, Rani V. Differential expression of novel MicroRNAs from developing fetal heart of Gallus gallus domesticus implies a role in cardiac development. Mol Cell Biochem 2019; 462:157-165. [PMID: 31494815 DOI: 10.1007/s11010-019-03618-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/24/2019] [Indexed: 01/26/2023]
Abstract
Heart development is a complex process regulated by multi-layered genetic as well epigenetic regulators many of which are still unknown. Besides their critical role during cardiac development, these molecular regulators emerge as key modulators of cardiovascular pathologies, where fetal cardiac genes' re-expression is witnessed. MicroRNAs have recently emerged as a crucial part of signalling cascade in both development and diseases. We aimed to identify, validate, and perform functional annotation of putative novel miRNAs using chicken as a cardiac development model system. Novel miRNAs were obtained through deep sequencing of small RNAs extracted from chicken embryonic cardiac tissue of different developmental stages. After filtering out real pre-miRNAs, their expression analysis, potential target gene's prediction and functional annotations were performed. Expression analysis revealed that miRNAs were differentially expressed during different developmental stages of chicken heart. The expression of selected putative novel miRNAs was further validated by real-time PCR. Our analysis indicated the presence of novel cardiac miRNAs that might be regulating critical cardiac development events such as cardiac cell growth, differentiation, cardiac action potential generation and signal transduction.
Collapse
Affiliation(s)
- Sharad Saxena
- Transcriptome Laboratory, Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, UP, 201307, India
| | - Priyanka Mathur
- Transcriptome Laboratory, Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, UP, 201307, India
| | - Vaibhav Shukla
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vibha Rani
- Transcriptome Laboratory, Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, UP, 201307, India.
| |
Collapse
|
23
|
Abstract
The health-promoting effects of physical activity to prevent and treat metabolic disorders are numerous. However, the underlying molecular mechanisms are not yet completely deciphered. In recent years, studies have referred to the liver as an endocrine organ, since it releases specific proteins called hepatokines. Some of these hepatokines are involved in whole body metabolic homeostasis and are theorized to participate in the development of metabolic disease. In this regard, the present review describes the role of Fibroblast Growth Factor 21, Fetuin-A, Angiopoietin-like protein 4, and Follistatin in metabolic disease and their production in response to acute exercise. Also, we discuss the potential role of hepatokines in mediating the beneficial effects of regular exercise and the future challenges to the discovery of new exercise-induced hepatokines.
Collapse
Affiliation(s)
- Gaël Ennequin
- PEPITE EA4267, EPSI, Université de Bourgogne Franche-Comté , Besançon , France
| | - Pascal Sirvent
- Université Clermont Auvergne, Laboratoire des Adaptations Métaboliques à l'Exercice en conditions Physiologiques et Pathologiques (AME2P), CRNH Auvergne, Clermont-Ferrand , France
| | - Martin Whitham
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham , Birmingham , United Kingdom
| |
Collapse
|
24
|
González-Prendes R, Mármol-Sánchez E, Quintanilla R, Castelló A, Zidi A, Ramayo-Caldas Y, Cardoso TF, Manunza A, Cánovas Á, Amills M. About the existence of common determinants of gene expression in the porcine liver and skeletal muscle. BMC Genomics 2019; 20:518. [PMID: 31234802 PMCID: PMC6591854 DOI: 10.1186/s12864-019-5889-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 06/07/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The comparison of expression QTL (eQTL) maps obtained in different tissues is an essential step to understand how gene expression is genetically regulated in a context-dependent manner. In the current work, we have compared the transcriptomic and eQTL profiles of two porcine tissues (skeletal muscle and liver) which typically show highly divergent expression profiles, in 103 Duroc pigs genotyped with the Porcine SNP60 BeadChip (Illumina) and with available microarray-based measurements of hepatic and muscle mRNA levels. Since structural variation could have effects on gene expression, we have also investigated the co-localization of cis-eQTLs with copy number variant regions (CNVR) segregating in this Duroc population. RESULTS The analysis of differential expresssion revealed the existence of 1204 and 1490 probes that were overexpressed and underexpressed in the gluteus medius muscle when compared to