1
|
DePaolo J, Bornstein MR, Judy R, Abramowitz S, Verma SS, Levin MG, Penn Medicine Biobank, Arany Z, Damrauer SM. Titin-Truncating variants predispose to dilated cardiomyopathy in populations genetically similar to african and european reference populations. PLoS Genet 2025; 21:e1011727. [PMID: 40512817 DOI: 10.1371/journal.pgen.1011727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/14/2025] [Indexed: 06/18/2025] Open
Abstract
The effect of high percentage spliced in (hiPSI) TTN truncating variants (TTNtvs) on risk of dilated cardiomyopathy (DCM) has historically been studied among population subgroups defined by genetic similarity to European reference populations. This has raised questions about the effect of TTNtvs in diverse populations, especially among individuals genetically similar to African reference populations. To determine the effect of TTNtvs on cardiovascular disease risk, we leveraged whole exome sequencing and electronic health record data from 43,731 Penn Medicine Biobank (PMBB) participants recruited from across the Penn Medicine healthcare system. Fraction of genetic similarity to the 1000 Genomes Project (1000G) African (AFR) reference population was determined using ADMIXTURE analysis. Logistic regression was performed to evaluate the association of hiPSI TTNtvs with prevalent DCM and atrial fibrillation (Afib), and linear regression was used to evaluate the association with reduced left ventricular ejection fraction (LVEF) either using dichotomized genetically similar population subgroup analysis or integrating ADMIXTURE population fraction. When individuals were assigned to population subgroups based on genetic similarity to the 1000G reference populations, hiPSI TTNtvs conferred significant risk of DCM among those genetically similar to the 1000G European (EUR) reference population (OR=6.12, 95% confidence intervals [CI] 4.33 to 8.65, P < 0.001) and individuals genetically similar to the AFR reference population (OR=3.44, 95% CI 1.97 to 5.99, P < 0.001). These results were consistent when considering the effect of change in fraction of similarity to the African reference population by ADMIXTURE as a continuous variable. Similar results were observed for the effect of TTNtvs on Afib and LVEF. Our findings demonstrate that TTNtvs are associated with increased risk of DCM, reduced LVEF, and Afib among a diverse cohort. There is no significant difference in effect of TTNtvs across fractions of similarity to the AFR reference population suggesting genetic background should not be considered when screening individuals for titin-related cardiovascular disease.
Collapse
Affiliation(s)
- John DePaolo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marc R Bornstein
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Renae Judy
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sarah Abramowitz
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael G Levin
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, United States of America
| | | | - Zoltan Arany
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, United States of America
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
2
|
Vahdat-Lasemi F, Farhoudi L, Hosseinikhah SM, Santos RD, Sahebkar A. Angiopoietin-like protein inhibitors: Promising agents for the treatment of familial hypercholesterolemia and atherogenic dyslipidemia. Atherosclerosis 2025; 405:119235. [PMID: 40344904 DOI: 10.1016/j.atherosclerosis.2025.119235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/23/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND AND AIMS This review examines the physiological functions of Angiopoietin-like proteins (ANGPTLs) in lipid metabolism and the epidemiology of atherosclerotic cardiovascular disease (ASCVD), while discussing their potential as therapies for dyslipidemias. METHODS A review of contemporary literature on ANGPTLs was conducted. RESULTS ANGPTLs comprise eight secreted proteins that share structural similarities with the angiopoietin family and serve as key regulators of various physiological and biochemical functions. Notably, ANGPTL3, ANGPTL4, and ANGPTL8 act as physiological inhibitors of lipoprotein lipase (LPL), playing a crucial role in lipoprotein and triglyceride metabolism in response to the body's nutritional status. A deficiency in these proteins is linked to hypolipidemia, characterized by a decrease in all lipid fractions, and genetic studies indicate a reduced risk of ASCVD in individuals with loss-of-function variants in ANGPTL3 and ANGPTL4. Conversely, elevated levels of ANGPTL3, ANGPTL4, and ANGPTL8 seem to increase the risk of cardiovascular disease. The role of ANGPTLs in regulating lipid metabolism underscores their potential in targeted therapies for managing dyslipidemias and lowering ASCVD risk, particularly in patients with difficult-to-control dyslipidemia phenotypes, such as homozygous Familial Hypercholesterolemia and mixed dyslipidemia. CONCLUSIONS The development of ANGPTL inhibitors could provide an effective strategy for preventing ASCVD.
Collapse
Affiliation(s)
- Fatemeh Vahdat-Lasemi
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Farhoudi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Maryam Hosseinikhah
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raul D Santos
- Academic Research Organization, Hospital Israelita Albert Einstein, Sao Paulo, Brazil; Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Cummings BB, Joing MP, Bouchard PR, Milton MN, Moesta PF, Ramanan V, Lane A, Hirman J, Trauger JW, Maratos-Flier E, Voznesensky A, Splawski I, Nimonkar AV, Flaherty MM, Yi BA, Meyers D, Huet F, Sahambi SK, Yates DP, Hom D, Hinder M, Basson CT, O'Donnell C, Siegelman ES, Garrett CE, Lehrer-Graiwer J, Juliano RA, Weiss EJ. Safety and efficacy of a novel ANGPTL4 inhibitory antibody for lipid lowering: results from phase 1 and phase 1b/2a clinical studies. Lancet 2025; 405:1923-1934. [PMID: 40383129 DOI: 10.1016/s0140-6736(25)00825-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Genetic studies have established angiopoietin-related protein 4 (ANGPTL4) as a key regulator of triglyceride metabolism and a promising target to reduce atherosclerotic cardiovascular disease (ASCVD) risk beyond traditional risk factors. Human ANGPTL4 loss-of-function shows no adverse consequences and is associated with reduced triglycerides and remnant cholesterol, and a reduced risk of type 2 diabetes and ASCVD. Nonetheless, development of ANGPTL4 inhibitors has been delayed due to adverse findings in ANGPTL4-knockout mice fed a high saturated fat diet, including lipid accumulation in mesenteric lymph nodes, systemic inflammation, adverse clinical signs, and reduced survival. We previously reported the development and preclinical characterisation of MAR001, an ANGPTL4 inhibitory antibody. Here, we report a comprehensive safety assessment of ANGPTL4 inhibition, including novel analysis of genetic ANGPTL4 loss on mesenteric lymph node architecture in humans and two early-phase clinical trials. METHODS MAR001 was evaluated in a first-in-human, randomised, placebo-controlled, single-ascending-dose phase 1 study with three parts in which participants received a single subcutaneous injection of MAR001 or placebo. The study was developed and conducted by Novartis Biomedical Research (Cambridge, MA, USA). Eligible participants enrolled in part 1A were healthy men and women aged between 18 years and 65 years with a bodyweight of at least 50 kg and a BMI of 18-30 kg/m2. Participants in part 1B weighed at least 70 kg and had a BMI of 30-40 kg/m2. Participants in part 1C weighed at least 59 kg and had fasting triglycerides in the range of 200-500 mg/dL. The primary objectives were to assess the safety and tolerability of a single subcutaneous injection of MAR001 up to and including 141 days post-dose and to assess the pharmacokinetics of single-dose subcutaneous administration in healthy participants. MAR001 was subsequently assessed in a randomised, double-blind, placebo-controlled phase 1b/2a study in participants with metabolic dysfunction. The study was done at two sites in Australia. Eligible participants were adults with hypertriglyceridaemia (in the screening range of ≥1·7 mmol/L and ≤5·6 mmol/L; ≥151 mg/dL and ≤496 mg/dL) and a history of type 2 diabetes, or a screening homeostatic model assessment for insulin resistance (HOMA-IR) value greater than 2·2 and abdominal obesity (defined as waist circumference >88 cm for women and >102 cm for men; > 80 cm for Asian women and >90 cm for Asian men). The primary objective was to characterise the safety and tolerability of multiple doses of MAR001 in participants with metabolic dysfunction. The phase 1b/2a study is registered with ClinicalTrials.gov, NCT05896254. FINDINGS We found no evidence of clinical adversity in human germline ANGPTL4 loss-of-function, adding to preclinical support for initiating human studies. Between Nov 20, 2017, and Sept 10, 2019, in the first-in-human, randomised, placebo-controlled, single-ascending-dose phase 1 study, part 1A enrolled 32 healthy participants: six each received 15 mg, 50 mg, 150 mg, or 450 mg of MAR001, and eight received placebo. Part 1B enrolled 12 participants: nine received 450 mg of MAR001 and three received placebo. Part 1C enrolled 12 participants: eight received 450 mg of MAR001 and four received placebo. Between Nov 24, 2013, and July 1, 2024, in the multidose phase 1b/2a randomised, double-blind, placebo-controlled study, 55 participants were randomly assigned to receive subcutaneous injections of placebo (19 participants) or MAR001 at doses of 150 mg (ten participants), 300 mg (nine participants), or 450 mg (17 participants), followed by a 12-week safety follow-up period. MAR001 was safe and generally well tolerated, and we observed no treatment-related systemic inflammatory biomarker elevations or changes in mesenteric lymph node size or inflammation assessed by MRI. MAR001 (450 mg) yielded placebo-adjusted week 12 mean reductions in triglycerides of 52·7% (90% CI -77·0 to -28·3) and in remnant cholesterol of 52·5% (-76·1 to -28·9). INTERPRETATION ANGPTL4 inhibition with MAR001 can safely and effectively reduce circulating triglycerides and remnant cholesterol. The findings of these trials support further research and development of MAR001 as a promising potential lipid-lowering therapy to reduce risk of ASCVD. FUNDING Marea Therapeutics.
Collapse
Affiliation(s)
| | | | - Page R Bouchard
- Marea Therapeutics, San Francisco, CA, USA; Novartis Biomedical Research, Cambridge, MA, USA
| | - Mark N Milton
- Marea Therapeutics, San Francisco, CA, USA; Novartis Biomedical Research, Cambridge, MA, USA
| | | | | | | | - Joe Hirman
- Marea Therapeutics, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Douglas Hom
- Novartis Biomedical Research, Cambridge, MA, USA
| | | | | | | | - Evan S Siegelman
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
4
|
Wulff AB. Safely lowering triglycerides through ANGPTL4 inhibition. Lancet 2025; 405:1885-1887. [PMID: 40383130 DOI: 10.1016/s0140-6736(25)00988-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Affiliation(s)
- Anders B Wulff
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, DK-2730 Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark.
| |
Collapse
|
5
|
Nordestgaard AT, Tybjærg-Hansen A, Mansbach H, Kersten S, Nordestgaard BG, Rosenson RS. Target Populations for Novel Triglyceride-Lowering Therapies. J Am Coll Cardiol 2025; 85:1876-1897. [PMID: 40368577 DOI: 10.1016/j.jacc.2025.02.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 05/16/2025]
Abstract
Lipoprotein lipase regulates triglyceride hydrolysis and contributes to cellular uptake of triglyceride-rich lipoprotein remnants. Multiple pathways modulate lipoprotein lipase activity, which has prompted interest in the development of drugs that increase lipoprotein lipase activity as means to reduce risk for acute pancreatitis, atherosclerotic cardiovascular disease, and metabolic dysfunction-associated steatohepatitis through reduction of circulating triglycerides and remnant cholesterol. The authors provide an overview of the target populations for agents that lower triglycerides and remnant cholesterol through increased lipoprotein lipase activity, the drugs being developed for these indications, including apolipoprotein C-III and angiopoietin-like protein 3, 3/8, and 4 inhibitors, and the epidemiologic and genetic evidence supporting the use of these drugs for the prevention of atherosclerotic cardiovascular disease and acute pancreatitis. In addition, the authors provide a corresponding overview of fibroblast growth factor-21 analogues that share many characteristics with these novel triglyceride-lowering drugs. Apolipoprotein C-III inhibitors, angiopoietin-like protein 3, 3/8, and 4 inhibitors, and fibroblast growth factor-21 analogues have pronounced triglyceride-lowering and remnant cholesterol-lowering effects. In clinical trials, apolipoprotein C-III inhibitors have been shown to lower risk for acute pancreatitis in patients with severe hypertriglyceridemia and are approved for this indication, while fibroblast growth factor-21 analogues reduce hepatic steatosis and fibrosis in patients with metabolic dysfunction-associated steatohepatitis. It remains to be seen whether these novel drugs may lower risk for atherosclerotic cardiovascular disease as well.
Collapse
Affiliation(s)
- Ask T Nordestgaard
- Center for Cardiovascular Disease Prevention, Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Clinical Biochemistry and the Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark.
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Sander Kersten
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry and the Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert S Rosenson
- Metabolism and Lipids Unit, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
6
|
Chen Y, Johnson SM, Burr SD, Povero D, Anderson AM, McMahon CE, Liu J. Absence of the intracellular lipolytic inhibitor G0S2 enhances intravascular triglyceride clearance and abolishes diet-induced hypertriglyceridemia. J Clin Invest 2025; 135:e181754. [PMID: 40100923 PMCID: PMC12077901 DOI: 10.1172/jci181754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 03/12/2025] [Indexed: 03/20/2025] Open
Abstract
The interplay between intracellular and intravascular lipolysis is crucial for maintaining circulating lipid levels and systemic energy homeostasis. Adipose triglyceride lipase (ATGL) and lipoprotein lipase (LPL), the primary triglyceride (TG) lipases responsible for these two spatially separate processes, are highly expressed in adipose tissue. Yet the mechanisms underlying their coordinated regulation remain undetermined. Here, we demonstrate that genetic ablation of G0S2, a specific inhibitory protein of ATGL, completely abolished diet-induced hypertriglyceridemia and significantly attenuated atherogenesis in mice. These effects were attributable to enhanced whole-body TG clearance, not altered hepatic TG secretion. Specifically, G0S2 deletion increased circulating LPL concentration and activity, predominantly through LPL production from white adipose tissue (WAT). Strikingly, transplantation of G0S2-deficient WAT normalized plasma TG levels in mice with hypertriglyceridemia. In conjunction with improved insulin sensitivity and decreased ANGPTL4 expression, the absence of G0S2 enhanced the stability of LPL protein in adipocytes, a phenomenon that could be reversed upon ATGL inhibition. Collectively, these findings highlight the pivotal role of adipocyte G0S2 in regulating both intracellular and intravascular lipolysis, and the possibility of targeting G0S2 as a viable pharmacological approach to reducing levels of circulating TGs.
Collapse
Affiliation(s)
- Yongbin Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Scott M. Johnson
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Stephanie D. Burr
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, and
| | - Davide Povero
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Division of Gastroenterology and Hepatology, Mayo Clinic in Rochester, Rochester, Minnesota, USA
| | - Aaron M. Anderson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Cailin E. McMahon
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Molecular Biology and Genetics Department, Cornell College of Agriculture and Life Sciences, Ithaca, New York, USA
| | - Jun Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, and
| |
Collapse
|
7
|
Cummings BB, Bouchard PR, Milton MN, Moesta PF, Ramanan V, Trauger JW, Maratos-Flier E, Voznesensky A, Splawski I, Nimonkar AV, DiPetrillo K, LaSala D, Pan M, Flaherty MM, Huet F, Sahambi SK, Dong J, Knee D, Cebe R, Huber T, Lehrer-Graiwer J, Juliano RA, Weiss EJ. An ANGPTL4 inhibitory antibody safely improves lipid profiles in non-human primates. EBioMedicine 2025:105748. [PMID: 40360349 DOI: 10.1016/j.ebiom.2025.105748] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Angiopoietin-like protein 4 (ANGPTL4) inhibition is a promising approach to manage atherogenic dyslipidaemia and residual atherosclerotic cardiovascular disease (ASCVD) risk. Human ANGPTL4 loss-of-function (LoF) is associated with reduced plasma triglyceride (TG), remnant cholesterol (RC), and apolipoprotein B (ApoB) levels, and lower risk of type 2 diabetes and ASCVD, without observable safety concerns. However, development of ANGPTL4 inhibitors has been stalled by adverse findings in Angptl4 knockout mice fed a high-saturated-fat diet (HSFD), which show lipid accumulation in mesenteric lymph nodes (MLNs), systemic inflammation, severe adverse clinical signs, and reduced survival. METHODS Here, we present the development and preclinical characterisation of MAR001, a humanised monoclonal ANGPTL4 inhibitor antibody. We assessed single-dose MAR001 efficacy in hypertriglyceridemic (HTG) non-human primates (NHPs, n = 4), and safety in two NHP toxicology studies: a 15-week subchronic study with a standard or HSFD (n = 36), and a 9-month chronic study exclusively on an HSFD (n = 24). FINDINGS In HTG monkeys, single-dose MAR001 treatment reduced plasma TG by up to 58%, non-high-density lipoprotein cholesterol by 38%, ApoB by 30%, and RC by 59%. In safety studies, MAR001 was well tolerated without clinically adverse findings with either diet. Animals fed an HSFD exhibited minimal to moderate foamy macrophage formation in MLNs, but importantly, these histological findings did not progress to degeneration, necrosis, inflammation, fibrosis, or other reactive changes, and with no evidence of systemic effects, including no evidence of systemic inflammation or clinical adverse signs. INTERPRETATION MAR001 improved plasma lipid profiles in NHPs without clinical adversity, even during prolonged HSFD feeding. The favourable NHP safety profile aligns with human ANGPTL4 LoF findings, and contrasts with the severe pathology in mouse knockout models on an HSFD. These findings supported MAR001 clinical studies reported in our concurrent publication, which demonstrated robust lipid improvements without lymphatic pathology. Overall, these findings support continued development of MAR001 as a promising new therapy for ASCVD risk reduction. FUNDING Marea Therapeutics.
