1
|
Ferri R, Falasca K, Vecchiet J, Ucciferri C. "Are new lipid lowering agents a good option for achieving lipid goals in people living with HIV? A case report". Int J STD AIDS 2025; 36:591-594. [PMID: 40123063 DOI: 10.1177/09564624251327237] [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] [Indexed: 03/25/2025]
Abstract
The dyslipidemia in people living with HIV differs from the general population because combination antiretroviral therapy may not only induce dyslipidemia but also interact with lipid-lowering agents.Monoclonal antibodies that target proprotein convertase subtilisin/kexin type 9 (PCSK9) have recently been demonstrated to dramatically reduce LDL-C level (>60%) in the majority of cases, and another interesting new option is inclisiran, a first-in-class, cholesterol-lowering small interfering RNA (siRNA) targeting PCSK9 mRNA and conjugated to triantennary N-acetylgalactosamine carbohydrates (GalNAc).We present the clinical case of a 62-year-old man living with HIV and dyslipidemia in whom new hypolipidemic drugs were fundamental in achieving adequate LDL values to prevent cardiovascular events.
Collapse
Affiliation(s)
- Raffaele Ferri
- Clinic of Infectious Diseases, Department of Medicine and Science of Aging, University "G. D'Annunzio", Chieti, Italy
| | - Katia Falasca
- Clinic of Infectious Diseases, Department of Medicine and Science of Aging, University "G. D'Annunzio", Chieti, Italy
| | - Jacopo Vecchiet
- Clinic of Infectious Diseases, Department of Medicine and Science of Aging, University "G. D'Annunzio", Chieti, Italy
| | - Claudio Ucciferri
- Clinic of Infectious Diseases, Department of Medicine and Science of Aging, University "G. D'Annunzio", Chieti, Italy
| |
Collapse
|
2
|
Delmas D, Mialhe A, Cotte AK, Connat JL, Bouyer F, Hermetet F, Aires V. Lipid metabolism in cancer: Exploring phospholipids as potential biomarkers. Biomed Pharmacother 2025; 187:118095. [PMID: 40311223 DOI: 10.1016/j.biopha.2025.118095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/03/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Aberrant lipid metabolism is increasingly recognized as a hallmark of cancer, contributing to tumor growth, metastatic dissemination, and resistance to therapy. Cancer cells reprogram key metabolic pathways-including de novo lipogenesis, lipid uptake, and phospholipid remodeling-to sustain malignant progression and adapt to microenvironmental demands. This review summarizes current insights into the role of lipid metabolic reprogramming in oncogenesis and highlights recent advances in lipidomics that have revealed cancer type- and stage-specific lipid signatures with diagnostic and prognostic relevance. We emphasize the dual potential of lipid metabolic pathways-particularly those involving phospholipids-as sources of clinically relevant biomarkers and therapeutic targets. Enzymes and transporters involved in these pathways have emerged as promising candidates for both diagnostic applications and pharmacological intervention. We also examine persistent challenges hindering the clinical translation of lipid-based approaches, including analytical variability, insufficient biological validation, and the lack of standardized integration into clinical workflows. Furthermore, the review explores strategies to overcome these barriers, highlighting the importance of incorporating lipidomics into multi-omics frameworks, supported by advanced computational tools and AI-driven analytics, to decipher the complexity of tumor-associated metabolic networks. We discuss how such integrative approaches can facilitate the identification of actionable metabolic targets, improve the specificity and robustness of lipid-based biomarkers, and enhance patient stratification in the context of precision oncology.