liver, respectively (|fold-change| > 1.5, q-value < 0.05). By performing genome scans in 103 Duroc pigs with available expression and genotypic data, we identified 76 and 28 genome-wide significant cis-eQTLs regulating gene expression in the gluteus medius muscle and liver, respectively. Twelve of these cis-eQTLs were shared by both tissues (i.e. 42.8% of the cis-eQTLs identified in the liver were replicated in the gluteus medius muscle). These results are consistent with previous studies performed in humans, where 50% of eQTLs were shared across tissues. Moreover, we have identified 41 CNVRs in a set of 350 pigs from the same Duroc population, which had been genotyped with the Porcine SNP60 BeadChip by using the PennCNV and GADA softwares, but only a small proportion of these CNVRs co-localized with the cis-eQTL signals. CONCLUSION Despite the fact that there are considerable differences in the gene expression patterns of the porcine liver and skeletal muscle, we have identified a substantial proportion of common cis-eQTLs regulating gene expression in both tissues. Several of these cis-eQTLs influence the mRNA levels of genes with important roles in meat (CTSF) and carcass quality (TAPT1), lipid metabolism (TMEM97) and obesity (MARC2), thus evidencing the practical importance of dissecting the genetic mechanisms involved in their expression.
Collapse
Affiliation(s)
- Rayner González-Prendes
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.,Departament de Producció Animal-Agrotecnio Center, Universitat de Lleida, 191 Rovira Roure, 25198, Lleida, Spain
| | - Emilio Mármol-Sánchez
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| | - Raquel Quintanilla
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140, Caldes de Montbui, Spain
| | - Anna Castelló
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.,Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Ali Zidi
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| | - Yuliaxis Ramayo-Caldas
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| | - Tainã Figueiredo Cardoso
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.,CAPES Foundation, Ministry of Education of Brazil, Brasilia D. F, Zip Code 70.040-020, Brazil
| | - Arianna Manunza
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| | - Ángela Cánovas
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.,Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Marcel Amills
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain. .,Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
| |
Collapse
|
25
|
Yang W, Feng B, Meng Y, Wang J, Geng B, Cui Q, Zhang H, Yang Y, Yang J. FAM3C-YY1 axis is essential for TGFβ-promoted proliferation and migration of human breast cancer MDA-MB-231 cells via the activation of HSF1. J Cell Mol Med 2019; 23:3464-3475. [PMID: 30887707 PMCID: PMC6484506 DOI: 10.1111/jcmm.14243] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/22/2018] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
Family with sequence similarity three member C (FAM3C) (interleukin‐like EMT inducer [ILEI]), heat shock factor 1 (HSF1) and Ying‐Yang 1 (YY1) have been independently reported to be involved in the pathogenesis of various cancers. However, whether they are coordinated to trigger the development of cancer remains unknown. This study determined the role and mechanism of YY1 and HSF1 in FAM3C‐induced proliferation and migration of breast cancer cells. In human MDA‐MB‐231 breast cancer cell line, transforming growth factor‐β (TGFβ) up‐regulated FAM3C, HSF1 and YY1 expressions. FAM3C overexpression promoted the proliferation and migration of MDA‐MB‐231 cells with YY1 and HSF1 up‐regulation, whereas FAM3C silencing exerted the opposite effects. FAM3C inhibition repressed TGFβ‐induced HSF1 activation, and proliferation and migration of breast cancer cells. YY1 was shown to directly activate HSF1 transcription to promote the proliferation and migration of breast cancer cells. YY1 silencing blunted FAM3C‐ and TGFβ‐triggered activation of HSF1‐Akt‐Cyclin D1 pathway, and proliferation and migration of breast cancer cells. Inhibition of HSF1 blocked TGFβ‐, FAM3C‐ and YY1‐induced proliferation and migration of breast cancer cells. YY1 and HSF1 had little effect on FAM3C expression. Similarly, inhibition of HSF1 also blunted FAM3C‐ and TGFβ‐promoted proliferation and migration of human breast cancer BT‐549 cells. In human breast cancer tissues, FAM3C, YY1 and HSF1 protein expressions were increased. In conclusion, FAM3C activated YY1‐HSF1 signalling axis to promote the proliferation and migration of breast cancer cells. Furthermore, novel FAM3C‐YY1‐HSF1 pathway plays an important role in TGFβ‐triggered proliferation and migration of human breast cancer MDA‐MB‐231 cells.