Collapse
Affiliation(s)
- Beryl B Cummings
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - Page R Bouchard
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA; Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Mark N Milton
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA; Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Peter F Moesta
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - Vyas Ramanan
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - John W Trauger
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | | | - Andrei Voznesensky
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Igor Splawski
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Amitabh V Nimonkar
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Keith DiPetrillo
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Daniel LaSala
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Meihui Pan
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Meghan M Flaherty
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Francois Huet
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Sukhdeep K Sahambi
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Jijun Dong
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Deborah Knee
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Regis Cebe
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Thomas Huber
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | | | - Rebecca A Juliano
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - Ethan J Weiss
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA.
| |
Collapse
|
8
|
Cuttone A, Cannavò V, Abdullah RMS, Fugazzotto P, Arena G, Brancati S, Muscarà A, Morace C, Quartarone C, Ruggeri D, Squadrito G, Russo GT. Expanding the Use of SGLT2 Inhibitors in T2D Patients Across Clinical Settings. Cells 2025; 14:668. [PMID: 40358192 PMCID: PMC12071329 DOI: 10.3390/cells14090668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are currently recommended in patients with type 2 diabetes (T2D) to reduce serum glucose levels. Moreover, robust evidence has clearly demonstrated their beneficial cardiovascular and renal effects, making this class of drugs pivotal for the treatment of T2D, especially when complicated by diabetic kidney disease or heart failure. However, several other comorbidities are frequently encountered in T2D patients beyond these long-term diabetes complications, especially in the internal medicine setting. For some of these comorbidities, such as MAFLD and cognitive impairment, the association with diabetes is increasingly recognized, with the hypothesis of a common pathophysiologic background, whereas, for others, a coincident epidemiology linked to the ageing of populations, including that of T2D subjects, may be advocated. In the effort of personalizing T2D treatment, evidence on the potential effects of SGLT2i in these different clinical conditions is accumulating. The purpose of this narrative review is to update current literature on the effects of SGLT2i for the treatment of T2D in different clinical settings beyond glycaemic control, and to elucidate potential molecular mechanisms by which they exert these effects.
Collapse
Affiliation(s)
- Alessandro Cuttone
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Vittorio Cannavò
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Raouf Mastan Sheik Abdullah
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Pierluigi Fugazzotto
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Giada Arena
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Simona Brancati
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Andrea Muscarà
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Carmela Morace
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Cristina Quartarone
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (C.Q.); (D.R.)
| | - Domenica Ruggeri
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (C.Q.); (D.R.)
| | - Giovanni Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Giuseppina Tiziana Russo
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| |
Collapse
|
9
|
Sun Z, Yi Z, Wei C, Wang W, Ren T, Cravedi P, Tedla F, Ward SC, Azeloglu E, Schrider DR, Li Y, Khan A, Zanoni F, Fu J, Ali S, Liu S, Liang D, Liu T, Li H, Xi C, Vy TH, Mosoyan G, Sun Q, Kumar A, Zhang Z, Farouk S, Campell K, Ochando J, Lee K, Coca S, Xiang J, Connolly P, Gallon L, O'Connell PJ, Colvin R, Menon MC, Nadkarni G, He JC, Kraft M, Jiang X, Zhang X, Kiryluk K, Cherukuri A, Lakkis FG, Zhang W, Chen SH, Heeger PS, Zhang W. LILRB3 genetic variation is associated with kidney transplant failure in African American recipients. Nat Med 2025; 31:1677-1687. [PMID: 40065170 DOI: 10.1038/s41591-025-03568-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 02/04/2025] [Indexed: 04/03/2025]
Abstract
African American (AA) kidney transplant recipients exhibit a higher rate of graft loss compared with other racial and ethnic populations, highlighting the need to identify causative factors. Here, in the Genomics of Chronic Allograft Rejection cohort, pretransplant blood RNA sequencing revealed a cluster of four consecutive missense single-nucelotide polymorphisms (SNPs), within the leukocyte immunoglobulin-like receptor B3 (LILRB3) gene, strongly associated with death-censored graft loss. This SNP cluster (named LILRB3-4SNPs) encodes missense mutations at amino acids 617-618 proximal to a SHP1/2 phosphatase-binding immunoreceptor tyrosine-based inhibitory motif. The LILRB3-4SNPs cluster is specifically enriched within AA individuals and exhibited a strong association with death-censored graft loss and estimated glomerular filtration rate decline in the AA participants from multiple transplant cohorts. In two large Biobanks (BioMe and All-of-Us), the LILRB3-4SNPs cluster was associated with the early onset of end-stage renal disease and acted synergistically with the apolipoprotein L1 (APOL1) G1/G2 allele to accelerate disease progression. The SNPs were also linked to multiple immune-related diseases in AA individuals. Last, on multiomics analysis of blood and biopsies, recipients with LILRB3-4SNPs showed enhanced inflammation and monocyte ferroptosis. While larger and prospective studies are needed, our data provide insights on the genetic variation underlying kidney transplant outcomes.
Collapse
Affiliation(s)
- Zeguo Sun
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhengzi Yi
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chengguo Wei
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wenlin Wang
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tianyuan Ren
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Paolo Cravedi
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fasika Tedla
- The Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen C Ward
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evren Azeloglu
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel R Schrider
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yun Li
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University, New York, NY, USA
| | - Francesca Zanoni
- Division of Nephrology, Department of Medicine, Columbia University, New York, NY, USA
| | - Jia Fu
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sumaria Ali
- Center for Immunotherapy Research, Houston Methodist Neal Cancer Center and Houston Methodist Research Institute, Houston, TX, USA
| | - Shun Liu
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Deguang Liang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tong Liu
- Center for Advanced Proteomics Research and Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, Newark, NJ, USA
| | - Hong Li
- Center for Advanced Proteomics Research and Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, Newark, NJ, USA
| | - Caixia Xi
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thi Ha Vy
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gohar Mosoyan
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Quan Sun
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ashwani Kumar
- Department of Medicine, Yale University, New Haven, CT, USA
| | - Zhongyang Zhang
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samira Farouk
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirk Campell
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kyung Lee
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steve Coca
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jenny Xiang
- Department of Microbiology and Immunology, Weil Cornell Medicine, New York, NY, USA
| | | | - Lorenzo Gallon
- Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Philip J O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Robert Colvin
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Madhav C Menon
- Department of Medicine, Yale University, New Haven, CT, USA
| | - Girish Nadkarni
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John C He
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xuewu Zhang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Columbia University, New York, NY, USA
| | - Aravind Cherukuri
- Departments of Surgery, Immunology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fadi G Lakkis
- Departments of Surgery, Immunology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Weiguo Zhang
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Shu-Hsia Chen
- Center for Immunotherapy Research, Houston Methodist Neal Cancer Center and Houston Methodist Research Institute, Houston, TX, USA
| | - Peter S Heeger
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Weijia Zhang
- Renal Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Datta S, Koka S, Boini KM. Understanding the Role of Adipokines in Cardiometabolic Dysfunction: A Review of Current Knowledge. Biomolecules 2025; 15:612. [PMID: 40427505 PMCID: PMC12109550 DOI: 10.3390/biom15050612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/13/2025] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiometabolic risk and associated dysfunctions contribute largely to the recent rise in mortality globally. Advancements in multi-omics in recent years promise a better understanding of potential biomarkers that enable an early diagnosis of cardiometabolic dysfunction. However, the molecular mechanisms driving the onset and progression of cardiometabolic disorders remain poorly understood. Adipokines are adipocyte-specific cytokines that are central to deleterious cardiometabolic alterations. They exhibit both pro-inflammatory and anti-inflammatory effects, complicating their association with cardiometabolic disturbances. Thus, understanding the cardiometabolic association of adipokines from a molecular and signaling perspective assumes great importance. This review presents a comprehensive outline of the most prominent adipokines exhibiting pro-inflammatory and/or anti-inflammatory functions in cardiometabolic dysfunction. The review also presents an insight into the pathophysiological implications of such adipokines in different cardiometabolic dysfunction conditions, the status of adipokine druggability, and future studies that can be undertaken to address the existing scientific gap. A clear understanding of the functional and mechanistic role of adipokines can potentially improve our understanding of cardiovascular disease pathophysiology and enhance our current therapeutic regimen in the years to come.
Collapse
Affiliation(s)
- Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd., Houston, TX 77204, USA;
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A & M University, Kingsville, TX 78363, USA
| | - Krishna M. Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4349 Martin Luther King Blvd., Houston, TX 77204, USA;
| |
Collapse
|
11
|
Wulff AB, Nordestgaard BG. Genetics of remnant cholesterol. Curr Opin Lipidol 2025:00041433-990000000-00119. [PMID: 40277396 DOI: 10.1097/mol.0000000000000991] [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] [Indexed: 04/26/2025]
Abstract
PURPOSE OF REVIEW Remnant cholesterol is receiving increasing attention as a target to reduce residual atherosclerotic cardiovascular disease (ASCVD) risk in individuals already treated with statins. New therapeutic options as antisense oligonucleotides, small interfering RNA, and monoclonal antibodies allow specific targeting of genes and proteins to counter pathological pathways promoted by these genes. Identifying genetic determinants of remnant cholesterol and relating these to risk of ASCVD is thus an appealing path to identifying and evaluating new and existing drug targets. RECENT FINDINGS Human genetic epidemiology has identified several genetic variants in genes involved in lipoprotein metabolism with effect on plasma concentrations of remnant cholesterol. Lipoprotein lipase (LPL) is central to the metabolism of remnant lipoproteins and plasma concentrations of remnant cholesterol, and several genes, including APOC3, ANGPTL3 and ANGPTL4, whose gene products regulate activity of LPL, are important determinants of remnant cholesterol. SUMMARY Current opinion is that remnant cholesterol is a likely causal factor in the development of ASCVD. Human genetic studies have identified several genes, many involved in LPL function, affecting remnant cholesterol concentrations, some of which are already used as therapeutic targets, and others which are subject to investigation of their remnant cholesterol and triglyceride-lowering effect in clinical trials.
Collapse
Affiliation(s)
- Anders B Wulff
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Balling M, Afzal S, Varbo A, Nordestgaard BG, Langsted A. Remnant Cholesterol: Quantification, Concentrations by Sex and Age, and Risk of Ischemic Heart Disease. Clin Chem 2025; 71:451-462. [PMID: 39723642 DOI: 10.1093/clinchem/hvae217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/22/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Observational and genetic causal studies have shown an association between high concentrations of remnant cholesterol and increased risk of ischemic heart disease. However, findings from randomized intervention trials that reduced plasma triglycerides, a surrogate marker of remnant cholesterol, have been conflicting. The exact mechanisms by which remnant cholesterol contributes to atherosclerosis and, ultimately, ischemic heart disease remain incompletely understood. Additionally, insight on sex and age differences and the importance of measurement differences of remnant cholesterol in plasma concentrations and risk of ischemic heart disease are sparse. CONTENT This review covers current knowledge regarding remnant cholesterol and its role in ischemic heart disease, with particular attention to measurement and sex- and age-specific differences. SUMMARY Findings from observational, genetic, and mechanistic studies support the notion that higher remnant cholesterol may be an important cause of ischemic heart disease in both women and men. Concentrations of remnant cholesterol vary by age, with a sharp increase at early adulthood for men and around menopause for women. Remnant cholesterol can be calculated from a standard lipid profile and in addition measured directly using manual ultracentrifugation, automated assays, and nuclear magnetic resonance spectroscopy. Irrespective of the method used to assess plasma concentrations, high concentrations of remnant cholesterol are consistently associated with increased risk of myocardial infarction and ischemic heart disease in observational and genetic causal studies; cholesterol rather than triglycerides in remnants drive this risk. Importantly, results from ongoing randomized clinical trials aiming specifically at lowering remnant cholesterol and ischemic heart disease are eagerly awaited.
Collapse
Affiliation(s)
- Mie Balling
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Shoaib Afzal
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anette Varbo
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Langsted
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
13
|
Gagnon E, Gill D, Chabot D, Cronjé HT, Yuan S, Brennan S, Thériault S, Burgess S, Arsenault BJ, Dib MJ. Evaluating the Cardiometabolic Efficacy and Safety of Lipoprotein Lipase Pathway Targets in Combination With Approved Lipid-Lowering Targets: A Drug Target Mendelian Randomization Study. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025; 18:e004933. [PMID: 40052268 PMCID: PMC7617573 DOI: 10.1161/circgen.124.004933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/29/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Therapies targeting the LPL (lipoprotein lipase) pathway are under development for cardiometabolic disease. Insights into their efficacy-both alone and in combination with existing lipid-lowering therapies-modes of action, and safety of these agents are essential to inform clinical development. Using Mendelian randomization, we aimed to (1) evaluate efficacy, (2) explore shared mechanisms, (3) assess additive effects with approved lipid-lowering drugs, and (4) identify secondary indications and potential adverse effects. METHODS We selected triglyceride-lowering genetic variants located in the genes encoding ANGPTL3 (angiopoietin-like 3), ANGPTL4 (angiopoietin-like 4), APOC3 (apolipoprotein C3), and LPL and conducted drug target Mendelian randomization on primary outcomes including coronary artery disease and type 2 diabetes, and secondary outcomes, including apolipoprotein B, waist-to-hip ratio, body mass index, and 233 metabolic biomarkers. We conducted interaction Mendelian randomization analyses in 488 139 UK Biobank participants to test the effect of combination therapy targeting the LPL and LDLR (low-density lipoprotein receptor) pathways. Finally, we investigated potential secondary indications and adverse effects by leveraging genetic association data on 1204 disease end points. RESULTS Genetically predicted triglyceride lowering through the perturbation of LPL pathway activation targets ANGPTL4, APOC3, and LPL was associated with a lower risk of coronary artery disease and type 2 diabetes and lower apolipoprotein B. Genetically predicted triglyceride lowering through ANGPTL4 was associated with a lower waist-to-hip ratio, suggestive of a favorable body fat distribution. There was no evidence of a multiplicative interaction between genetically proxied perturbation of ANGPTL4, APOC3, and LPL and that of HMGCR (HMG-CoA reductase) and PCSK9 (proprotein convertase subtilisin/kexin type 9) on coronary artery disease and type 2 diabetes, consistent with additive effects. Finally, associations of genetically predicted LPL pathway targeting were supportive of the broad safety of these targets. CONCLUSIONS Our findings provide genetic evidence supporting the efficacy and safety of LPL pathway activation therapies for the prevention of coronary artery disease and type 2 diabetes, alone or in combination with statins or PCSK9 inhibitors.