Collapse
Affiliation(s)
- Dominique Delmas
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France; Centre de Lutte Contre le Cancer Georges François Leclerc Center, Dijon F-21000, France; Inserm UMS58 - Biologie Santé Dijon (BioSanD), Dijon F-21000, France.
| | - Aurélie Mialhe
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Alexia K Cotte
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Jean-Louis Connat
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Florence Bouyer
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - François Hermetet
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Virginie Aires
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| |
Collapse
|
3
|
Supplee JG, Marmorstein R, Wellen KE. Molecular targets of bempedoic acid and related decoy fatty acids. Trends Endocrinol Metab 2025:S1043-2760(25)00077-3. [PMID: 40345862 DOI: 10.1016/j.tem.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 05/11/2025]
Abstract
Disorders of lipid metabolism, including hyperlipidemia, atherosclerosis, and metabolic dysfunction-associated steatotic liver disease, are increasing across the globe. Bempedoic acid (BPA) is a first-in-class drug for the treatment of hypercholesterolemia and cardiac risk reduction, which may particularly benefit those who do not tolerate statins. Inhibition of hepatic ATP-citrate lyase (ACLY) is widely accepted as the main mediator of its observed clinical effects. However, BPA treatment also has ACLY-independent effects on lipid metabolism, as the structural similarity of BPA to endogenous fatty acids allows it to trigger multiple lipid-signaling pathways. Here, we review the molecular targets of BPA and related 'decoy fatty acid' drugs and identify areas where further study is warranted as these molecules are evaluated for clinical indications.
Collapse
Affiliation(s)
- Julianna G Supplee
- Graduate Group in Biochemistry, Biophysics and Chemical Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ronen Marmorstein
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Kathryn E Wellen
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Noble JT, Bimpeh K, Pisciotta MA, Reyes Ballista JM, Hines KM, Brindley MA. Chikungunya Replication and Infection Is Dependent upon and Alters Cellular Hexosylceramide Levels in Vero Cells. Viruses 2025; 17:509. [PMID: 40284952 PMCID: PMC12031450 DOI: 10.3390/v17040509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/15/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Chikungunya virus (CHIKV), a mosquito-borne alphavirus, causes significant global morbidity, including fever, rash, and persistent arthralgia. Utilizing untargeted lipidomics, we investigated how CHIKV infection alters host cell lipid metabolism in Vero cells. CHIKV infection induced marked catabolism of hexosylceramides, reducing their levels while increasing ceramide byproducts. Functional studies revealed a reliance on fatty acid synthesis, β-oxidation, and glycosphingolipid biosynthesis. Notably, inhibition of uridine diphosphate glycosyltransferase 8 (UGT8), essential for galactosylceramide production, significantly impaired CHIKV replication and entry in Vero cells. Sensitivity of CHIKV to UGT8 inhibition was reproduced in a disease-relevant cell line, mouse hepatocytes (Hepa1-6). CHIKV was also sensitive to evacetrapib, a cholesterol ester transfer protein (CETP) inhibitor, though the mechanism of inhibition appeared independent of CETP itself, suggesting an off-target effect. These findings highlight specific lipid pathways, particularly glycosphingolipid metabolism, as critical for CHIKV replication and further refine our understanding of how CHIKV exploits host lipid networks. This study provides new insights into CHIKV biology and suggests that targeted investigation of host lipid pathways may inform future therapeutic strategies.
Collapse
Affiliation(s)
- Joseph Thomas Noble
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.T.N.); (M.A.P.); (J.M.R.B.)
| | - Kingsley Bimpeh
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA; (K.B.); (K.M.H.)
| | - Michael Anthony Pisciotta
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.T.N.); (M.A.P.); (J.M.R.B.)
| | - Judith Mary Reyes Ballista
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.T.N.); (M.A.P.); (J.M.R.B.)
| | - Kelly Marie Hines
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA; (K.B.); (K.M.H.)
| | - Melinda Ann Brindley
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.T.N.); (M.A.P.); (J.M.R.B.)