Collapse
Affiliation(s)
- Weili Yang
- Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Department of Biomedical Informatics, School of Basic Medical Sciences, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Biaoqi Feng
- Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Yuhong Meng
- Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Junpei Wang
- Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Department of Biomedical Informatics, School of Basic Medical Sciences, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Bin Geng
- State Key Laboratory of Cardiovascular Disease, Hypertension Center, Fuwai Hospital, Peking University Health Science Center, CAMS & PUMC, Beijing, China
| | - Qinghua Cui
- Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Department of Biomedical Informatics, School of Basic Medical Sciences, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Hongquan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| | - Yang Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jichun Yang
- Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| |
Collapse
|
26
|
Shi M, Duan G, Nie S, Shen S, Zou X. Elevated FAM3C promotes cell epithelial- mesenchymal transition and cell migration in gastric cancer. Onco Targets Ther 2018; 11:8491-8505. [PMID: 30584315 PMCID: PMC6287415 DOI: 10.2147/ott.s178455] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background Tumor metastasis is an important factor in treatment failure for advanced gastric cancer. Family with sequence similarity 3 member C (FAM3C) is known to play a critical role in inducing epithelial–mesenchymal transition in several cancer types, while its role in gastric cancer is unidentified. The aim of this study was to investigate the role of FAM3C in gastric cancer and provide new information on the receptor tyrosine-kinase pathway and cytokine-based therapies. Methods FAM3C expression was tested in human gastric cancer tissue and adjacent normal mucosa, and the prognostic effect of FAM3C was analyzed in data from the Cancer Genome Atlas (TCGA). The role of FAM3C in gastric cancer proliferation and metastasis was investigated in vitro and in vivo. Western blot analysis and immunofluorescence were used to detect the underlying mechanisms. Results FAM3C expression was increased in gastric cancer tissue and showed cytoplasmic distribution. Gastric cancer patients with FAM3C overexpression had significantly worse prognoses based on TCGA data. In the gastric cancer cell lines MKN45 and AGS, knockdown of FAM3C dramatically attenuated cell migration, but had almost no influence on proliferation, while exogenous FAM3C promoted cell migration in a cell line with low FAM3C expression. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment of TCGA data showed that FAM3C was mainly associated with genes involved in focal adhesion, extracellular matrix–receptor interactions and the PI3K–Akt signaling pathway. Knockdown of FAM3C in gastric cancer cell lines significantly suppressed epithelial–mesenchymal transition, as demonstrated by increased expression of E-cadherin and decreased expression of Snail and Slug. Furthermore, knockdown of FAM3C strongly suppressed activation of the PI3K–Akt signaling pathway. Finally, we confirmed that FAM3C knockdown significantly decreased metastatic lesions in vivo. Conclusion Our study demonstrated that FAM3C can promote gastric cancer metastasis both in vitro and in vivo. FAM3C should be taken into consideration for gastric cancer treatments involving inhibition of the ligands and downstream pathways of receptor tyrosine kinases.