Collapse
Affiliation(s)
- Eloi Gagnon
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec (QC), Canada
| | - Dipender Gill
- Sequoia Genetics LTD., Translation & Innovation Hub, 84 Wood Lane, London, England
| | - Dominique Chabot
- Department of Medicine, Faculty of Medicine, Université Laval, Québec (QC), Canada
| | - Héléne T. Cronjé
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Denmark
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stephen Brennan
- Department of Neurology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Sébastien Thériault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec (QC), Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec (QC), Canada
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Benoit J. Arsenault
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec (QC), Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec (QC), Canada
| | - Marie-Joe Dib
- Cardiovascular Division, Perelman School for Advanced Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
14
|
Gagnon E, Arsenault BJ. Leveraging drug-target Mendelian randomization for tailored lipoprotein-lipid lowering. Curr Opin Lipidol 2025; 36:71-77. [PMID: 39973804 DOI: 10.1097/mol.0000000000000977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
PURPOSE OF REVIEW The study of naturally occurring genetic variation in human populations has laid the foundation for proprotein converts subtilisin/kexin type 9 inhibitors, and more recently new classes of lipid-lowering drugs such as lipoprotein(a) inhibitors and lipoprotein lipase pathway activators. These emerging therapies lower plasma lipoprotein-lipid levels that are not adequately managed by traditional low-density lipoprotein (LDL) cholesterol-lowering medications. By targeting different risk factors, these therapies could help manage the important residual cardiovascular risk of LDL cholesterol medications. RECENT FINDINGS We review the latest insights into the pharmacological and genetic modulation of these new therapeutic targets. We highlight that the drugs remarkably recapitulate the lipid effects observed in genetic studies. In addition to lowering lipoprotein-lipid levels, robust genetic evidence support that these drugs may prevent cardiometabolic outcomes. SUMMARY Emerging lipid-lowering therapies could launch a new era for preventive medicine in which treatments are optimally tailored to patient's lipoprotein-lipid profiles.
Collapse
Affiliation(s)
- Eloi Gagnon
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| |
Collapse
|
15
|
Kumari A, Larsen SWR, Bondesen S, Qian Y, Tian HD, Walker SG, Davies BSJ, Remaley AT, Young SG, Konrad RJ, Jørgensen TJD, Ploug M. ANGPTL3/8 is an atypical unfoldase that regulates intravascular lipolysis by catalyzing unfolding of lipoprotein lipase. Proc Natl Acad Sci U S A 2025; 122:e2420721122. [PMID: 40112106 PMCID: PMC11962473 DOI: 10.1073/pnas.2420721122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
Lipoprotein lipase (LPL) carries out the lipolytic processing of triglyceride-rich lipoproteins (TRL) along the luminal surface of capillaries. LPL activity is regulated by the angiopoietin-like proteins (ANGPTL3, ANGPTL4, ANGPTL8), which control the delivery of TRL-derived lipid nutrients to tissues in a temporal and spatial fashion. This regulation of LPL mediates the partitioning of lipid delivery to adipose tissue and striated muscle according to nutritional status. A complex between ANGPTL3 and ANGPTL8 (ANGPTL3/8) inhibits LPL activity in oxidative tissues, but its mode of action has remained unknown. Here, we used biophysical techniques to define how ANGPTL3/8 and ANGPTL3 interact with LPL and how they drive LPL inactivation. We demonstrate, by mass photometry, that ANGPTL3/8 is a heterotrimer with a 2:1 ANGPTL3:ANGPTL8 stoichiometry and that ANGPTL3 is a homotrimer. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) studies revealed that ANGPTL3/8 and ANGPTL3 use the proximal portion of their N-terminal α-helices to interact with sequences surrounding the catalytic pocket in LPL. That binding event triggers unfolding of LPL's α/β-hydrolase domain and irreversible loss of LPL catalytic activity. The binding of LPL to its endothelial transporter protein (GPIHBP1) or to heparan-sulfate proteoglycans protects LPL from unfolding and inactivation, particularly against the unfolding triggered by ANGPTL3. Pulse-labeling HDX-MS studies revealed that ANGPTL3/8 and ANGPTL3 catalyze LPL unfolding in an ATP-independent fashion, which categorizes these LPL inhibitors as atypical unfoldases. The catalytic nature of LPL unfolding by ANGPTL3/8 explains why low plasma concentrations of ANGPTL3/8 are effective in inhibiting a molar excess of LPL in capillaries.
Collapse
Affiliation(s)
- Anni Kumari
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, Copenhagen NDK–2200, Denmark
- Finsen Laboratory, Biotechnology Research and Innovation Centre, University of Copenhagen, Copenhagen NDK-2200, Denmark
| | - Sanne W. R. Larsen
- Finsen Laboratory, Biotechnology Research and Innovation Centre, University of Copenhagen, Copenhagen NDK-2200, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense MDK–5320, Denmark
| | - Signe Bondesen
- Finsen Laboratory, Biotechnology Research and Innovation Centre, University of Copenhagen, Copenhagen NDK-2200, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense MDK–5320, Denmark
| | - Yuewei Qian
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN462585
| | - Hao D. Tian
- Laboratory of Lipoprotein Metabolism, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20814
| | - Sydney G. Walker
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa, IA52242
| | - Brandon S. J. Davies
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa, IA52242
| | - Alan T. Remaley
- Laboratory of Lipoprotein Metabolism, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20814
| | - Stephen G. Young
- Department of Medicine, University of California, Los Angeles, CA90095
- Department of Human Genetics, University of California, Los Angeles, CA90095
| | - Robert J. Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN462585
| | - Thomas J. D. Jørgensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense MDK–5320, Denmark
| | - Michael Ploug
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, Copenhagen NDK–2200, Denmark
- Finsen Laboratory, Biotechnology Research and Innovation Centre, University of Copenhagen, Copenhagen NDK-2200, Denmark
| |
Collapse
|
16
|
Murdock DR, Guo DC, DePaolo JS, Schwarze U, Duan XY, Cecchi AC, Marin IC, Tang Y, Chong JX, Bamshad MJ, Leppig KA, Byers PH, Damrauer SM, Milewicz DM. Non-canonical splice variants in thoracic aortic dissection cases and Marfan syndrome with negative genetic testing. NPJ Genom Med 2025; 10:25. [PMID: 40118890 PMCID: PMC11928670 DOI: 10.1038/s41525-025-00472-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/27/2025] [Indexed: 03/24/2025] Open
Abstract
Individuals with heritable thoracic aortic disease (HTAD) face a high risk of deadly aortic dissections, but genetic testing identifies causative variants in only a minority of cases. We explored the contribution of non-canonical splice variants (NCVAS) to thoracic aortic disease (TAD) using SpliceAI and sequencing data from diverse cohorts, including 551 early-onset sporadic dissection cases and 437 HTAD probands with exome sequencing, 57 HTAD pedigrees with whole genome sequencing, and select sporadic cases with clinical panel testing. NCVAS were identified in syndromic HTAD genes such as FBN1, SMAD3, and COL3A1, including intronic variants in FBN1 in two Marfan syndrome (MFS) families. Validation in the Penn Medicine BioBank and UK Biobank showed enrichment of NCVAS in HTAD-associated genes among dissections. These findings suggest NCVAS are an underrecognized contributor to TAD, particularly in sporadic dissection and unsolved MFS cases, highlighting the potential of advanced splice prediction tools in genetic diagnostics.
Collapse
Affiliation(s)
- David R Murdock
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Dong-Chuan Guo
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - John S DePaolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ulrike Schwarze
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Xue-Yan Duan
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Alana C Cecchi
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Isabella C Marin
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - YingYing Tang
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, New York, NY, USA
| | - Jessica X Chong
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Michael J Bamshad
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Kathleen A Leppig
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Peter H Byers
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Scott M Damrauer
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Dianna M Milewicz
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA.
| |
Collapse
|
17
|
Qi Y, Jiang H, Lun Y, Gang Q, Shen S, Zhang H, Liu M, Wang Y, Zhang J. Protein Drug Targets for Abdominal Aortic Aneurysm and Proteomic Associations Between Modifiable Risk Factors and Abdominal Aortic Aneurysm. J Am Heart Assoc 2025; 14:e037802. [PMID: 40008516 DOI: 10.1161/jaha.124.037802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a severe aortic disease for which no pharmacological interventions have yet been developed. This investigation focused on identifying protein-based therapeutic targets and assessing how proteins mediate the interplay between modifiable risk factors and AAA development. METHODS Causal inferences between plasma proteins and AAA were drawn using 2-sample Mendelian randomization, followed by comprehensive sensitivity testing, colocalization, and replication efforts. Further analyses included database interrogation, single-cell RNA data analysis, enrichment analysis, protein-protein interaction networks, and immunohistochemistry to map the tissue-specific expression of these proteins, their expression within AAA tissues, and their biological roles. Mediation Mendelian randomization was employed to evaluate the mediating effects of AAA-related proteins on the associations between AAA and 3 risk factors: hypertension, smoking, and obesity. RESULTS A total of 43 proteins were identified as having causal links to AAA. Colocalization analysis pinpointed 13 proteins with strong evidence of colocalization with AAA. Of these, the causal involvement of 10 proteins was substantiated by external validation data. Consistent evidence for PCSK9 (proprotein convertase subtilisin/kexin type 9), IL6R (interleukin-6R), ECM1 (extracellular matrix protein 1), and ANGPTL4 (angiopoietin-related protein 4) was further validated through tissue immunohistochemistry and blood data. Moreover, Mendelian randomization analysis identified 10 proteins as mediators of the influence of hypertension, smoking, and obesity on AAA development. CONCLUSIONS This analysis identifies 4 proteins (PCSK9, IL6R, ECM1, and ANGPTL4) as high-priority therapeutic targets for AAA and emphasizes the intermediary role of plasma proteins in linking hypertension, smoking, obesity, and AAA. Further investigations are needed to clarify the specific roles of these proteins in AAA pathology.
Collapse
Affiliation(s)
- Yao Qi
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Han Jiang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Yu Lun
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Qingwei Gang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Shikai Shen
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Han Zhang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Mingyu Liu
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Yixian Wang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Jian Zhang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| |
Collapse
|
18
|
Xu D, Lu J, Yang Y, Hu W, Chen J, Xue J, Yang S, Cao N, Hu H, Qian N, Zhou D, Dai H, Wang J, Liu X. Identifying novel drug targets for calcific aortic valve disease through Mendelian randomization. Atherosclerosis 2025; 402:119110. [PMID: 39922081 DOI: 10.1016/j.atherosclerosis.2025.119110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/10/2024] [Accepted: 01/26/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND AND AIMS Calcific aortic valve disease (CAVD) is characterized by progressive leaflet thickening and calcification, with no available pharmacological treatments. Plasma proteins play a pivotal role in disease regulation. This study aimed to uncover novel therapeutic targets for CAVD using Mendelian randomization (MR) integrated with transcriptomic analysis. METHODS Protein quantitative trait loci (pQTL) from the deCODE and UK Biobank Pharma Proteomics Project (UKB-PPP) plasma protein databases were used as exposure data. The FinnGen cohort (9870 cases, 402,311 controls) served as the discovery set, while the TARGET cohort (13,765 cases, 640,102 controls) provided validation. MR and summary data-based Mendelian randomization (SMR) were employed to screen for potential causal targets of CAVD. Colocalization analysis was conducted to assess whether CAVD and target proteins shared common causal SNPs. Additional analyses included trancriptomic profiling at multiple RNA levels. Protein-level validation was conducted via Western blot and immunostaining. RESULTS Six proteins (ANGPTL4, PCSK9, ITGAV, CTSB, GNPTG, and FURIN) with strong genetic colocalization were identified by MR and SMR analysis. Among these, cellular trancriptomic analysis revealed ANGPTL4 and ITGAV with significantly greater expression in osteogenic group, which was further validated in calcified aortic valves and osteogenic valvular interstitial cells in protein level. CONCLUSIONS This study identified six causal proteins with strong genetic colocalization for CAVD, with ANGPTL4 and ITGAV emerging as the most promising targets for further investigation.
Collapse
Affiliation(s)
- Dilin Xu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Jin Lu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Yanfang Yang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Wangxing Hu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Jinyong Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Junhui Xue
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Shuangshuang Yang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Naifang Cao
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Haochang Hu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Ningjing Qian
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Dao Zhou
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Hanyi Dai
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China
| | - Jian'an Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, PR China.
| | - Xianbao Liu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, PR China; State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, PR China; Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, PR China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, PR China.
| |
Collapse
|
19
|
Fang Z, Liu C, Cheng Y, Ji Y, Liu C. Combined analysis of bulk, single-cell RNA sequencing, and spatial transcriptomics reveals the expression patterns of lipid metabolism and ferroptosis in the immune microenvironment of metabolic-associated fatty liver disease. Life Sci 2025; 362:123377. [PMID: 39793853 DOI: 10.1016/j.lfs.2025.123377] [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: 09/19/2024] [Revised: 12/20/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
AIMS This study aims to identify key biomarkers associated with ferroptosis and lipid metabolism and investigate their roles in the progression of metabolic dysfunction-associated fatty liver disease (MAFLD). It further explores interactions between these biomarkers and the immune-infiltration environment, shedding light on how ferroptosis and lipid metabolism influence immune dynamics in MAFLD. MAIN METHODS Single-cell RNA sequencing data from liver samples were analyzed to evaluate expression variations related to ferroptosis and lipid metabolism in MAFLD patients. Gene scores were assessed to explore their impact on the immune microenvironment, particularly hepatocyte-macrophage communication. Weighted Gene Co-expression Network Analysis (WGCNA) was applied to Bulk-RNA-Seq data to identify gene clusters associated with ferroptosis and lipid metabolism. The analyses were integrated into a risk assessment system and predictive model, with validation conducted through in vivo experiments. KEY FINDINGS Integration of single-cell and WGCNA data identified 11 key genes linked to ferroptosis and lipid metabolism (e.g., IER5L, SOCS2, KLF9), significantly influencing the liver's immune microenvironment. The risk assessment system and predictive model achieved an AUC of 0.92 and revealed distinct immune and biological characteristics in MAFLD patients across risk levels. The expression patterns and biological roles of these genes were confirmed in in vivo studies. SIGNIFICANCE This study establishes a strong link between ferroptosis- and lipid metabolism-related gene expression and MAFLD's complexity. It provides novel insights into disease mechanisms, supporting personalized prognosis and targeted therapeutic strategies for MAFLD patients.
Collapse
Affiliation(s)
- Zhihao Fang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Changxu Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Cheng
- Cardiovascular Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanchao Ji
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
20
|
Li G, Zhao H, Cheng Z, Liu J, Li G, Guo Y. Single-cell transcriptomic profiling of heart reveals ANGPTL4 linking fibroblasts and angiogenesis in heart failure with preserved ejection fraction. J Adv Res 2025; 68:215-230. [PMID: 38346487 PMCID: PMC11785561 DOI: 10.1016/j.jare.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/19/2024] Open
Abstract
INTRODUCTION Despite the high morbidity and mortality, the effective therapies for heart failure with preserved fraction (HFpEF) are limited as the poor understand of its pathophysiological basis. OBJECTIVE This study was aimed to characterize the cellular heterogeneity and potential mechanisms of HFpEF at single-cell resolution. METHODS An HFpEF mouse model was induced by a high-fat diet with N-nitro-L-arginine methyl ester. Cells from the hearts were subjected to single-cell sequencing. The key protein expression was measured with Immunohistochemistry and immunofluorescence staining. RESULTS In HFpEF hearts, myocardial fibroblasts exhibited higher levels of fibrosis. Furthermore, an increased number of fibroblasts differentiated into high-metabolism and high-fibrosis phenotypes. The expression levels of genes encoding certain pro-angiogenic secreted proteins were decreased in the HFpEF group, as confirmed by bulk RNA sequencing. Additionally, the proportion of the endothelial cell (EC) lineages in the HFpEF group was significantly downregulated, with low angiogenesis and high apoptosis phenotypes observed in these EC lineages. Interestingly, the fibroblasts in the HFpEF heart might cross-link with the EC lineages via over-secretion of ANGPTL4, thus displaying an anti-angiogenic function. Immunohistochemistry and immunofluorescence staining then revealed the downregulation of vascular density and upregulation of ANGPTL4 expression in HFpEF hearts. Finally, we predicted ANGPTL4as a potential druggable target using DrugnomeAI. CONCLUSION In conclusion, this study comprehensively characterized the angiogenesis impairment in HFpEF hearts at single-cell resolution and proposed that ANGPTL4 secretion by fibroblasts may be a potential mechanism underlying this angiogenic abnormality.