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
5
|
Argun Baris S, Baris O, Kaya H, Karasal Guliyev E, Olsen CA, Vural C, Boyaci H, Basyigit I. The effect of bempedoic acid on histopathologic changes associated with natural aging in rat lungs. BMC Pulm Med 2025; 25:142. [PMID: 40155848 PMCID: PMC11951572 DOI: 10.1186/s12890-025-03608-x] [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: 11/26/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND The process of aging is characterized by a series of physiological, cellular, and immunological changes in tissues. Bempedoic acid is an antioxidant, anti-inflammatory, and cholesterol-lowering drug that does not belong to the statin class. The objective of this study was to assess the impact of bempedoic acid on age-related histopathological alterations in rat lungs. METHODS A total of 40 Wistar-Albino male rats (275-357 g) were included in the study, with 10 rats in each experimental group [Young control (YC), Young + Bempedoic acid (YD), Elderly control (EC), Elderly + Bempedoic acid (ED)]. Bempedoic acid (30 mg/kg/day) was administered orally for one month. The rats were housed under controlled conditions to minimize external stressors. The Geropathological Grading System (GGP) was used to standardize the evaluation of age-related findings in the lungs. Some lesions were classified as either present or absent, whereas others were evaluated on a scale of 0-4 according to their severity. Composite lesion scores were calculated for each rat. Additionally, the presence and severity of emphysema in the rat lungs were recorded. RESULTS Although the median composite lesion score was higher in the elderly groups, the difference was not statistically significant (p = 0.7). The distribution of cells associated with passive congestion, heart failure, and atelectasis was higher in the elderly drug group (p = 0.024 and p = 0.015, respectively). The prevalence of perivascular inflammation was significantly higher in the elderly control group compared to the other groups. Moreover, no cases of moderate-to-severe perivascular inflammation were observed in the elderly drug group (p = 0.019). The prevalence of severe emphysema was higher in the elderly control group compared to the other groups, whereas no cases of severe emphysema were observed in the elderly drug group (p = 0.044). CONCLUSION It is hypothesized that statins, a class of antihyperlipidemic drugs, exert a protective effect against aging due to their ability to correct oxidative damage. Similarly, bempedoic acid's effect on fat oxidation and cholesterol metabolism may be associated with its protective role in the lungs.
Collapse
Affiliation(s)
- Serap Argun Baris
- Department of Chest Diseases, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| | - Ozgur Baris
- Department of Cardiovascular Surgery, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| | - Huseyin Kaya
- Department of Chest Diseases, Kocaeli City Hospital, Kocaeli, Turkey.
| | | | - Cenk Anil Olsen
- Department of Cardiovascular Surgery, Kosuyolu High Specialization Education and Research Hospital, Istanbul, Turkey
| | - Cigdem Vural
- Department of Pathology, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| | - Hasim Boyaci
- Department of Chest Diseases, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| | - Ilknur Basyigit
- Department of Chest Diseases, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| |
Collapse
|
6
|
Liu Y, Yu C, Zhang Y, Xie Z, Wang Y, Qian H, Liang L, Liu Y, Chen Q, Jia J, Yan S, Lai X, Li W, Li J, Zhang Y, Nan F, Yu C. A Phase I Study to Evaluate the Safety, Tolerability, Pharmacokinetics of BGT-002, a Novel ATP-Citrate Lyase Inhibitor, in Healthy Chinese Subjects. Drug Des Devel Ther 2025; 19:1783-1794. [PMID: 40093645 PMCID: PMC11910062 DOI: 10.2147/dddt.s504814] [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: 11/06/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Objective This Phase I study evaluated the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of BGT-002, a novel ATP-citrate lyase (ACLY) inhibitor, in healthy Chinese adults. Methods This study included three parts: Part I (single-ascending-dose study), Part II (multiple-ascending-dose study), and Part III (food effect study). A total of 104 healthy subjects were enrolled in the study and were given BGT-002 tablet or placebo per protocol requirements. Blood samples were collected for pharmacokinetic and pharmacodynamic analysis. Safety was assessed by clinical examinations and adverse events. Results In Part I, BGT-002 demonstrated rapid absorption with a Tmax of 0.67 to 1.75 hours, and slow elimination with a T1/2 of 24.53 to 72.86 hours, prolonged with increased dosages. Cmax and AUC0-∞ ranged from 1.55 to 48.39 μg/mL, and 31.09 to 2930.69 h·μg/mL, respectively. In Part II, the accumulation index (Rac) of Cmax and AUCtau following 14 days of consecutive administration were 3.53 to 3.62 and 5.29 to 5.59, respectively, with a dose-proportionality PK profile. The levels of total cholesterol (TC), non-high-density lipoprotein cholesterol (non-HDL-C), and low-density lipoprotein cholesterol (LDL-C) were maximally decreased by 15.80%, 18.50%, and 22.37%, respectively. In Part III, the geometric mean ratio (90% CI) of fed to fasting condition in Cmax and AUC0-∞ of BGT-002 were 73.11% and 98.36%, respectively, indicating a minor food effect on the absorption rate. Across the study, two cases of Grade 3 adverse events (elevated blood triglycerides) were reported, both of which were assessed as not related to BGT-002. No serious adverse events were observed. Conclusion BGT-002 demonstrated favorable safety, tolerability, and lipid-lowering effects, supporting its potential for further clinical development. Clinical Trial Registration ChiCTR2200057793(https://www.chictr.org.cn/showproj.html?proj=160210); ChiCTR2300067474(https://www.chictr.org.cn/showproj.html?proj=182183); ChiCTR2300067472(https://www.chictr.org.cn/showproj.html?proj=184079).