Collapse
Affiliation(s)
- Mengyue Shi
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China, ;
| | - Guihua Duan
- Department of Gastroenterology, First People's Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming 650032, China
| | - Shuang Nie
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China, ;
| | - Shanshan Shen
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China, ;
| | - Xiaoping Zou
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China, ;
| |
Collapse
|
27
|
Chen X, Chen C, Hao J, Qin R, Qian B, Yang K, Zhang J, Zhang F. AKR1B1 Upregulation Contributes to Neuroinflammation and Astrocytes Proliferation by Regulating the Energy Metabolism in Rat Spinal Cord Injury. Neurochem Res 2018; 43:1491-1499. [DOI: 10.1007/s11064-018-2570-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 06/01/2018] [Accepted: 06/06/2018] [Indexed: 12/29/2022]
|
28
|
Wang J, Yang W, Chen Z, Chen J, Meng Y, Feng B, Sun L, Dou L, Li J, Cui Q, Yang J. Long Noncoding RNA lncSHGL Recruits hnRNPA1 to Suppress Hepatic Gluconeogenesis and Lipogenesis. Diabetes 2018; 67:581-593. [PMID: 29382663 DOI: 10.2337/db17-0799] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 01/16/2018] [Indexed: 11/13/2022]
Abstract
Mammalian genomes encode a huge number of long noncoding RNAs (lncRNAs) with unknown functions. This study determined the role and mechanism of a new lncRNA, lncRNA suppressor of hepatic gluconeogenesis and lipogenesis (lncSHGL), in regulating hepatic glucose/lipid metabolism. In the livers of obese mice and patients with nonalcoholic fatty liver disease, the expression levels of mouse lncSHGL and its human homologous lncRNA B4GALT1-AS1 were reduced. Hepatic lncSHGL restoration improved hyperglycemia, insulin resistance, and steatosis in obese diabetic mice, whereas hepatic lncSHGL inhibition promoted fasting hyperglycemia and lipid deposition in normal mice. lncSHGL overexpression increased Akt phosphorylation and repressed gluconeogenic and lipogenic gene expression in obese mouse livers, whereas lncSHGL inhibition exerted the opposite effects in normal mouse livers. Mechanistically, lncSHGL recruited heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) to enhance the translation efficiency of CALM mRNAs to increase calmodulin (CaM) protein level without affecting their transcription, leading to the activation of the phosphatidyl inositol 3-kinase (PI3K)/Akt pathway and repression of the mTOR/SREBP-1C pathway independent of insulin and calcium in hepatocytes. Hepatic hnRNPA1 overexpression also activated the CaM/Akt pathway and repressed the mTOR/SREBP-1C pathway to ameliorate hyperglycemia and steatosis in obese mice. In conclusion, lncSHGL is a novel insulin-independent suppressor of hepatic gluconeogenesis and lipogenesis. Activating the lncSHGL/hnRNPA1 axis represents a potential strategy for the treatment of type 2 diabetes and steatosis.
Collapse
Affiliation(s)
- Junpei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
- Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Weili Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
- Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Zhenzhen Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Ji Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Yuhong Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Biaoqi Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Libo Sun
- Beijing You An Hospital, Capital Medical University, Beijing, China
| | - Lin Dou
- Key Laboratory of Geriatrics, Beijing Institute of Geriatrics & Beijing Hospital, Ministry of Health, Beijing, China
| | - Jian Li
- Key Laboratory of Geriatrics, Beijing Institute of Geriatrics & Beijing Hospital, Ministry of Health, Beijing, China
| | - Qinghua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing, China
| |
Collapse
|
29
|
Zhang X, Yang W, Wang J, Meng Y, Guan Y, Yang J. FAM3 gene family: A promising therapeutical target for NAFLD and type 2 diabetes. Metabolism 2018; 81:71-82. [PMID: 29221790 DOI: 10.1016/j.metabol.2017.12.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/08/2017] [Accepted: 12/01/2017] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) and diabetes are severe public health issues worldwide. The Family with sequence similarity 3 (FAM3) gene family consists of four members designated as FAM3A, FAM3B, FAM3C and FAM3D, respectively. Recently, there had been increasing evidence that FAM3A, FAM3B and FAM3C are important regulators of glucose and lipid metabolism. FAM3A expression is reduced in the livers of diabetic rodents and NAFLD patients. Hepatic FAM3A restoration activates ATP-P2 receptor-Akt and AMPK pathways to attenuate steatosis and hyperglycemia in obese diabetic mice. FAM3C expression is also reduced in the liver under diabetic condition. FAM3C is a new hepatokine that activates HSF1-CaM-Akt pathway and represses mTOR-SREBP1-FAS pathway to suppress hepatic gluconeogenesis and lipogenesis. In contrast, hepatic expression of FAM3B, also called PANDER, is increased under obese state. FAM3B promotes hepatic lipogenesis and gluconeogenesis by repressing Akt and AMPK activities, and activating lipogenic pathway. Under obese state, the imbalance among hepatic FAM3A, FAM3B and FAM3C signaling networks plays important roles in the pathogenesis of NAFLD and type 2 diabetes. This review briefly discussed the latest research progress on the roles and mechanisms of FAM3A, FAM3B and FAM3C in the regulation of hepatic glucose and lipid metabolism.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Weili Yang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Junpei Wang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Yuhong Meng