Collapse
Affiliation(s)
- Guoxing Li
- Institute of Life Sciences, Chongqing Medical University, 400016, China
| | - Huilin Zhao
- Institute of Life Sciences, Chongqing Medical University, 400016, China
| | - Zhe Cheng
- Department of Cardiology, Chongqing University Three Gorges Hospital, Chongqing 404199, China
| | - Junjin Liu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Gang Li
- Institute of Life Sciences, Chongqing Medical University, 400016, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, 400016, China.
| | - Yongzheng Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
21
|
Silbernagel G, Chen YQ, Li H, Lemen D, Wen Y, Zhen EY, Rief M, Kleber ME, Delgado GE, Sarzynski MA, Qian YW, Schmidt B, Erbel R, Trampisch US, Moissl AP, Rudolf H, Schunkert H, Stang A, März W, Trampisch HJ, Scharnagl H, Konrad RJ. Associations of Circulating ANGPTL3, C-Terminal Domain-Containing ANGPTL4, and ANGPTL3/8 and ANGPTL4/8 Complexes with LPL Activity, Diabetes, Inflammation, and Cardiovascular Mortality. Circulation 2025; 151:218-234. [PMID: 39392008 DOI: 10.1161/circulationaha.124.069272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND ANGPTL3/4/8 (angiopoietin-like proteins 3, 4, and 8) are important regulators of LPL (lipoprotein lipase). ANGPTL8 forms complexes with ANGPTL3 and ANGPTL4. ANGPTL4/8 complex formation converts ANGPTL4 from a furin substrate to a plasmin substrate, and both cleavages generate similar C-terminal domain-containing (CD)-ANGPTL4 fragments. Whereas several studies have investigated associations of free ANGPTL proteins with cardiovascular risk, there are no data describing associations of the complexes and CD-ANGPTL4 with outcomes or describing the effects of the complexes on LPL bound to GPIHBP1 (glycosylphosphatidylinositol HDL-binding protein 1). METHODS Recombinant protein assays were used to study ANGPTL protein and complex effects on GPIHBP1-LPL activity. ANGPTL3/8, ANGPTL3, ANGPTL4/8, and CD-ANGPTL4 were measured with dedicated immunoassays in 2394 LURIC (Ludwigshafen Risk and Cardiovascular Health) study participants undergoing coronary angiography and 6188 getABI study (German Epidemiological Trial on Ankle Brachial Index) participants undergoing ankle brachial index measurement. There was a follow-up for cardiovascular death with a median (interquartile range) duration of 9.80 (8.75-10.40) years in the LURIC study and 7.06 (7.00-7.14) years in the getABI study. RESULTS ANGPTL3/8 potently inhibited GPIHBP1-LPL activity and showed positive associations with LDL-C (low-density lipoprotein cholesterol) and triglycerides (both P<0.001). However, in neither study did ANGPTL3/8 correlate with cardiovascular death. Free ANGPTL3 was positively associated with cardiovascular death in the getABI study but not the LURIC study. ANGPTL4/8 and especially CD-ANGPTL4 were positively associated with the prevalence of diabetes, CRP (C-reactive protein; all P<0.001), and cardiovascular death in both studies. In the LURIC and getABI studies, respective hazard ratios for cardiovascular mortality comparing the third with the first ANGPTL4/8 tertile were 1.47 (1.15-1.88) and 1.68 (1.25-2.27) when adjusted for sex, age, body mass index, and diabetes. For CD-ANGPTL4, these hazard ratios were 2.44 (1.86-3.20) and 2.76 (2.00-3.82). CONCLUSIONS ANGPTL3/8 potently inhibited GPIHBP1-LPL enzymatic activity, consistent with its positive association with serum lipids. However, ANGPTL3/8, LDL-C, and triglyceride levels were not associated with cardiovascular death in the LURIC and getABI cohorts. In contrast, concentrations of ANGPTL4/8 and particularly CD-ANGPTL4 were positively associated with inflammation, the prevalence of diabetes, and cardiovascular mortality.
Collapse
Affiliation(s)
- Günther Silbernagel
- Division of Angiology, Department of Internal Medicine (G.S.), Medical University of Graz, Austria
| | - Yan Q Chen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Hongxia Li
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Deven Lemen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Eugene Y Zhen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Martin Rief
- Anaesthesiology and Intensive Care Medicine (M.R.), Medical University of Graz, Austria
| | - Marcus E Kleber
- 5th Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (W.M., M.E.K., G.E.D.)
- Synlab Medizinisches Versorgungszentrum Humangenetik Mannheim, GmbH, Germany (M.E.K.)
| | - Graciela E Delgado
- 5th Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (W.M., M.E.K., G.E.D.)
| | - Mark A Sarzynski
- Department of Exercise Science, University of South Carolina, Columbia (M.A.S.)
| | - Yue-Wei Qian
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Boerge Schmidt
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Germany (A.S., B.S., R.E.)
| | - Raimund Erbel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Germany (A.S., B.S., R.E.)
| | - Ulrike S Trampisch
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr University, Bochum, Germany (U.S.T., H.J.T.)
| | - Angela P Moissl
- Institute of Nutritional Sciences Friedrich Schiller University and Competence Cluster for Nutrition and Cardiovascular Health, Halle-Jena-Leipzig, Jena, Germany (A.P.M.)
| | - Henrik Rudolf
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University, Germany (H.R.)
| | - Heribert Schunkert
- Department of Cardiology, German Heart Center Munich, Technical University of Munich and Partner Site Munich Heart Alliance, German Center for Cardiovascular Disease, Germany (H.S.)
| | - Andreas Stang
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Germany (A.S., B.S., R.E.)
- School of Public Health, Department of Epidemiology, Boston University, MA (A.S.)
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., H.S.), Medical University of Graz, Austria
- 5th Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (W.M., M.E.K., G.E.D.)
- Synlab Academy, Synlab Holding Germany, Mannheim (W.M.)
| | - Hans J Trampisch
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr University, Bochum, Germany (U.S.T., H.J.T.)
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., H.S.), Medical University of Graz, Austria
| | - Robert J Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| |
Collapse
|
22
|
Li Y, Zhang Y, Cao M, Yuan T, Ou S. Angiopoietin-like protein 4 dysregulation in kidney diseases: a promising biomarker and therapeutic target. Front Pharmacol 2025; 15:1475198. [PMID: 39840089 PMCID: PMC11747783 DOI: 10.3389/fphar.2024.1475198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
The global burden of renal diseases is increasingly severe, underscoring the need for in-depth exploration of the molecular mechanisms underlying renal disease progression and the development of potential novel biomarkers or therapeutic targets. Angiopoietin-like protein 4 (ANGPTL4) is a multifunctional cytokine involved in the regulation of key biological processes, such as glucose and lipid metabolism, inflammation, vascular permeability, and angiogenesis, all of which play crucial roles in the pathogenesis of kidney diseases. Over the past 2 decades, ANGPTL4 has been regarded as playing a pivotal role in the progression of various kidney diseases, prompting significant interest from the scientific community regarding its potential clinical utility in renal disorders. This review synthesizes the available literature, provides a concise overview of the molecular biological effects of ANGPTL4, and highlights its relationship with multiple renal diseases and recent research advancements. These findings underscore the important gaps that warrant further investigation to develop novel targets for the prediction or treatment of various renal diseases.
Collapse
Affiliation(s)
- Yan Li
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephrology, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Yuxin Zhang
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephrology, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Mengxia Cao
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephrology, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Tingting Yuan
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephrology, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Santao Ou
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephrology, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| |
Collapse
|
23
|
Ko S, Anzai A, Liu X, Kinouchi K, Yamanoi K, Torimitsu T, Ichihara G, Kitakata H, Shirakawa K, Katsumata Y, Endo J, Hayashi K, Yoshida M, Nishimori K, Tanaka KF, Onaka T, Sano M, Ieda M. Social Bonds Retain Oxytocin-Mediated Brain-Liver Axis to Retard Atherosclerosis. Circ Res 2025; 136:78-90. [PMID: 39601150 DOI: 10.1161/circresaha.124.324638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUNDS Social interaction with others is essential to life. Although social isolation and loneliness have been implicated as increased risks of cardiometabolic and cardiovascular diseases and all-cause mortality, the cellular and molecular mechanisms by which social connection maintains cardiometabolic and cardiovascular health remain largely unresolved. METHODS To investigate how social connection protects against cardiometabolic and cardiovascular diseases, atherosclerosis-prone, high-fat diet-fed Apoe-/- mouse siblings were randomly assigned to either individual or grouped housing for 12 weeks. Histological, flow cytometric, biochemical, gene, and protein analyses were performed to assess atherosclerotic lesions, systemic metabolism, inflammation, and stress response. The effects of oxytocin on hepatocytes and subsequent cardiometabolic and cardiovascular function were investigated by in vivo and in vitro approaches. RESULTS Apoe-/- mice housed individually developed larger vulnerable atherosclerotic lesions by disrupted lipid metabolism compared with those of mice in regular group housing, irrespective of body weight, eating behavior, feeding conditions, sympathetic nervous activity, glucocorticoid response, or systemic inflammation. Mechanistically, the chronic isolation reduced the hypothalamic production of oxytocin, which controls bile acid production and LPL (lipoprotein lipase) activity through the peripheral OXTR (oxytocin receptor) in hepatocytes, whose downstream targets include Cyp7a1, Angptl4, and Angptl8. While hepatocyte-specific OXTR-null mice and mice receiving adeno-associated virus targeting OXTR on hepatocytes led to severe dyslipidemia and aggravated atherosclerosis, oral oxytocin supplementation to socially isolated mice, but not to hepatocyte-specific OXTR conditional knockout mice, improved lipid profiles and retarded atherosclerosis development. CONCLUSIONS These results identify a novel brain-liver axis that links sociality to hepatic lipid metabolism, thus proposing a potential therapeutic strategy for loneliness-associated atherosclerosis progression.
Collapse
Affiliation(s)
- Seien Ko
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Anzai
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Xueyuan Liu
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Kenichiro Kinouchi
- Department of Endocrinology, Metabolism, and Nephrology (K.K., T.T., K.H.), Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiro Yamanoi
- Department of Pathology (K.Y.), Keio University School of Medicine, Tokyo, Japan
| | - Takuto Torimitsu
- Department of Endocrinology, Metabolism, and Nephrology (K.K., T.T., K.H.), Keio University School of Medicine, Tokyo, Japan
| | - Genki Ichihara
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Hiroki Kitakata
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Kohsuke Shirakawa
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Yoshinori Katsumata
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Jin Endo
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| | - Kaori Hayashi
- Department of Endocrinology, Metabolism, and Nephrology (K.K., T.T., K.H.), Keio University School of Medicine, Tokyo, Japan
| | - Masahide Yoshida
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan (M.Y., T.O.)
| | - Katsuhiko Nishimori
- Department of Obesity and Internal Inflammation, Fukushima Medical University, Japan (K.N.)
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research (K.F.T.), Keio University School of Medicine, Tokyo, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Tochigi, Japan (M.Y., T.O.)
| | - Motoaki Sano
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Japan (M.S.)
| | - Masaki Ieda
- Department of Cardiology (S.K., A.A., X.L., G.I., H.K., K.S., Y.K., J.E., M.S., M.I.), Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Ispir E, Saruhan E, Topcu DI, Varol B, Akbaba E, Cakmak T. Relationship between serum levels of ANGPTL8, Apo C2, and human placental lactogen (hPL) in patients with gestational diabetes mellitus: Interaction of LPL regulators with hPL, a possible contributing factor to insulin resistance. Placenta 2025; 159:119-125. [PMID: 39700905 DOI: 10.1016/j.placenta.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 11/05/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM) is defined as glucose intolerance during pregnancy. We aimed to investigate the potential effects of betatrophin and ApoC2 in GDM, focusing on their roles in LPL (lipoprotein lipase) regulation and their relationship with hPL to elucidate the possible impact of hPL on lipid metabolism and its potential contribution to the development of GDM. METHODS Thirty pregnant women with normal glucose tolerance and 29 with gestational diabetes mellitus (diagnosed by 75g OGTT between 24 and 28 weeks) were included in the study. Serum betatrophin, hPL, and ApoC2 were measured by Elisa and HOMA-IR was calculated. RESULTS In the GDM group, hPL levels correlated with betatrophin and ApoC2 (r = 0.552, p < 0.05; r = 0.588, p < 0.05 respectively) while betatrophin correlated with the ApoC2 (r = 0.584, p < 0.05). A linear relationship between hPL and betatropin and also between hPL and ApoC2 values in the control group (r = 0.454, p < 0.05; r = 0.779, p < 0.01 respectively) were observed. ApoC2 levels in the GDM group (n = 20) with HOMA-IR cut-off >2.5 were significantly higher than the control group (n = 10) (p < 0.05). There was also a positive relationship between betatrophin and ApoC2 (r = 0.591) (p < 0.05). DISCUSSION GDM patients may have impaired LPL enzyme regulation in addition to insulin resistance, with hPL potentially contributing to this disruption. Impaired lipoprotein lipase activity and its dysregulation secondary to genetic disorders may play a role in the etiopathogenesis of GDM. Further investigation into the correlation between betatrophin, ApoC2, and other LPL modulators in patients with various forms of diabetes could be beneficial for understanding this interaction more comprehensively.
Collapse
Affiliation(s)
- Emre Ispir
- Department of Medical Biochemistry, Bozyaka Research and Training Hospital, Izmir, Turkey.
| | - Ercan Saruhan
- Department of Medical Biochemistry, Faculty of Medicine, Mugla Sitki Kocman University, Mugla, Turkey.
| | - Deniz Ilhan Topcu
- Department of Medical Biochemistry, Izmir City Hospital, Izmir, Turkey.
| | - Bugra Varol
- Adnan Menderes University, Institute of Health Sciences, Division of Biostatistics, Aydin, Turkey.
| | - Eren Akbaba
- Department of Obstetrics and Gynecology, Faculty of Medicine, Mugla Sitki Kocman University, Mugla, Turkey.
| | - Tuba Cakmak
- Department of Medical Biochemistry, Bozyaka Research and Training Hospital, Izmir, Turkey.
| |
Collapse
|
25
|
Modder M, het Panhuis WI, Li M, Afkir S, Dorn AL, Pronk ACM, Streefland TCM, Lalai RA, Pierrou S, Nilsson SK, Olivecrona G, Kooijman S, Rensen PCN, Schönke M. Liver-targeted Angptl4 silencing by antisense oligonucleotide treatment attenuates hyperlipidaemia and atherosclerosis development in APOE*3-Leiden.CETP mice. Cardiovasc Res 2024; 120:2179-2190. [PMID: 39259836 PMCID: PMC11687395 DOI: 10.1093/cvr/cvae195] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 09/13/2024] Open
Abstract
AIMS Angiopoietin-like 3 (ANGPTL3) and 4 (ANGPTL4) inhibit lipoprotein lipase to regulate tissue fatty acid (FA) uptake from triglyceride (TG)-rich lipoproteins such as very low density lipoproteins (VLDL). While pharmacological inhibition of ANGPTL3 is being evaluated as a lipid-lowering strategy, systemic ANGPTL4 inhibition is not pursued due to adverse effects. This study aims to compare the therapeutic potential of liver-specific Angptl3 and Angptl4 silencing to attenuate hyperlipidemia and atherosclerosis development in APOE*3-Leiden.CETP mice, a well-established humanized model for lipoprotein metabolism. METHODS AND RESULTS Mice were subcutaneously injected twice per week with saline or liver-targeted antisense oligonucleotides against Angptl3, Angptl4, both, or a scrambled oligonucleotide. Plasma lipid levels, VLDL clearance, and hepatic VLDL production were determined, and atherosclerosis development was assessed. For toxicological evaluation, cynomolgus monkeys were treated with three dosages of liver-targeted ANGPTL4-silencing oligonucleotides. Liver-targeted Angptl4 silencing reduced plasma TGs (-48%) and total cholesterol (-56%), explained by higher VLDL-derived FA uptake by brown adipose tissue and lower VLDL production by the liver. Accordingly, Angptl4 silencing reduced atherosclerotic lesion size (-86%) and improved lesion stability. Hepatic Angptl3 silencing similarly attenuated hyperlipidemia and atherosclerosis development. While Angptl3 and Angptl4 silencing lowered plasma TGs in the refed and fasted state, respectively, combined Angptl3/4 silencing lowered plasma TGs independent of the nutritional state. In cynomolgus monkeys, anti-ANGPTL4 ASO treatment was well tolerated without adverse effects. CONCLUSION Liver-targeted Angptl4 silencing potently attenuates hyperlipidemia and atherosclerosis development in APOE*3-Leiden.CETP mice, and liver-targeted ANGPTL4 silencing is well tolerated in non-human primates. These data warrant further clinical development of liver-targeted ANGPTL4 silencing.