Collapse
Affiliation(s)
- Yun Liu
- Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai, People’s Republic of China
- Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai Engineering Research Center, Shanghai, People’s Republic of China
| | - Chengyin Yu
- Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai, People’s Republic of China
- Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai Engineering Research Center, Shanghai, People’s Republic of China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Yifan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Zhifu Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Yating Wang
- Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai, People’s Republic of China
- Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai Engineering Research Center, Shanghai, People’s Republic of China
| | - Hongjie Qian
- Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai, People’s Republic of China
- Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai Engineering Research Center, Shanghai, People’s Republic of China
| | - Liyu Liang
- Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai, People’s Republic of China
- Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai Engineering Research Center, Shanghai, People’s Republic of China
| | - Yanmei Liu
- Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai, People’s Republic of China
- Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai Engineering Research Center, Shanghai, People’s Republic of China
| | - Qian Chen
- Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai, People’s Republic of China
- Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai Engineering Research Center, Shanghai, People’s Republic of China
| | - Jingying Jia
- Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai, People’s Republic of China
- Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai Engineering Research Center, Shanghai, People’s Republic of China
| | - Sai Yan
- Burgeon Therapeutics Co., Ltd., Shanghai, People’s Republic of China
| | - Xiaoyin Lai
- Burgeon Therapeutics Co., Ltd., Shanghai, People’s Republic of China
| | - Wei Li
- Burgeon Therapeutics Co., Ltd., Shanghai, People’s Republic of China
| | - Jingya Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Yangming Zhang
- Burgeon Therapeutics Co., Ltd., Shanghai, People’s Republic of China
| | - Fajun Nan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Chen Yu
- Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai, People’s Republic of China
- Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai Engineering Research Center, Shanghai, People’s Republic of China
| |
Collapse
|
7
|
Li P, Jiang W. A New Insight on Atherosclerosis Mechanism and Lipid-Lowering Drugs. Rev Cardiovasc Med 2025; 26:25321. [PMID: 40160588 PMCID: PMC11951287 DOI: 10.31083/rcm25321] [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: 06/21/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 04/02/2025] Open
Abstract
Atherosclerosis (AS) is a chronic vascular disease primarily affecting large and medium-sized arteries, involving complex pathological mechanisms such as inflammatory responses, lipid metabolism disorders and vascular plaque formation. In recent years, several emerging research hotspots have appeared in the field of atherosclerosis, including gut microbiota, pyroptosis, ferroptosis, autophagy, cuproptosis, exosomes and non-coding RNA. Traditional lipid-lowering drugs play a crucial role in the treatment of AS but are not able to significantly reverse the pathological changes. This article aims to summarize the latest research progress in the pathogenesis of AS and the diagnosis and treatment of the disease by comprehensively analyzing relevant literature mainly from the past five years. Additionally, the mechanisms of action and research advances of statins, cholesterol absorption inhibitors, fibrates and novel lipid-lowering drugs are reviewed to provide new insights into the diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Penghui Li
- Binhai New Area Hospital of TCM, 300000 Tianjin, China
| | - Wei Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300000 Tianjin, China
| |
Collapse
|
8
|