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Jichun Yang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
30
|
Howley BV, Howe PH. TGF-beta signaling in cancer: post-transcriptional regulation of EMT via hnRNP E1. Cytokine 2018; 118:19-26. [PMID: 29396052 DOI: 10.1016/j.cyto.2017.12.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 12/29/2017] [Indexed: 12/12/2022]
Abstract
The TGFβ signaling pathway is a critical regulator of cancer progression in part through induction of the epithelial to mesenchymal transition (EMT). This process is aberrantly activated in cancer cells, facilitating invasion of the basement membrane, survival in the circulatory system, and dissemination to distant organs. The mechanisms through which epithelial cells transition to a mesenchymal state involve coordinated transcriptional and post-transcriptional control of gene expression. One such mechanism of control is through the RNA binding protein hnRNP E1, which regulates splicing and translation of a cohort of EMT and stemness-associated transcripts. A growing body of evidence indicates a major role for hnRNP E1 in the control of epithelial cell plasticity, especially in the context of carcinoma progression. Here, we review the multiple mechanisms through which hnRNP E1 functions to control EMT and metastatic progression.
Collapse
Affiliation(s)
- Breege V Howley
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Philip H Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
31
|
Chen Z, Wang J, Yang W, Chen J, Meng Y, Feng B, Chi Y, Geng B, Zhou Y, Cui Q, Yang J. FAM3C activates HSF1 to suppress hepatic gluconeogenesis and attenuate hyperglycemia of type 1 diabetic mice. Oncotarget 2017; 8:106038-106049. [PMID: 29285313 PMCID: PMC5739700 DOI: 10.18632/oncotarget.22524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/30/2017] [Indexed: 12/17/2022] Open
Abstract
FAM3C, a member of FAM3 gene family, has been shown to improve insulin resistance and hyperglycemia in obese mice. This study further determined whether FAM3C functions as a hepatokine to suppress hepatic gluconeogenesis of type 1 diabetic mice. In STZ-induced type 1 diabetic mouse liver, the FAM3C-HSF1-CaM signaling axis was repressed. Hepatic FAM3C overexpression activated HSF1-CaM-Akt pathway to repress gluconeogenic gene expression and ameliorate hyperglycemia of type 1 diabetic mice. Moreover, hepatic HSF1 overexpression also activated CaM-Akt pathway to repress gluconeogenic gene expression and improve hyperglycemia of type 1 diabetic mice. Hepatic FAM3C and HSF1 overexpression had little effect on serum insulin levels in type 1 diabetic mice. In cultured hepatocytes, conditioned medium of Ad-FAM3C-infected cells induced Akt phosphorylation. Moreover, Akt activation and gluconeogenesis repression induced by FAM3C overexpression were reversed by the treatment with anti-FAM3C antibodies. Treatment with recombinant FAM3C protein induced Akt activation in a HSF1- and CaM-dependent manner in cultured hepatocytes. Furthermore, recombinant FAM3C protein repressed gluconeogenic gene expression and gluconeogenesis by inactivating FOXO1 in a HSF1-dependent manner in cultured hepatocytes. In conclusion, FAM3C is a new hepatokine that suppresses hepatic gluconeogenic gene expression and gluconeogenesis independent of insulin by activating HSF1-CaM-Akt pathway.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Junpei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Weili Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Ji Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Yuhong Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Biaoqi Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Yujing Chi
- Institute of Clinical Molecular Biology & Central Laboratory, Peking University People's Hospital, Beijing 100044, China
| | - Bin Geng
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Yong Zhou
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qinghua Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China.,Department of Biomedical Informatics, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education, Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of The Ministry of Education Center for Non-Coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|