Collapse
Affiliation(s)
- Melanie Modder
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Wietse In het Panhuis
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Mohan Li
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Salwa Afkir
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Alexandra L Dorn
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Amanda C M Pronk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Trea C M Streefland
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Reshma A Lalai
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Stefan Pierrou
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36 Umeå, Sweden
| | - Stefan K Nilsson
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36 Umeå, Sweden
| | - Gunilla Olivecrona
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36 Umeå, Sweden
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
26
|
Srivastava SP, Zhou H, Shenoi R, Morris M, Lainez-Mas B, Goedeke L, Rajendran BK, Setia O, Aryal B, Kanasaki K, Koya D, Inoki K, Dardik A, Bell T, Fernández-Hernando C, Shulman GI, Goodwin JE. Renal Angptl4 is a key fibrogenic molecule in progressive diabetic kidney disease. SCIENCE ADVANCES 2024; 10:eadn6068. [PMID: 39630889 PMCID: PMC11616692 DOI: 10.1126/sciadv.adn6068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
Angiopoietin-like 4 (ANGPTL4), a key protein involved in lipoprotein metabolism, has diverse effects. There is an association between Angptl4 and diabetic kidney disease; however, this association has not been well investigated. We show that both podocyte- and tubule-specific ANGPTL4 are crucial fibrogenic molecules in diabetes. Diabetes accelerates the fibrogenic phenotype in control mice but not in ANGPTL4 mutant mice. The protective effect observed in ANGPTL4 mutant mice is correlated with a reduction in stimulator of interferon genes pathway activation, expression of pro-inflammatory cytokines, reduced epithelial-to-mesenchymal transition and endothelial-to-mesenchymal transition, lessened mitochondrial damage, and increased fatty acid oxidation. Mechanistically, we demonstrate that podocyte- or tubule-secreted Angptl4 interacts with Integrin β1 and influences the association between dipeptidyl-4 with Integrin β1. We demonstrate the utility of a targeted pharmacologic therapy that specifically inhibits Angptl4 gene expression in the kidneys and protects diabetic kidneys from proteinuria and fibrosis. Together, these data demonstrate that podocyte- and tubule-derived Angptl4 is fibrogenic in diabetic kidneys.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Han Zhou
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
| | - Rachel Shenoi
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Myshal Morris
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Begoña Lainez-Mas
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
| | - Leigh Goedeke
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ocean Setia
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT, USA
| | - Binod Aryal
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Keizo Kanasaki
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
- Department of Internal Medicine 1, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
- The Center for Integrated Kidney Research and Advance, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT, USA
| | | | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Gerald I. Shulman
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
27
|
Wen Y, Zhang X, Liu H, Ye H, Wang R, Ma C, Duo T, Wang J, Yang X, Yu M, Wang Y, Wu L, Zhao Y, Wang L. SGLT2 inhibitor downregulates ANGPTL4 to mitigate pathological aging of cardiomyocytes induced by type 2 diabetes. Cardiovasc Diabetol 2024; 23:430. [PMID: 39633372 PMCID: PMC11619200 DOI: 10.1186/s12933-024-02520-8] [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: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Senescence is recognized as a principal risk factor for cardiovascular diseases, with a significant association between the senescence of cardiomyocytes and inferior cardiac function. Furthermore, type 2 diabetes exacerbates this aging process. Sodium-glucose co-transporter 2 inhibitor (SGLT2i) has well-established cardiovascular benefits and, in recent years, has been posited to possess anti-aging properties. However, there are no reported data on their improvement of cardiomyocytes function through the alleviation of aging. Consequently, our study aims to investigate the mechanism by which SGLT2i exerts anti-aging and protective effects at the cardiac level through its action on the FOXO1-ANGPTL4 pathway. METHODS To elucidate the underlying functions and mechanisms, we established both in vivo and in vitro disease models, utilizing mice with diabetic cardiomyopathy (DCM) induced by type 2 diabetes mellitus (T2DM) through high-fat diet combined with streptozotocin (STZ) administration, and AC16 human cardiomyocyte cell subjected to stimulation with high glucose (HG) and palmitic acid (PA). These models were employed to assess the changes in the senescence phenotype of cardiomyocytes and cardiac function following treatment with SGLT2i. Concurrently, we identified ANGPTL4, a key factor contributing to senescence in DCM, using RNA sequencing (RNA-seq) technology and bioinformatics methods. We further clarified ANGPTL4 role in promoting pathological aging of cardiomyocytes induced by hyperglycemia and hyperlipidemia through knockdown and overexpression of the factor, as well as analyzed the impact of SGLT2i intervention on ANGPTL4 expression. Additionally, we utilized chromatin immunoprecipitation followed by quantitative real-time PCR (ChIP-qPCR) to confirm that FOXO1 is essential for the transcriptional activation of ANGPTL4. RESULTS The therapeutic intervention with SGLT2i alleviated the senescence phenotype in cardiomyocytes of the DCM mouse model constructed by high-fat feeding combined with STZ, as well as in the AC16 model stimulated by HG and PA, while also improving cardiac function in DCM mice. We observed that the knockdown of ANGPTL4, a key senescence-promoting factor in DCM identified through RNA-seq technology and bioinformatics, mitigated the senescence of cardiomyocytes, whereas overexpression of ANGPTL4 exacerbated it. Moreover, SGLT2i improved the senescence phenotype by suppressing the overexpression of ANGPTL4. In fact, we discovered that SGLT2i exert their effects by regulating the upstream transcription factor FOXO1 of ANGPTL4. Under conditions of hyperglycemia and hyperlipidemia, compared to the control group without FOXO1, the overexpression of FOXO1 in conjunction with SGLT2i intervention significantly reduced both ANGPTL4 mRNA and protein levels. This suggests that the FOXO1-ANGPTL4 axis may be a potential target for the cardioprotective effects of SGLT2i. CONCLUSIONS Collectively, our study demonstrates that SGLT2i ameliorate the pathological aging of cardiomyocytes induced by a high glucose and high fat metabolic milieu by regulating the interaction between FOXO1 and ANGPTL4, thereby suppressing the transcriptional synthesis of the latter, and consequently restoring cardiac function.
Collapse
MESH Headings
- Sodium-Glucose Transporter 2 Inhibitors/pharmacology
- Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
- Diabetic Cardiomyopathies/blood
- Diabetic Cardiomyopathies/drug therapy
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/pathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/pathology
- Streptozocin/toxicity
- Diet, High-Fat/adverse effects
- Angiopoietin-Like Protein 4/antagonists & inhibitors
- Angiopoietin-Like Protein 4/genetics
- Angiopoietin-Like Protein 4/metabolism
- Cell Line
- Humans
- Animals
- Mice
- Cellular Senescence/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Gene Knockdown Techniques
- Mice, Inbred C57BL
- Male
- Forkhead Box Protein O1/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Down-Regulation/drug effects
- Blood Glucose/metabolism
Collapse
Affiliation(s)
- Yun Wen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xiaofang Zhang
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, China
| | - Han Liu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Haowen Ye
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ruxin Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Caixia Ma
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Tianqi Duo
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jiaxin Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xian Yang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Meixin Yu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Liangyan Wu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yongting Zhao
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lihong Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China.
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
28
|
Lanzer JD, Wienecke LM, Ramirez Flores RO, Zylla MM, Kley C, Hartmann N, Sicklinger F, Schultz JH, Frey N, Saez-Rodriguez J, Leuschner F. Single-cell transcriptomics reveal distinctive patterns of fibroblast activation in heart failure with preserved ejection fraction. Basic Res Cardiol 2024; 119:1001-1028. [PMID: 39311911 PMCID: PMC11628589 DOI: 10.1007/s00395-024-01074-w] [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: 12/12/2023] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 12/10/2024]
Abstract
Inflammation, fibrosis and metabolic stress critically promote heart failure with preserved ejection fraction (HFpEF). Exposure to high-fat diet and nitric oxide synthase inhibitor N[w]-nitro-l-arginine methyl ester (L-NAME) recapitulate features of HFpEF in mice. To identify disease-specific traits during adverse remodeling, we profiled interstitial cells in early murine HFpEF using single-cell RNAseq (scRNAseq). Diastolic dysfunction and perivascular fibrosis were accompanied by an activation of cardiac fibroblast and macrophage subsets. Integration of fibroblasts from HFpEF with two murine models for heart failure with reduced ejection fraction (HFrEF) identified a catalog of conserved fibroblast phenotypes across mouse models. Moreover, HFpEF-specific characteristics included induced metabolic, hypoxic and inflammatory transcription factors and pathways, including enhanced expression of Angiopoietin-like 4 (Angptl4) next to basement membrane compounds, such as collagen IV (Col4a1). Fibroblast activation was further dissected into transcriptional and compositional shifts and thereby highly responsive cell states for each HF model were identified. In contrast to HFrEF, where myofibroblast and matrifibrocyte activation were crucial features, we found that these cell states played a subsidiary role in early HFpEF. These disease-specific fibroblast signatures were corroborated in human myocardial bulk transcriptomes. Furthermore, we identified a potential cross-talk between macrophages and fibroblasts via SPP1 and TNFɑ with estimated fibroblast target genes including Col4a1 and Angptl4. Treatment with recombinant ANGPTL4 ameliorated the murine HFpEF phenotype and diastolic dysfunction by reducing collagen IV deposition from fibroblasts in vivo and in vitro. In line, ANGPTL4, was elevated in plasma samples of HFpEF patients and particularly high levels associated with a preserved global-longitudinal strain. Taken together, our study provides a comprehensive characterization of molecular fibroblast activation patterns in murine HFpEF, as well as the identification of Angiopoietin-like 4 as central mechanistic regulator with protective effects.
Collapse
Affiliation(s)
- Jan D Lanzer
- Institute for Computational Biomedicine, Heidelberg University, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
- Internal Medicine II, Heidelberg University Hospital, Heidelberg, Germany
- Informatics for Life, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Laura M Wienecke
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
- Department of Cardiology, Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Ricardo O Ramirez Flores
- Institute for Computational Biomedicine, Heidelberg University, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
- Informatics for Life, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Maura M Zylla
- Department of Cardiology, Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Celina Kley
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
- Department of Cardiology, Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Niklas Hartmann
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
- Department of Cardiology, Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Florian Sicklinger
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
- Department of Cardiology, Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | | | - Norbert Frey
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
- Department of Cardiology, Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Heidelberg University, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany.
- Informatics for Life, Heidelberg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany.
| | - Florian Leuschner
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany.
- Department of Cardiology, Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| |
Collapse
|
29
|
Izquierdo MC, Cabodevilla AG, Basu D, Nasias D, Kanter JE, Ho W, Gjini J, Fisher EA, Kim J, Lee W, Bornfeldt KE, Goldberg IJ. Hyperchylomicronemia causes endothelial cell inflammation and increases atherosclerosis. RESEARCH SQUARE 2024:rs.3.rs-5451391. [PMID: 39649171 PMCID: PMC11623764 DOI: 10.21203/rs.3.rs-5451391/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
The effect of increased triglycerides (TGs) as an independent factor in atherosclerosis development has been contentious, in part, because severe hypertriglyceridemia associates with low levels of low-density lipoprotein cholesterol (LDL-C). To test whether hyperchylomicronemia, in the absence of markedly reduced LDL-C levels, contributes to atherosclerosis, we created mice with induced whole-body lipoprotein lipase (LpL) deficiency combined with LDL receptor (LDLR) deficiency. On an atherogenic Western-type diet (WD), male and female mice with induced global LpL deficiency (iLpl -/-) and LDLR knockdown (Ldlr kd ) developed hypertriglyceridemia and elevated cholesterol levels; all the increased cholesterol was in chylomicrons or large VLDL. After 12 weeks on a WD, atherosclerotic lesions both in the brachiocephalic artery and the aortic root were more severe in iLpl -/- /Ldlr kd mice compared to the control Ldlr kd mice. One likely mechanism for this is that exposure of the aorta to hyperchylomicronemia led to endothelial cell inflammation. Thus, our data show that intact chylomicrons contribute to atherosclerosis, explain the association of postprandial lipemia and vascular disease, and prove that hyperchylomicronemia is not benign.
Collapse
Affiliation(s)
- Maria Concepcion Izquierdo
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Ainara G. Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Dimitris Nasias
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Jenny E. Kanter
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Winnie Ho
- Keenan Centre for Biomedical Research, St. Michael’s Hospital and Division of Critical Care, Department of Medicine, University of Toronto, Canada
| | - Jana Gjini
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| | - Edward A. Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Jeffrey Kim
- Comparative Medicine Research Unit, University of Louisville School of Medicine, Louisville, KY
| | - Warren Lee
- Keenan Centre for Biomedical Research, St. Michael’s Hospital and Division of Critical Care, Department of Medicine, University of Toronto, Canada
| | - Karin E. Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
30
|
Maidman SD, Hegele RA, Rosenson RS. The Emerging Potential of Apolipoprotein C-III Inhibition for ASCVD Prevention: A State-of-the-Art Review. Curr Atheroscler Rep 2024; 27:3. [PMID: 39541062 DOI: 10.1007/s11883-024-01258-8] [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] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW Multiple agents are being developed that inhibit apolipoprotein (apo) C-III. This state-of-the-art review examines their potential for atherosclerotic cardiovascular disease (ASCVD) risk reduction. RECENT FINDINGS Apo C-III, an apolipoprotein on the surface of triglyceride-rich lipoproteins (TRLs), impairs clearance of TRLs through both lipoprotein lipase dependent and independent pathways, thereby resulting in increased concentrations of triglycerides. Apo C-III has also been shown to have pro-atherogenic effects when bound to high-density lipoprotein (HDL) particles. Classical and genetic epidemiology studies provide support for the concept that apo C-III is associated with an increased risk of ASCVD events. Drug efficacy of agents that silence APOC3 mRNA has been studied in populations with varying hypertriglyceridemia severity, including those with familial chylomicronemia syndrome, multifactorial chylomicronemia syndrome/severe hypertriglyceridemia, and mixed hyperlipidemia. Randomized controlled trials have reported significant reductions in TG and non-HDL cholesterol levels among these patients treated with APOC3 inhibitors. Upcoming clinical outcomes trials seek to establish a role for APOC3 inhibitors to reduce risk of ASCVD.
Collapse
Affiliation(s)
- Samuel D Maidman
- Metabolism and Lipids Program, Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Robert S Rosenson
- Metabolism and Lipids Program, Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029, USA.
| |
Collapse
|
31
|
DePaolo J, Biagetti G, Judy R, Wang GJ, Kelly JJ, Iyengar A, Goel NJ, Desai ND, Szeto WY, Bavaria JE, Levin MG, Damrauer SM. Polygenic Scoring for Detection of Ascending Thoracic Aortic Dilation. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004512. [PMID: 39324273 PMCID: PMC11540195 DOI: 10.1161/circgen.123.004512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 08/30/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Ascending thoracic aortic dilation is a complex heritable trait that involves modifiable and nonmodifiable risk factors. Polygenic scores (PGS) are increasingly used to assess risk for complex diseases. The degree to which a PGS can improve aortic diameter prediction in diverse populations is unknown. Presently, we tested whether adding a PGS to clinical prediction algorithms improves performance in a diverse biobank. METHODS The analytic cohort comprised 6235 Penn Medicine Biobank participants with available echocardiography and clinical data linked to genome-wide genotype data. Linear regression models were used to integrate PGS weights derived from a genome-wide association study of thoracic aortic diameter performed in the UK Biobank and were compared with the performance of the previously published aorta optimized regression for thoracic aneurysm (AORTA) score. RESULTS Cohort participants had a median age of 61 years (IQR, 53-70) and a mean ascending aortic diameter of 3.36 cm (SD, 0.49). Fifty-five percent were male, and 33% were genetically similar to an African reference population. Compared with the AORTA score, which explained 30.6% (95% CI, 29.9%-31.4%) of the variance in aortic diameter, AORTA score+UK Biobank-derived PGS explained 33.1%, (95% CI, 32.3%-33.8%), the reweighted AORTA score explained 32.5% (95% CI, 31.8%-33.2%), and the reweighted AORTA score+UK Biobank-derived PGS explained 34.9% (95% CI, 34.2%-35.6%). When stratified by population, models including the UK Biobank-derived PGS consistently improved upon the clinical AORTA score among individuals genetically similar to a European reference population but conferred minimal improvement among individuals genetically similar to an African reference population. Comparable performance disparities were observed in models developed to discriminate cases/noncases of thoracic aortic dilation (≥4.0 cm). CONCLUSIONS We demonstrated that inclusion of a UK Biobank-derived PGS to the AORTA score confers a clinically meaningful improvement in model performance only among individuals genetically similar to European reference populations and may exacerbate existing health care disparities.