Puertas-Umbert L, Alonso J, Blanco-Casoliva L, Almendra-Pegueros R, Camacho M, Rodríguez-Sinovas A, Galán M, Roglans N, Laguna JC, Martínez-González J, Rodríguez C. Inhibition of ATP-citrate lyase by bempedoic acid protects against abdominal aortic aneurysm formation in mice. Biomed Pharmacother 2025; 184:117876. [PMID: 39889383 DOI: 10.1016/j.biopha.2025.117876] [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: 11/20/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a prevalent degenerative disease characterized by an exacerbated inflammation and destructive vascular remodeling. Unfortunately, effective pharmacological tools for the treatment of this disease remain a challenge. ATP-citrate lyase (ACLY), the primary enzyme responsible for acetyl-CoA biosynthesis, is a key regulator of inflammatory signaling in macrophages and lymphocytes. Here, we found increased levels of the active (phosphorylated) form of ACLY (p-ACLY) in the inflammatory infiltrate of AAA from patients and in aneurysmal lesions from angiotensin II (Ang II)-infused apolipoprotein E-deficient mice (ApoE-/-). Furthermore, plasma ACLY levels positively correlates with IL6 and IFNγ levels in patients with AAA, while inflammatory stimuli strongly upregulated ACLY expression in macrophages and Jurkat cells. The administration of the ACLY inhibitor bempedoic acid (BemA) protected against Ang II-induced AAA formation in ApoE-/- mice, limiting the progression of aortic dilatation and reducing mortality due to aortic rupture. BMS-303141, another ACLY inhibitor, also ameliorated AAA formation, although to a lesser extent. BemA attenuated vascular remodeling and the disorganization and rupture of elastic fibers induced by Ang II, as well as vascular inflammation, decreasing the recruitment of macrophages (CD68 +) and neutrophils (Ly-6G+) into the aortic wall. Moreover, BemA shifted splenic monocytes toward a functionally anti-inflammatory phenotype, and increased the percentage of CD4+CD69+ cells. Taken together, these results support the contribution of ACLY to AAA and point to BemA as a promising tool to be considered for future clinical trials addressing the management of this disease which is quite often associated with disorders of lipoprotein metabolism.
Collapse
Affiliation(s)
- Lídia Puertas-Umbert
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Laia Blanco-Casoliva
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain
| | | | - Mercedes Camacho
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Antonio Rodríguez-Sinovas
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain; Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Barcelona 08035, Spain
| | - María Galán
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain; Facultad de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Madrid 28922, Spain
| | - Nuria Roglans
- Dept. Farmacologia, Toxicologia i Química Terapèutica. Facultat de Farmàcia i Ciències de l'Alimentació, Institut de Biomedicina, Universitat de Barcelona, Barcelona 08028, Spain; CIBER de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Juan Carlos Laguna
- Dept. Farmacologia, Toxicologia i Química Terapèutica. Facultat de Farmàcia i Ciències de l'Alimentació, Institut de Biomedicina, Universitat de Barcelona, Barcelona 08028, Spain; CIBER de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Cristina Rodríguez
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain.
| |
Collapse
|
9
|
Nicholls SJ, Nelson AJ. Achieving More Optimal Lipid Control with Non-Statin Lipid Lowering Therapy. Curr Atheroscler Rep 2025; 27:32. [PMID: 39954169 PMCID: PMC11829850 DOI: 10.1007/s11883-025-01280-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE OF REVIEW The use of statins has transformed approaches to the prevention of cardiovascular disease. However, many patients remain at a major risk of experiencing cardiovascular events, due to a range of factors including suboptimal control of low-density lipoprotein cholesterol (LDL-C). Accordingly, there is an ongoing need to develop additional strategies, beyond the use of statins, to achieve more effective reductions in cardiovascular risk. RECENT FINDINGS Genomic studies have implicated the causal role of LDL in atherosclerosis and identified that polymorphisms influencing factors involved in lipid metabolism influence both the level of LDL-C and cardiovascular risk. These findings have highlighted the potential for cardiovascular benefit from development of therapies targeting these factors and incremental benefit when used in combination with statins. Clinical trials have demonstrated that these new agents have favourable effects on both atherosclerotic plaque and cardiovascular events. Additional work has sought to improve intensification of statin therapy and adherence with lipid lowering therapy, to achieve more effective cardiovascular prevention via lipid lowering. Emerging therapies, beyond statins, have the potential to optimise lipid levels and play an effective role in the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Stephen J Nicholls
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne, Australia.