Collapse
Affiliation(s)
| | - Gina Biagetti
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery (G.B., G.J.W., S.M.D.)
| | | | - Grace J Wang
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery (G.B., G.J.W., S.M.D.)
| | - John J Kelly
- Division of Cardiovascular Surgery, Department of Surgery (J.J.K., A.I., N.J.G., N.D.D., W.Y.S., J.E.B.)
| | - Amit Iyengar
- Division of Cardiovascular Surgery, Department of Surgery (J.J.K., A.I., N.J.G., N.D.D., W.Y.S., J.E.B.)
| | - Nicholas J Goel
- Division of Cardiovascular Surgery, Department of Surgery (J.J.K., A.I., N.J.G., N.D.D., W.Y.S., J.E.B.)
| | - Nimesh D Desai
- Division of Cardiovascular Surgery, Department of Surgery (J.J.K., A.I., N.J.G., N.D.D., W.Y.S., J.E.B.)
| | - Wilson Y Szeto
- Division of Cardiovascular Surgery, Department of Surgery (J.J.K., A.I., N.J.G., N.D.D., W.Y.S., J.E.B.)
| | - Joseph E Bavaria
- Division of Cardiovascular Surgery, Department of Surgery (J.J.K., A.I., N.J.G., N.D.D., W.Y.S., J.E.B.)
| | - Michael G Levin
- Department of Medicine, Division of Cardiology (M.G.L.)
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA (M.G.L., S.M.D.)
| | - Scott M Damrauer
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery (G.B., G.J.W., S.M.D.)
- Department of Genetics (S.M.D.)
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania (S.M.D.)
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA (M.G.L., S.M.D.)
| |
Collapse
|
32
|
Gugliucci A. Angiopoietin-like Proteins and Lipoprotein Lipase: The Waltz Partners That Govern Triglyceride-Rich Lipoprotein Metabolism? Impact on Atherogenesis, Dietary Interventions, and Emerging Therapies. J Clin Med 2024; 13:5229. [PMID: 39274442 PMCID: PMC11396212 DOI: 10.3390/jcm13175229] [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: 08/22/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Over 50% of patients who take statins are still at risk of developing atherosclerotic cardiovascular disease (ASCVD) and do not achieve their goal LDL-C levels. This residual risk is largely dependent on triglyceride-rich lipoproteins (TRL) and their remnants. In essence, remnant cholesterol-rich chylomicron (CM) and very-low-density lipoprotein (VLDL) particles play a role in atherogenesis. These remnants increase when lipoprotein lipase (LPL) activity is inhibited. ApoCIII has been thoroughly studied as a chief inhibitor and therapeutic options to curb its effect are available. On top of apoCIII regulation of LPL activity, there is a more precise control of LPL in various tissues, which makes it easier to physiologically divide the TRL burden according to the body's requirements. In general, oxidative tissues such as skeletal and cardiac muscle preferentially take up lipids during fasting. Conversely, LPL activity in adipocytes increases significantly after feeding, while its activity in oxidative tissues decreases concurrently. This perspective addresses the recent improvements in our understanding of circadian LPL regulations and their therapeutic implications. Three major tissue-specific lipolysis regulators have been identified: ANGPTL3, ANGPTL4, and ANGPTL8. Briefly, during the postprandial phase, liver ANGPTL8 acts on ANGPTL3 (which is released continuously from the liver) to inhibit LPL in the heart and muscle through an endocrine mechanism. On the other hand, when fasting, ANGPTL4, which is released by adipocytes, inhibits lipoprotein lipase in adipose tissue in a paracrine manner. ANGPTL3 inhibitors may play a therapeutic role in the treatment of hypertriglyceridemia. Several approaches are under development. We look forward to future studies to clarify (a) the nature of hormonal and nutritional factors that determine ANGPTL3, 4, and 8 activities, along with what long-term impacts may be expected if their regulation is impaired pharmacologically; (b) the understanding of the quantitative hierarchy and interaction of the regulatory actions of apoCIII, apoAV, and ANGPTL on LPL activity; (c) strategies for the safe and proper treatment of postprandial lipemia; and (d) the effect of fructose restriction on ANGPTL3, ANGPTL4, and ANGPTL8.
Collapse
Affiliation(s)
- Alejandro Gugliucci
- Glycation, Oxidation and Disease Laboratory, Touro University California, Vallejo, CA 94592, USA
| |
Collapse
|
33
|
Romero G, Martin B, Gabris B, Salama G. Relaxin suppresses atrial fibrillation, reverses fibrosis and reduces inflammation in aged hearts. Biochem Pharmacol 2024; 227:116407. [PMID: 38969298 DOI: 10.1016/j.bcp.2024.116407] [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: 03/19/2024] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Healthy aging results in cardiac structural and electrical remodeling that increase susceptibility to cardiovascular diseases. Relaxin has shown broad cardioprotective effects including anti-fibrotic, anti-arrhythmic and anti-inflammatory outcomes in multiple models. This paper focuses on the cardioprotective effects of Relaxin in a rat model of aging. Sustained atrial or ventricular fibrillation are readily induced in the hearts of aged but not young control animals. Treatment with Relaxin suppressed this arrhythmogenic response by increasing conduction velocity, decreasing fibrosis and promoting substantial cardiac remodeling. Relaxin treatment resulted in a significant increase in the levels of: Nav1.5, Cx43, βcatenin and Wnt1 in rat hearts. In isolated cardiomyocytes, Relaxin increased Nav1.5 expression. These effects were mimicked by CHIR 99021, a pharmacological activator of canonical Wnt signaling, but blocked by the canonical Wnt inhibitor Dickkopf1. Relaxin prevented TGF-β-dependent differentiation of cardiac fibroblasts into myofibroblasts while increasing the expression of Wnt1; the effects of Relaxin on cardiac fibroblast differentiation were blocked by Dickkopf1. RNASeq studies demonstrated reduced expression of pro-inflammatory cytokines and an increase in the expression of α- and β-globin in Relaxin-treated aged males. Relaxin reduces arrhythmogenicity in the hearts of aged rats by reduction of fibrosis and increased conduction velocity. These changes are accompanied by substantial remodeling of the cardiac tissue and appear to be mediated by increased canonical Wnt signaling. Relaxin also exerts significant anti-inflammatory and anti-oxidant effects in the hearts of aged rodents. The mechanisms by which Relaxin increases the expression of Wnt ligands, promotes Wnt signaling and reprograms gene expression remain to be determined.
Collapse
Affiliation(s)
- Guillermo Romero
- Departments of Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Brian Martin
- Departments of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Beth Gabris
- Departments of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guy Salama
- Departments of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
34
|
Fan KC, Chen SC, Yen IW, Lin CH, Kuo CH, Lyu YP, Juan HC, Lin MS, Wang SH, Li HY. Plasma angiopoietin-like protein 4 as a novel biomarker predicting 10-year mortality in a community-based population: a longitudinal cohort study. Arch Med Sci 2024; 21:51-59. [PMID: 40190305 PMCID: PMC11969511 DOI: 10.5114/aoms/189504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/30/2024] [Indexed: 04/09/2025] Open
Abstract
Introduction Angiopoietin-like protein 4 (ANGPTL4) is a hepatokine implicated in fat metabolism regulation. Its genetic inactivation has been associated with improved glucose homeostasis, while elevated plasma ANGPTL4 levels are observed in diabetic and obese individuals. However, the potential link between ANGPTL4 and diabetes- or obesity-related complications remains uncertain. This study aimed to explore whether plasma ANGPTL4 level could serve as a predictor of cancer mortality, cardiovascular mortality, and all-cause mortality in a community-based cohort. Material and methods A community-based cohort study was conducted, where fasting plasma ANGPTL4 concentrations were measured at baseline, and vital status was ascertained through linkage with the National Health Insurance Research Database in Taiwan. Results During a 10.46-year follow-up period, 29 (2.49%) of the 1163 participants died. Subjects within the highest tertile of plasma ANGPTL4 levels exhibited the lowest survival rate. In unadjusted models, plasma ANGPTL4 significantly predicted all-cause mortality, cancer mortality, and cardiovascular or cancer-related mortality. Upon adjustment for confounders including age, sex, smoking, body mass index (BMI), hypertension, diabetes mellitus (DM), and renal function, each standard deviation increase in plasma ANGPTL4 was associated with HRs of 1.35 (95% CI: 1.01-1.80, p < 0.05) for all-cause mortality, 1.41 (95% CI: 0.94-2.10, p = 0.094) for cancer mortality, and 1.40 (95% CI: 1.02-1.94, p < 0.05) for cardiovascular or cancer-related mortality. Additionally, plasma ANGPTL4 contributed more significantly to predicting cardiovascular or cancer-related mortality and all-cause mortality compared to other predictors, such as sex, smoking, BMI, history of hypertension, history of diabetes, and eGFR. Conclusions Plasma ANGPTL4 emerges as a promising biomarker capable of predicting 10-year mortality and enhancing risk prediction beyond established risk factors.
Collapse
Affiliation(s)
- Kang-Chih Fan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Szu-Chi Chen
- Department of Internal Medicine, Taipei City Hospital Renai Branch, Taipei, Taiwan
| | - I-Weng Yen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Hung Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Heng Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Ya-Pin Lyu
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsien-Chia Juan
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mao-Shin Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Huei Wang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hung-Yuan Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
35
|
Liu Y, Lin H, Liu M, Lin L, Wen Y. Establishment of a Mitochondrial Metabolism-Related Diagnostic Model in Schizophrenia Based on LASSO Algorithm. Psychiatry Investig 2024; 21:618-628. [PMID: 38960439 PMCID: PMC11222072 DOI: 10.30773/pi.2024.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 07/05/2024] Open
Abstract
OBJECTIVE Schizophrenia is a common mental disorder, and mitochondrial function represents a potential therapeutic target for psychiatric diseases. The role of mitochondrial metabolism-related genes (MRGs) in the diagnosis of schizophrenia remains unknown. This study aimed to identify candidate genes that may influence the diagnosis and treatment of schizophrenia based on MRGs. METHODS Three schizophrenia datasets were obtained from the Gene Expression Omnibus database. MRGs were collected from relevant literature. The differentially expressed genes between normal samples and schizophrenia samples were screened using the limma package. Venn analysis was performed to identify differentially expressed MRGs (DEMRGs) in schizophrenia. Based on the STRING database, hub genes in DEMRGs were identified using the MCODE algorithm in Cytoscape. A diagnostic model containing hub genes was constructed using LASSO regression and logistic regression analysis. The relationship between hub genes and drug sensitivity was explored using the DSigDB database. An interaction network between miRNA-transcription factor (TF)-hub genes was created using the Network-Analyst website. RESULTS A total of 1,234 MRGs, 172 DEMRGs, and 6 hub genes with good diagnostic performance were identified. Ten potential candidate drugs (rifampicin, fulvestrant, pentadecafluorooctanoic acid, etc.) were selected. Thirty-four miRNAs targeting genes in the diagnostic model (ANGPTL4, CPT2, GLUD1, MED1, and MED20), as well as 137 TFs, were identified. CONCLUSION Six potential candidate genes showed promising diagnostic significance. rifampicin, fulvestrant, and pentadecafluorooctanoic acid were potential drugs for future research in the treatment of schizophrenia. These findings provided valuable evidence for the understanding of schizophrenia pathogenesis, diagnosis, and drug treatment.
Collapse
Affiliation(s)
- Yinfang Liu
- Department of Pharmacy, The Third Hospital of Longyan, Longyan, China
| | - Han Lin
- Department of Pharmacy, The Third Hospital of Longyan, Longyan, China
| | - Meicen Liu
- Department of Pharmacy, The First Hospital of Longyan, Longyan, China
| | - Liping Lin
- Department of Pharmacy, The Third Hospital of Longyan, Longyan, China
| | - Yaohui Wen
- Department of Laboratory Medicine, The Third Hospital of Longyan, Longyan, China
| |
Collapse
|
36
|
Le A, Peng H, Golinsky D, Di Scipio M, Lali R, Paré G. What Causes Premature Coronary Artery Disease? Curr Atheroscler Rep 2024; 26:189-203. [PMID: 38573470 DOI: 10.1007/s11883-024-01200-y] [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] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW This review provides an overview of genetic and non-genetic causes of premature coronary artery disease (pCAD). RECENT FINDINGS pCAD refers to coronary artery disease (CAD) occurring before the age of 65 years in women and 55 years in men. Both genetic and non-genetic risk factors may contribute to the onset of pCAD. Recent advances in the genetic epidemiology of pCAD have revealed the importance of both monogenic and polygenic contributions to pCAD. Familial hypercholesterolemia (FH) is the most common monogenic disorder associated with atherosclerotic pCAD. However, clinical overreliance on monogenic genes can result in overlooked genetic causes of pCAD, especially polygenic contributions. Non-genetic factors, notably smoking and drug use, are also important contributors to pCAD. Cigarette smoking has been observed in 25.5% of pCAD patients relative to 12.2% of non-pCAD patients. Finally, myocardial infarction (MI) associated with spontaneous coronary artery dissection (SCAD) may result in similar clinical presentations as atherosclerotic pCAD. Recognizing the genetic and non-genetic causes underlying pCAD is important for appropriate prevention and treatment. Despite recent progress, pCAD remains incompletely understood, highlighting the need for both awareness and research.
Collapse
Affiliation(s)
- Ann Le
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Helen Peng
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Danielle Golinsky
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- School of Nursing, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Matteo Di Scipio
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Ricky Lali
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Medical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, 237 Barton Street East, Hamilton, ON, L8L 2X2, Canada.
- Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main Street West, Hamilton, ON, L8L 4K1, Canada.
| |
Collapse
|
37
|
Gagnon E, Bourgault J, Gobeil É, Thériault S, Arsenault BJ. Impact of loss-of-function in angiopoietin-like 4 on the human phenome. Atherosclerosis 2024; 393:117558. [PMID: 38703417 DOI: 10.1016/j.atherosclerosis.2024.117558] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Carriers of the E40K loss-of-function variant in Angiopoietin-like 4 (ANGPTL4), have lower plasma triglyceride levels as well as lower rates of coronary artery disease (CAD) and type 2 diabetes (T2D). These genetic data suggest ANGPTL4 inhibition as a potential therapeutic target for cardiometabolic diseases. However, it is unknown whether the association between E40K and human diseases is due to linkage disequilibrium confounding. The broader impact of genetic ANGPTL4 inhibition is also unknown, raising uncertainties about the safety and validity of this target. METHODS To assess the impact of ANGPLT4 inhibition, we evaluated whether E40K and other loss-of-function variants in ANGPTL4 influenced a wide range of health markers and diseases using 29 publicly available genome-wide association meta-analyses of cardiometabolic traits and diseases, as well as 1589 diseases assessed in electronic health records within FinnGen (n = 309,154). To determine whether these relationships were likely causal, and not driven by other correlated variants, we used the Bayesian fine mapping algorithm CoPheScan. RESULTS The CoPheScan posterior probability of E40K being the causal variant for triglyceride levels was 99.99 %, validating the E40K to proxy lifelong lower activity of ANGPTL4. The E40K variant was associated with lower risk of CAD (odds ratio [OR] = 0.84, 95 % CI = 0.81 to 0.87, p=3.6e-21) and T2D (OR = 0.91, 95 % CI = 0.87 to 0.95, p=2.8e-05) in GWAS meta-analyses, with results replicated in FinnGen. These significant results were also replicated using other rare loss-of-function variants identified through whole exome sequencing in 488,278 participants of the UK Biobank. Using a Mendelian randomization study design, the E40K variant effect on cardiometabolic diseases was concordant with lipoprotein lipase enhancement (r = 0.82), but not hepatic lipase enhancement (r = -0.10), suggesting that ANGPTL4 effects on cardiometabolic diseases are potentially mainly mediated through lipoprotein lipase. After correction for multiple testing, the E40K variant did not significantly increase the risk of any of the 1589 diseases tested in FinnGen. CONCLUSIONS ANGPTL4 inhibition may represent a potentially safe and effective target for cardiometabolic diseases prevention or treatment.
Collapse
Affiliation(s)
- Eloi Gagnon
- Centre de Recherche de L'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | - Jérome Bourgault
- Centre de Recherche de L'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | - Émilie Gobeil
- Centre de Recherche de L'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
| | - Sébastien Thériault
- Centre de Recherche de L'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada; Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Benoit J Arsenault
- Centre de Recherche de L'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada; Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
38
|
Sharma P, Klarin D, Voight BF, Tsao PS, Levin MG, Damrauer SM. Evaluation of Plasma Biomarkers for Causal Association With Peripheral Artery Disease. Arterioscler Thromb Vasc Biol 2024; 44:1114-1123. [PMID: 38545784 PMCID: PMC11043009 DOI: 10.1161/atvbaha.124.320674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Hundreds of biomarkers for peripheral artery disease (PAD) have been reported in the literature; however, the observational nature of these studies limits causal inference due to the potential of reverse causality and residual confounding. We sought to evaluate the potential causal impact of putative PAD biomarkers identified in human observational studies through genetic causal inference methods. METHODS Putative circulating PAD biomarkers were identified from human observational studies through a comprehensive literature search based on terms related to PAD using PubMed, Cochrane, and Embase. Genetic instruments were generated from publicly available genome-wide association studies of circulating biomarkers. Two-sample Mendelian randomization was used to test the association of genetically determined biomarker levels with PAD using summary statistics from a genome-wide association study of 31 307 individuals with and 211 753 individuals without PAD in the Veterans Affairs Million Veteran Program and replicated in data from FinnGen comprised of 11 924 individuals with and 288 638 individuals without PAD. RESULTS We identified 204 unique circulating biomarkers for PAD from the observational literature, of which 173 were genetically instrumented using genome-wide association study results. After accounting for multiple testing (false discovery rate, <0.05), 10 of 173 (5.8%) biomarkers had significant associations with PAD. These 10 biomarkers represented categories including plasma lipoprotein regulation, lipid homeostasis, and protein-lipid complex remodeling. Observational literature highlighted different pathways including inflammatory response, negative regulation of multicellular organismal processes, and regulation of response to external stimuli. CONCLUSIONS Integrating human observational studies and genetic causal inference highlights several key pathways in PAD pathophysiology. This work demonstrates that a substantial portion of biomarkers identified in observational studies are not well supported by human genetic evidence and emphasizes the importance of triangulating evidence to understand PAD pathophysiology. Although the identified biomarkers offer insights into atherosclerotic development in the lower limb, their specificity to PAD compared with more widespread atherosclerosis requires further study.
Collapse
Affiliation(s)
- Pranav Sharma
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Derek Klarin
- Veterans Affairs Palo Alto Healthcare System, CA
- Division of Vascular Surgery, Stanford University School of Medicine, CA
| | - Benjamin F. Voight
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United State
| | - Philip S. Tsao
- Veterans Affairs Palo Alto Healthcare System, CA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, CA
| | - Michael G. Levin
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Scott M. Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
39
|
Landfors F, Henneman P, Chorell E, Nilsson SK, Kersten S. Drug-target Mendelian randomization analysis supports lowering plasma ANGPTL3, ANGPTL4, and APOC3 levels as strategies for reducing cardiovascular disease risk. EUROPEAN HEART JOURNAL OPEN 2024; 4:oeae035. [PMID: 38895109 PMCID: PMC11182694 DOI: 10.1093/ehjopen/oeae035] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/30/2024] [Accepted: 04/26/2024] [Indexed: 06/21/2024]
Abstract
Aims APOC3, ANGPTL3, and ANGPTL4 are circulating proteins that are actively pursued as pharmacological targets to treat dyslipidaemia and reduce the risk of atherosclerotic cardiovascular disease. Here, we used human genetic data to compare the predicted therapeutic and adverse effects of APOC3, ANGPTL3, and ANGPTL4 inactivation. Methods and results We conducted drug-target Mendelian randomization analyses using variants in proximity to the genes associated with circulating protein levels to compare APOC3, ANGPTL3, and ANGPTL4 as drug targets. We obtained exposure and outcome data from large-scale genome-wide association studies and used generalized least squares to correct for linkage disequilibrium-related correlation. We evaluated five primary cardiometabolic endpoints and screened for potential side effects across 694 disease-related endpoints, 43 clinical laboratory tests, and 11 internal organ MRI measurements. Genetically lowering circulating ANGPTL4 levels reduced the odds of coronary artery disease (CAD) [odds ratio, 0.57 per s.d. protein (95% CI 0.47-0.70)] and Type 2 diabetes (T2D) [odds ratio, 0.73 per s.d. protein (95% CI 0.57-0.94)]. Genetically lowering circulating APOC3 levels also reduced the odds of CAD [odds ratio, 0.90 per s.d. protein (95% CI 0.82-0.99)]. Genetically lowered ANGPTL3 levels via common variants were not associated with CAD. However, meta-analysis of protein-truncating variants revealed that ANGPTL3 inactivation protected against CAD (odds ratio, 0.71 per allele [95%CI, 0.58-0.85]). Analysis of lowered ANGPTL3, ANGPTL4, and APOC3 levels did not identify important safety concerns. Conclusion Human genetic evidence suggests that therapies aimed at reducing circulating levels of ANGPTL3, ANGPTL4, and APOC3 reduce the risk of CAD. ANGPTL4 lowering may also reduce the risk of T2D.
Collapse
Affiliation(s)
- Fredrik Landfors
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, B41, Norrlands universitetssjukhus, S-901 87 Umeå, Sweden
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, S-907 36 Umeå, Sweden
| | - Peter Henneman
- Department of Human Genetics, Amsterdam University Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Section of Medicine, Umeå University, B41, Norrlands universitetssjukhus, S-901 87 Umeå, Sweden
| | - Stefan K Nilsson
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, S-907 36 Umeå, Sweden
- Department of Medical Biosciences, Umeå University, B41, Norrlands universitetssjukhus, S-901 87 Umeå, Sweden
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics group, Division of Human Nutrition and Health, Wageningen University, 6708WE Wageningen, the Netherlands
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
40
|
Sagris M, Apostolos A, Theofilis P, Ktenopoulos N, Katsaros O, Tsalamandris S, Tsioufis K, Toutouzas K, Tousoulis D. Myocardial Ischemia-Reperfusion Injury: Unraveling Pathophysiology, Clinical Manifestations, and Emerging Prevention Strategies. Biomedicines 2024; 12:802. [PMID: 38672157 PMCID: PMC11048318 DOI: 10.3390/biomedicines12040802] [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: 01/30/2024] [Revised: 03/02/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) remains a challenge in the context of reperfusion procedures for myocardial infarction (MI). While early revascularization stands as the gold standard for mitigating myocardial injury, recent insights have illuminated the paradoxical role of reperfusion, giving rise to the phenomenon known as ischemia-reperfusion injury. This comprehensive review delves into the intricate pathophysiological pathways involved in MIRI, placing a particular focus on the pivotal role of endothelium. Beyond elucidating the molecular intricacies, we explore the diverse clinical manifestations associated with MIRI, underscoring its potential to contribute substantially to the final infarct size, up to 50%. We further navigate through current preventive approaches and highlight promising emerging strategies designed to counteract the devastating effects of the phenomenon. By synthesizing current knowledge and offering a perspective on evolving preventive interventions, this review serves as a valuable resource for clinicians and researchers engaged in the dynamic field of MIRI.
Collapse
Affiliation(s)
- Marios Sagris
- Correspondence: ; Tel.: +30-213-2088099; Fax: +30-2132088676
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Aghasizadeh M, Ahmadi Hoseini A, Sahebi R, Kazemi T, Asadiyan‐Sohan P, Esmaily H, Samadi S, Avan A, Ferns GA, Khosravi S, Ghazizadeh H, Miri‐Moghaddam E, Ghayour‐Mobarhan M. Association of a genetic variant in angiopoietin-like 3 with serum HDL-C and risk of cardiovascular disease: A study of the MASHAD cohort over 6 years. Mol Genet Genomic Med 2024; 12:e2418. [PMID: 38634215 PMCID: PMC11024632 DOI: 10.1002/mgg3.2418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 01/17/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Loss-of-function (LOF) variants of the angiopoietin-like 3 (ANGPTL3) gene are reported to be associated with serum triglyceride (TG) and high-density lipoprotein cholesterol (HDL-C) concentrations and thereby affect the risk of cardiovascular disease (CVD). OBJECTIVE In the present study, we examined the association of rs10789117 in the ANGPTL 3 gene locus and the risk of CVD in the group of people who were part of the Mashhad-Stroke and Heart-Atherosclerotic-Disorders (MASHAD) cohort. METHODS One thousand and two healthy individuals enrolled in this study of whom 849 subjects were healthy and 153 subjects developed CVD outcomes after 6 years of follow-up. After a 12-h overnight fasting, 20 mL of blood samples were collected for the measurement of fasting blood glucose and lipid profile. DNA was extracted, and the Tetra-ARMS PCR (amplification refractory mutation system) was used for genotyping of rs10789117 in the ANGPTL3 gene. The genotype frequencies of the variant of rs10789117 in the ANGPTL3 gene were estimated using χ2 tests. Eventually, the statistical analysis was done by SPSS version 20. RESULTS Individuals with AC/CC genotypes (rs10789117) were found to have to greater risk of CVD events compared to AA genotype (OR = 1.43, 95%CI = 1.01-2.02, p = 0.041). There was a 1.3-fold increase in cardiovascular events in individuals carrying the C allele of rs10789117 variant compared to non-carriers (OR = 1.32, 95%CI = 1.06-1.72, p value = 0.038). There were significant differences between different genotypes for serum triglyceride levels within the control group, but this difference was not significant in the group with CVD. Moreover, there was a significant association between CC genotype and CVD risk in the individuals with a normal serum HDL-C. CONCLUSION We have found that a rs10789117 C>A in ANGPTL3 gene polymorphism was associated with incident CVD events, and this may be of value as a risk stratification biomarker in CVD in the Iranian population.
Collapse
Affiliation(s)
- Malihe Aghasizadeh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IranMashhad University of Medical SciencesMashhadIran
| | - Asieh Ahmadi Hoseini
- Department of Nutrition, Ghaem HospitalMashhad University of Medical SciencesMashhadIran
| | - Reza Sahebi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IranMashhad University of Medical SciencesMashhadIran
- Department of Modern Sciences and Technologies, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Tooba Kazemi
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
- Razi Clinical Research Development Unit (RCRDU)Birjand University of Medical SciencesBirjandIran
| | - Parisa Asadiyan‐Sohan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IranMashhad University of Medical SciencesMashhadIran
| | - Habibollah Esmaily
- Social Determinants of Health Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Biostatistics, School of HealthMashhad University of Medical SciencesMashhadIran
| | - Sara Samadi
- Department of Modern Sciences and Technologies, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Amir Avan
- Student Research Committee, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
| | - Gordon A. Ferns
- Division of Medical EducationBrighton & Sussex Medical SchoolBrightonUK
| | - Saeede Khosravi
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
| | - Hamideh Ghazizadeh
- Department of Modern Sciences and Technologies, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Ebrahim Miri‐Moghaddam
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
| | - Majid Ghayour‐Mobarhan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IranMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
42
|
Deng M, Kersten S. Characterization of sexual dimorphism in ANGPTL4 levels and function. J Lipid Res 2024; 65:100526. [PMID: 38431115 PMCID: PMC10973588 DOI: 10.1016/j.jlr.2024.100526] [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: 01/30/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
ANGPTL4 is an attractive pharmacological target for lowering plasma triglycerides and cardiovascular risk. Since most preclinical studies on ANGPTL4 were performed in male mice, little is known about sexual dimorphism in ANGPTL4 regulation and function. Here, we aimed to study potential sexual dimorphism in ANGPTL4 mRNA and protein levels and ANGPTL4 function. Additionally, we performed exploratory studies on the function of ANGPTL4 in the liver during fasting using Angptl4-transgenic and Angptl4-/- mice. Compared to female mice, male mice showed higher hepatic and adipose ANGPTL4 mRNA and protein levels, as well as a more pronounced effect of genetic ANGPTL4 modulation on plasma lipids. By contrast, very limited sexual dimorphism in ANGPTL4 levels was observed in human liver and adipose tissue. In human and mouse adipose tissue, ANGPTL8 mRNA and/or protein levels were significantly higher in females than males. Adipose LPL protein levels were higher in female than male Angptl4-/- mice, which was abolished by ANGPTL4 (over) expression. At the human genetic level, the ANGPTL4 E40K loss-of-function variant was associated with similar plasma triglyceride reductions in women and men. Finally, ANGPTL4 ablation in fasted mice was associated with changes in hepatic gene expression consistent with PPARα activation. In conclusion, the levels of ANGPTL4 and the magnitude of the effect of ANGPTL4 on plasma lipids exhibit sexual dimorphism. Nonetheless, inactivation of ANGPTL4 should confer a similar metabolic benefit in women and men. Expression levels of ANGPTL8 in human and mouse adipose tissue are highly sexually dimorphic, showing higher levels in females than males.
Collapse
Affiliation(s)
- Mingjuan Deng
- Nutrition, Metabolism, and Genomics group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands; Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
43
|
Zhao Y, Zhuang Z, Li Y, Xiao W, Song Z, Huang N, Wang W, Dong X, Jia J, Clarke R, Huang T. Elevated blood remnant cholesterol and triglycerides are causally related to the risks of cardiometabolic multimorbidity. Nat Commun 2024; 15:2451. [PMID: 38503751 PMCID: PMC10951224 DOI: 10.1038/s41467-024-46686-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 02/28/2024] [Indexed: 03/21/2024] Open
Abstract
The connection between triglyceride-rich lipoproteins and cardiometabolic multimorbidity, characterized by the concurrence of at least two of type 2 diabetes, ischemic heart disease, and stroke, has not been definitively established. We aim to examine the prospective associations between serum remnant cholesterol, triglycerides, and the risks of progression from first cardiometabolic disease to multimorbidity via multistate modeling in the UK Biobank. We also evaluate the causality of these associations via Mendelian randomization using 13 biologically relevant SNPs as the genetic instruments. Here we show that elevated remnant cholesterol and triglycerides are significantly associated with gradually higher risks of cardiometabolic multimorbidity, particularly the progression of ischemic heart disease to the multimorbidity of ischemic heart disease and type 2 diabetes. These results advocate for effective management of remnant cholesterol and triglycerides as a potential strategy in mitigating the risks of cardiometabolic multimorbidity.
Collapse
Affiliation(s)
- Yimin Zhao
- Department of Sports Medicine, Peking University Third Hospital, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Zhenhuang Zhuang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Yueying Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Wendi Xiao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Zimin Song
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Ninghao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Wenxiu Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xue Dong
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Robert Clarke
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
- Center for Intelligent Public Health, Academy for Artificial Intelligence, Peking University, Beijing, China.
| |
Collapse
|
44
|
Su J, Yang L, Sun Z, Zhan X. Personalized Drug Therapy: Innovative Concept Guided With Proteoformics. Mol Cell Proteomics 2024; 23:100737. [PMID: 38354979 PMCID: PMC10950891 DOI: 10.1016/j.mcpro.2024.100737] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024] Open
Abstract
Personalized medicine can reduce adverse effects, enhance drug efficacy, and optimize treatment outcomes, which represents the essence of personalized medicine in the pharmacy field. Protein drugs are crucial in the field of personalized drug therapy and are currently the mainstay, which possess higher target specificity and biological activity than small-molecule chemical drugs, making them efficient in regulating disease-related biological processes, and have significant potential in the development of personalized drugs. Currently, protein drugs are designed and developed for specific protein targets based on patient-specific protein data. However, due to the rapid development of two-dimensional gel electrophoresis and mass spectrometry, it is now widely recognized that a canonical protein actually includes multiple proteoforms, and the differences between these proteoforms will result in varying responses to drugs. The variation in the effects of different proteoforms can be significant and the impact can even alter the intended benefit of a drug, potentially making it harmful instead of lifesaving. As a result, we propose that protein drugs should shift from being targeted through the lens of protein (proteomics) to being targeted through the lens of proteoform (proteoformics). This will enable the development of personalized protein drugs that are better equipped to meet patients' specific needs and disease characteristics. With further development in the field of proteoformics, individualized drug therapy, especially personalized protein drugs aimed at proteoforms as a drug target, will improve the understanding of disease mechanisms, discovery of new drug targets and signaling pathways, provide a theoretical basis for the development of new drugs, aid doctors in conducting health risk assessments and making more cost-effective targeted prevention strategies conducted by artificial intelligence/machine learning, promote technological innovation, and provide more convenient treatment tailored to individualized patient profile, which will benefit the affected individuals and society at large.