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, Melbourne, Australia
| |
Collapse
|
10
|
Ewida H, Benson H, Tareq S, Ahmed MS. Molecular Targets and Small Molecules Modulating Acetyl Coenzyme A in Physiology and Diseases. ACS Pharmacol Transl Sci 2025; 8:36-46. [PMID: 39816789 PMCID: PMC11729435 DOI: 10.1021/acsptsci.4c00476] [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: 08/07/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
Acetyl coenzyme A (acetyl-CoA), a pivotal regulatory metabolite, is a product of numerous catabolic reactions and a substrate for various anabolic responses. Its role extends to crucial physiological processes, such as glucose homeostasis and free fatty acid utilization. Moreover, acetyl-CoA plays a significant part in reshaping the metabolic microenvironment and influencing the progression of several diseases and conditions, including cancer, insulin resistance, diabetes, heart failure, fear, and neuropathic pain. This Review delves into the role of acetyl-CoA in both physiological and pathological conditions, shedding light on the key players in its formation within the cytosol. We specifically focus on the physiological impact of malonyl-CoA decarboxylase (MCD), acetyl-CoA synthetase2 (ACSS2), and ATP-citrate lyase (ACLY) on metabolism, glucose homeostasis, free fatty acid utilization, and post-translational modification cellular processes. Additionally, we present the pathological implications of MCD, ACSS2, and ACLY in various clinical manifestations. This Review also explores the potential and limitations of targeting MCD, ACSS2, and ACLY using small molecules in different clinical settings.
Collapse
Affiliation(s)
- Heba Ewida
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, Texas 79106, United States
- Department
of Biochemistry, Faculty of Pharmacy, Future
University in Egypt, Cairo 11835, Egypt
| | - Harrison Benson
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, Texas 79106, United States
| | - Syed Tareq
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, Texas 79106, United States
| | - Mahmoud Salama Ahmed
- Department
of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, Texas 79106, United States
| |
Collapse
|
11
|
Liu JY, Kuna RS, Pinheiro LV, Nguyen PTT, Welles JE, Drummond JM, Murali N, Sharma PV, Supplee JG, Shiue M, Zhao S, Farria AT, Kumar A, Ruchhoeft ML, Demetriadou C, Kantner DS, Chatoff A, Megill E, Titchenell PM, Snyder NW, Metallo CM, Wellen KE. Bempedoic acid suppresses diet-induced hepatic steatosis independently of ATP-citrate lyase. Cell Metab 2025; 37:239-254.e7. [PMID: 39471816 PMCID: PMC11711013 DOI: 10.1016/j.cmet.2024.10.014] [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: 11/03/2023] [Revised: 08/07/2024] [Accepted: 10/18/2024] [Indexed: 11/01/2024]
Abstract
ATP citrate lyase (ACLY) synthesizes acetyl-CoA for de novo lipogenesis (DNL), which is elevated in metabolic dysfunction-associated steatotic liver disease. Hepatic ACLY is inhibited by the LDL-cholesterol-lowering drug bempedoic acid (BPA), which also improves steatosis in mice. While BPA potently suppresses hepatic DNL and increases fat catabolism, it is unclear if ACLY is its primary molecular target in reducing liver triglyceride. We show that on a Western diet, loss of hepatic ACLY alone or together with the acetyl-CoA synthetase ACSS2 unexpectedly exacerbates steatosis, linked to reduced PPARα target gene expression and fatty acid oxidation. Importantly, BPA treatment ameliorates Western diet-mediated triacylglyceride accumulation in both WT and liver ACLY knockout mice, indicating that its primary effects on hepatic steatosis are ACLY independent. Together, these data indicate that hepatic ACLY plays an unexpected role in restraining diet-dependent lipid accumulation and that BPA exerts substantial effects on hepatic lipid metabolism independently of ACLY.