Collapse
Affiliation(s)
- Junwen Su
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lamei Yang
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziran Sun
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
45
|
Andersson DP, Littmann K, Kindborg G, Eklund D, Sejersen K, Yan J, Eriksson Hogling D, Parini P, Brinck J. Relation among hypertriglyceridaemia, cardiometabolic disease, and hereditary factors-design and rationale of the Stockholm hyperTRIglyceridaemia REGister study. EUROPEAN HEART JOURNAL OPEN 2024; 4:oeae010. [PMID: 38487365 PMCID: PMC10937219 DOI: 10.1093/ehjopen/oeae010] [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: 09/18/2023] [Revised: 01/17/2024] [Accepted: 02/12/2024] [Indexed: 03/17/2024]
Abstract
Aims Hypertriglyceridaemia (hTG) is associated with atherosclerotic cardiovascular disease, pancreatitis, and non-alcoholic fatty liver disease (NAFLD) in large population-based studies. The understanding of the impact of hereditary hTG and cardiometabolic disease status on the development of hTG and its associated cardiometabolic outcomes is more limited. We aimed to establish a multigenerational cohort to enable studies of the relationship between hTG, cardiometabolic disease and hereditary factors. Methods and results The population-based observational Stockholm hyperTRIglyceridaemia REGister (STRIREG) study includes 1 460 184 index individuals who have measured plasma triglycerides in the clinical routine in Region Stockholm, Sweden, between 1 January 2000 and 31 December 2021. The laboratory measurements also included basic haematology, blood lipid panel, liver function tests, and HbA1c. Using the Swedish Multi-Generation register, 2 147 635 parents and siblings to the indexes were identified to form the complete study cohort. Laboratory data from participants were combined with data from several national registers that provided information on the cause of death, medical diagnoses, dispensed medicines, and socioeconomic factors including country of birth, education level, and marital status. Conclusion The multi-generational longitudinal STRIREG cohort provides a unique opportunity to investigate different aspects of hTG as well as heredity for other metabolic diseases. Important outcome measures include mortality, cardiovascular mortality, major cardiovascular events, development of incident diabetes, and NAFLD. The STRIREG study will provide a deeper understanding of the impact of hereditary factors and associated cardiometabolic complications.
Collapse
Affiliation(s)
- Daniel P Andersson
- Department of Medicine Huddinge, Karolinska Institutet, Cardio Metabolic Unit, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
- Medical Unit Endocrinology, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Karin Littmann
- Department of Medicine Huddinge, Karolinska Institutet, Cardio Metabolic Unit, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
- Medical Unit Endocrinology, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Gustav Kindborg
- Department of Medicine Huddinge, Karolinska Institutet, Cardio Metabolic Unit, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
- Medical Unit Endocrinology, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Daniel Eklund
- Medical Unit Clinical Chemistry, C1-62, Karolinska University Laboratory, 141 86 Stockholm, Sweden
| | - Kristina Sejersen
- Department of Medical Sciences, Section of Clinical Chemistry, Uppsala University, Uppsala University Hospital, 751 85 Uppsala, Sweden
- Unilabs AB, Unilabs Laboratory Medicine Stockholm, Section of Clinical Chemistry, 171 54 Solna, Sweden
| | - Jane Yan
- Institute of Environmental Medicine, Unit of Biostatistics, Karolinska Institutet, Nobels väg 13, 17 177 Stockholm, Sweden
| | - Daniel Eriksson Hogling
- Department of Medicine Huddinge, Karolinska Institutet, Cardio Metabolic Unit, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
- Medical Unit Endocrinology, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Paolo Parini
- Department of Medicine Huddinge, Karolinska Institutet, Cardio Metabolic Unit, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
- Medical Unit Endocrinology, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
- Department of Laboratory Medicine, Cardio Metabolic Unit, Karolinska Institutet, Alfred Nobels Allé 8, 141 52 Huddinge, Sweden
| | - Jonas Brinck
- Department of Medicine Huddinge, Karolinska Institutet, Cardio Metabolic Unit, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
- Medical Unit Endocrinology, C2:94, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| |
Collapse
|
46
|
Li W, Wang Y, Huang R, Lian F, Xu G, Wang W, Xue S. Rare and common coding variants in lipid metabolism-related genes and their association with coronary artery disease. BMC Cardiovasc Disord 2024; 24:97. [PMID: 38336686 PMCID: PMC10858582 DOI: 10.1186/s12872-024-03759-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Coronary artery disease (CAD) is a complex disease that is influenced by environmental and genetic factors. In this study, we aimed to investigate the relationship between coding variants in lipid metabolism-related genes and CAD in a Chinese Han population. METHODS A total of 252 individuals were recruited for this study, including 120 CAD patients and 132 healthy control individuals. Rare and common coding variants in 12 lipid metabolism-related genes (ANGPTL3, ANGPTL4, APOA1, APOA5, APOC1, APOC3, CETP, LDLR, LIPC, LPL, PCSK9 and SCARB1) were detected via next-generation sequencing (NGS)-based targeted sequencing. Associations between common variants and CAD were evaluated by Fisher's exact test. A gene-based association test of rare variants was performed by the sequence kernel association test-optimal (SKAT-O test). RESULTS We found 51 rare variants and 17 common variants in this study. One common missense variant, LIPC rs6083, was significantly associated with CAD after Bonferroni correction (OR = 0.47, 95% CI = 0.29-0.76, p = 1.9 × 10- 3). Thirty-three nonsynonymous rare variants were identified, including two novel variants located in the ANGPTL4 (p.Gly47Glu) and SCARB1 (p.Leu233Phe) genes. We did not find a significant association between rare variants and CAD via gene-based analysis via the SKAT-O test. CONCLUSIONS Targeted sequencing is a powerful tool for identifying rare and common variants in CAD. The common missense variant LIPC rs6083 confers protection against CAD. The clinical relevance of rare variants in CAD aetiology needs to be investigated in larger sample sizes in the future.
Collapse
Affiliation(s)
- Wei Li
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Yongyi Wang
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Ritai Huang
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Feng Lian
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Genxing Xu
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Weijun Wang
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
47
|
Yen IW, Lin SY, Lin MW, Lee CN, Kuo CH, Chen SC, Tai YY, Kuo CH, Kuo HC, Lin HH, Juan HC, Lin CH, Fan KC, Wang CY, Li HY. The association between plasma angiopoietin-like protein 4, glucose and lipid metabolism during pregnancy, placental function, and risk of delivering large-for-gestational-age neonates. Clin Chim Acta 2024; 554:117775. [PMID: 38220135 DOI: 10.1016/j.cca.2024.117775] [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: 09/11/2023] [Revised: 12/18/2023] [Accepted: 01/08/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND Large-for-gestational-age (LGA) neonates have increased risk of adverse pregnancy outcomes and adult metabolic diseases. We aimed to investigate the relationship between plasma angiopoietin-like protein 4 (ANGPTL4), a protein involved in lipid and glucose metabolism during pregnancy, placental function, growth factors, and the risk of LGA. METHODS We conducted a prospective cohort study and recruited women with singleton pregnancies at the National Taiwan University Hospital between 2013 and 2018. First trimester maternal plasma ANGPTL4 concentrations were measured. RESULTS Among 353 pregnant women recruited, the LGA group had higher first trimester plasma ANGPTL4 concentrations than the appropriate-for-gestational-age group. Plasma ANGPTL4 was associated with hemoglobin A1c, post-load plasma glucose, plasma triglyceride, plasma free fatty acid concentrations, plasma growth hormone variant (GH-V), and birth weight, but was not associated with cord blood growth factors. After adjusting for age, body mass index, hemoglobin A1c, and plasma triglyceride concentrations, plasma ANGPTL4 concentrations were significantly associated with LGA risk, and its predictive performance, as measured by the area under the receiver operating characteristic curve, outperformed traditional risk factors for LGA. CONCLUSIONS Plasma ANGPTL4 is associated with glucose and lipid metabolism during pregnancy, plasma GH-V, and birth weight, and is an early biomarker for predicting the risk of LGA.
Collapse
Affiliation(s)
- I-Weng Yen
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu County, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shin-Yu Lin
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Wei Lin
- Department of Obstetrics and Gynecology, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu County, Taiwan
| | - Chien-Nan Lee
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Heng Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | | | - Yi-Yun Tai
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan; The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Han-Chun Kuo
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Heng-Huei Lin
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsien-Chia Juan
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Hung Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, Taiwan
| | - Kang-Chih Fan
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu County, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Yuan Wang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, Taiwan
| | - Hung-Yuan Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, Taiwan.
| |
Collapse
|
48
|
Zhao Z, Fu Y, Lian H, Liu Y, Liu J, Sun L, Zhang Y. Correlation between the serum FABP4, ANGPTL3, and ANGPTL4 levels and coronary artery disease. Clin Cardiol 2024; 47:e24246. [PMID: 38425231 PMCID: PMC10905153 DOI: 10.1002/clc.24246] [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: 10/28/2023] [Revised: 02/01/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Lipid metabolism related factors, such as angiopoietin-like protein 3 (ANGPTL3), angiopoietin-like 4 (ANGPTL4), fatty acid-binding protein 4 (FABP4) are newly discovered factors that can affect coronary artery disease (CAD). In this study, we aimed to investigate the relationship between CAD and these lipid metabolism factors. HYPOTHESIS ANGPTL3, ANGPTL4, and FABP4 may provide a new method for the control of CAD risk factors and the prevention and treatment of CAD. METHODS We enrolled 284 consecutive inpatients with suspected CAD and divided them into CAD and non-CAD groups based on the coronary angiography results. Serum ANGPTL3, ANGPTL4, FABP4, and tumor necrosis factor-α (TNF-α) levels were estimated using the enzyme-linked immunosorbent assay. Multivariate logistic regression was used to assess the risk factors for CAD. The receiver operating characteristic curve was used to determine the cutoff and diagnostic values. RESULTS The serum TNF-α, FABP4, ANGPTL3, and ANGPTL4 values showed a significant difference between the CAD and non-CAD groups (p < .05). After adjusting for confounding factors, the FABP4, ANGPTL3, and ANGPTL4 levels were independently associated with CAD (p < .05). The ANGPTL3 expression level was an independent risk factor for CAD in patients with hypertension, but not in those without hypertension. The ANGPTL3 > 67.53 ng/mL, ANGPTL4 > 29.95 ng/mL, and FABP4 > 1421.25 ng/L combination had the highest diagnostic value for CAD. CONCLUSION ANGPTL3, ANGPTL4, and FABP4 were identified as independent risk factors for CAD and have valuable clinical implications for the diagnosis and treatment of CAD.
Collapse
Affiliation(s)
- Zhuoyan Zhao
- Department of CardiologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Ying Fu
- Department of CardiologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Huan Lian
- Department of CardiologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Yixiang Liu
- Department of CardiologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Jingyi Liu
- Department of CardiologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
- Hebei Key Laboratory of Panvascular DiseasesChengdeChina
| | - Lixian Sun
- Department of CardiologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
- Hebei Key Laboratory of Panvascular DiseasesChengdeChina
| | - Ying Zhang
- Department of CardiologyThe Affiliated Hospital of Chengde Medical UniversityChengdeChina
- Hebei Key Laboratory of Panvascular DiseasesChengdeChina
| |
Collapse
|
49
|
Zhang Y, Zhang ZT, Wan SY, Yang J, Wei YJ, Chen HJ, Zhou WZ, Song QY, Niu SX, Zheng L, Huang K. ANGPTL3 negatively regulates IL-1β-induced NF-κB activation by inhibiting the IL1R1-associated signaling complex assembly. J Mol Cell Biol 2024; 15:mjad053. [PMID: 37634084 PMCID: PMC11149415 DOI: 10.1093/jmcb/mjad053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 05/15/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023] Open
Abstract
Interleukin-1β (IL-1β)-induced signaling is one of the most important pathways in regulating inflammation and immunity. The assembly of the receptor complex, consisting of the ligand IL-1β, the IL-1 receptor (IL-1R) type 1 (IL1R1), and the IL-1R accessory protein (IL1RAP), initiates this signaling. However, how the IL1R1-associated complex is regulated remains elusive. Angiopoietin like 3 (ANGPTL3), a key inhibitor of plasma triglyceride clearance, is mainly expressed in the liver and exists in both intracellular and extracellular secreted forms. Currently, ANGPTL3 has emerged as a highly promising drug target for hypertriglyceridemia and associated cardiovascular diseases. However, most studies have focused on the secreted form of ANGPTL3, while its intracellular role is still largely unknown. Here, we report that intracellular ANGPTL3 acts as a negative regulator of IL-1β-triggered signaling. Overexpression of ANGPTL3 inhibited IL-1β-induced NF-κB activation and the transcription of inflammatory genes in HepG2, THP1, and HEK293T cells, while knockdown or knockout of ANGPTL3 resulted in opposite effects. Mechanistically, ANGPTL3 interacted with IL1R1 and IL1RAP through its intracellular C-terminal fibrinogen-like domain and disrupted the assembly of the IL1R1-associated complex. Taken together, our study reveals a novel role for ANGPTL3 in inflammation, whereby it inhibits the physiological interaction between IL1R1 and IL1RAP to maintain immune tolerance and homeostasis in the liver.
Collapse
Affiliation(s)
- Yu Zhang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zi-tong Zhang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shi-yuan Wan
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Yang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu-juan Wei
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hui-jing Chen
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wan-zhu Zhou
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiu-yi Song
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shu-xuan Niu
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
50
|
DePaolo J, Bornstein M, Judy R, Abramowitz S, Verma SS, Levin MG, Arany Z, Damrauer SM. Titin-Truncating variants Predispose to Dilated Cardiomyopathy in Diverse Populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.17.24301405. [PMID: 38293092 PMCID: PMC10827233 DOI: 10.1101/2024.01.17.24301405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Importance The effect of high percentage spliced in (hiPSI) TTN truncating variants (TTNtvs) on risk of dilated cardiomyopathy (DCM) has historically been studied among population subgroups defined by genetic similarity to European reference populations. This has raised questions about the effect of TTNtvs in diverse populations, especially among individuals genetically similar to African reference populations. Objective To determine the effect of TTNtvs on risk of DCM in diverse population as measured by genetic distance (GD) in principal component (PC) space. Design Cohort study. Setting Penn Medicine Biobank (PMBB) is a large, diverse biobank. Participants Participants were recruited from across the Penn Medicine healthcare system and volunteered to have their electronic health records linked to biospecimen data including DNA which has undergone whole exome sequencing. Main Outcomes and Measures Risk of DCM among individuals carrying a hiPSI TTNtv. Results Carrying a hiPSI TTNtv was associated with DCM among PMBB participants across a range of GD deciles from the 1000G European centroid; the effect estimates ranged from odds ratio (OR) = 3.29 (95% confidence interval [CI] 1.26 to 8.56) to OR = 9.39 (95% CI 3.82 to 23.13). When individuals were assigned to population subgroups based on genetic similarity to the 1000G reference populations, hiPSI TTNtvs conferred significant risk of DCM among those genetically similar to the 1000G European reference population (OR = 7.55, 95% CI 4.99 to 11.42, P<0.001) and individuals genetically similar to the 1000G African reference population (OR 3.50, 95% CI 1.48 to 8.24, P=0.004). Conclusions and Relevance TTNtvs are associated with increased risk of DCM among a diverse cohort. There is no significant difference in effect of TTNtvs on DCM risk across deciles of GD from the 1000G European centroid, suggesting genetic background should not be considered when screening individuals for titin-related DCM.
Collapse
Affiliation(s)
- John DePaolo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marc Bornstein
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Renae Judy
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Abramowitz
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Michael G Levin
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Zoltan Arany
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|