Collapse
Affiliation(s)
- Joyce Y Liu
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ramya S Kuna
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Laura V Pinheiro
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phuong T T Nguyen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jaclyn E Welles
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jack M Drummond
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nivitha Murali
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Prateek V Sharma
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julianna G Supplee
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mia Shiue
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven Zhao
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aimee T Farria
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Avi Kumar
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mauren L Ruchhoeft
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina Demetriadou
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Daniel S Kantner
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Adam Chatoff
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Emily Megill
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Paul M Titchenell
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel W Snyder
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Christian M Metallo
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
McGuigan A, Blair HA. Bempedoic Acid: A Review in Cardiovascular Risk Reduction in Statin-Intolerant Patients. Am J Cardiovasc Drugs 2025; 25:7-16. [PMID: 39847220 DOI: 10.1007/s40256-024-00714-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 01/24/2025]
Abstract
Oral bempedoic acid (NEXLETOL® in the USA; Nilemdo® in the EU) and the fixed dose combination (FDC) of bempedoic acid/ezetimibe (NEXLIZET® in the USA; Nustendi® in the EU) are approved to reduce cardiovascular (CV) risk in statin-intolerant patients who are at high risk for, or have, CV disease. A first-in-class therapy, bempedoic acid inhibits the adenosine triphosphate-citrate lyase enzyme in the cholesterol biosynthesis pathway. In the multinational phase III CLEAR Outcomes trial in statin-intolerant patients, once-daily bempedoic acid 180 mg significantly reduced the risk of the primary endpoint (a four-component major adverse CV event composite of CV death, nonfatal myocardial infarction, nonfatal stroke, or coronary revascularization) compared with placebo. Bempedoic acid was generally well tolerated and, unlike statins, was associated with a low incidence of musculoskeletal adverse events (AEs). In conclusion, bempedoic acid as a monotherapy or adjunctive to other lipid-lowering therapies expands the treatment options available for the pharmacological reduction of CV risk in statin-intolerant patients, supporting achievement of low-density lipoprotein cholesterol (LDL-C) targets required for CV risk reduction.
Collapse
Affiliation(s)
- Aisling McGuigan
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| | - Hannah A Blair
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand
| |
Collapse
|
13
|
Jiang Y, Liu P, Qiu Z, Zhou M, Cheng M, Yang T. The U.S. FDA approved cardiovascular drugs from 2011 to 2023: A medicinal chemistry perspective. Eur J Med Chem 2024; 275:116593. [PMID: 38889609 DOI: 10.1016/j.ejmech.2024.116593] [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: 04/21/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024]
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality worldwide. A total of 28 new molecular entities (NMEs) were approved by the U.S. Food and Drug Administration (FDA) for the treatment of cardiovascular diseases from 2011 to 2023. Approximately 25 % of the medications were sanctioned for the management of diverse vascular disorders. The other major therapeutic areas of focus included antilipemic agents (15 %), blood pressure disease (11 %), heart failure, hyperkalemia, and cardiomyopathy (7-8% each). Among all the approved drugs, there are a total of 22 new chemical entities (NCEs), including inhibitors, agonists, polymers, and inorganic compounds. In addition to NCEs, 6 biological agents (BLAs), including monoclonal antibodies, small interfering RNAs (siRNAs), and antisense oligonucleotides, have also obtained approval for the treatment of cardiovascular diseases. From this perspective, approved NCEs are itemized and discussed based on their disease, targets, chemical classes, major drug metabolites, and biochemical and pharmacological properties. Systematic analysis has been conducted to examine the binding modes of these approved drugs with their targets using cocrystal structure information or docking studies to provide valuable insights for designing next-generation agents. Furthermore, the synthetic approaches employed in the creation of these drug molecules have been emphasized, aiming to inspire the development of novel, efficient, and applicable synthetic methodologies. Generally, the primary objective of this review is to provide a comprehensive examination of the clinical applications, pharmacology, binding modes, and synthetic methodologies employed in small-molecule drugs approved for treating CVD. This will facilitate the development of more potent and innovative therapeutics for effectively managing cardiovascular diseases.
Collapse
Affiliation(s)
- Yunhan Jiang
- Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China; Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pingxian Liu
- Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiqiang Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Zhou
- Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mengdi Cheng
- Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yang
- Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Espadas I, Cáliz‐Molina MÁ, López‐Fernández‐Sobrino R, Panadero‐Morón C, Sola‐García A, Soriano‐Navarro M, Martínez‐Force E, Venegas‐Calerón M, Salas JJ, Martín F, Gauthier BR, Alfaro‐Cervelló C, Martí‐Aguado D, Capilla‐González V, Martín‐Montalvo A. Hydroxycitrate delays early mortality in mice and promotes muscle regeneration while inducing a rich hepatic energetic status. Aging Cell 2024; 23:e14205. [PMID: 38760909 PMCID: PMC11488303 DOI: 10.1111/acel.14205] [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: 01/31/2024] [Revised: 04/09/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024] Open
Abstract
ATP citrate lyase (ACLY) inhibitors have the potential of modulating central processes in protein, carbohydrate, and lipid metabolism, which can have relevant physiological consequences in aging and age-related diseases. Here, we show that hepatic phospho-active ACLY correlates with overweight and Model for End-stage Liver Disease score in humans. Wild-type mice treated chronically with the ACLY inhibitor potassium hydroxycitrate exhibited delayed early mortality. In AML12 hepatocyte cultures, the ACLY inhibitors potassium hydroxycitrate, SB-204990, and bempedoic acid fostered lipid accumulation, which was also observed in the liver of healthy-fed mice treated with potassium hydroxycitrate. Analysis of soleus tissue indicated that potassium hydroxycitrate produced the modulation of wound healing processes. In vivo, potassium hydroxycitrate modulated locomotor function toward increased wire hang performance and reduced rotarod performance in healthy-fed mice, and improved locomotion in mice exposed to cardiotoxin-induced muscle atrophy. Our findings implicate ACLY and ACLY inhibitors in different aspects of aging and muscle regeneration.
Collapse
Affiliation(s)
- Isabel Espadas
- Andalusian Molecular Biology and Regenerative Medicine Centre‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevilleSpain
| | - María Ángeles Cáliz‐Molina
- Andalusian Molecular Biology and Regenerative Medicine Centre‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevilleSpain
| | - Raúl López‐Fernández‐Sobrino
- Andalusian Molecular Biology and Regenerative Medicine Centre‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevilleSpain
| | - Concepción Panadero‐Morón
- Andalusian Molecular Biology and Regenerative Medicine Centre‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevilleSpain
| | - Alejandro Sola‐García
- Andalusian Molecular Biology and Regenerative Medicine Centre‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevilleSpain
| | - Mario Soriano‐Navarro
- Electron Microscopy Core Facility, Centro de Investigación Príncipe Felipe (CIPF)ValenciaSpain
| | | | | | - Joaquin J. Salas
- Instituto de la Grasa (CSIC)Universidad Pablo de OlavideSevillaSpain
| | - Franz Martín
- Andalusian Molecular Biology and Regenerative Medicine Centre‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevilleSpain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases‐CIBERDEMInstituto de Salud Carlos IIIMadridSpain
| | - Benoit R. Gauthier
- Andalusian Molecular Biology and Regenerative Medicine Centre‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevilleSpain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases‐CIBERDEMInstituto de Salud Carlos IIIMadridSpain
| | - Clara Alfaro‐Cervelló
- Pathology Department, INCLIVA Health Research Institute, Clinic University HospitalUniversity of ValenciaValenciaSpain
| | - David Martí‐Aguado
- Digestive Disease Department, Clinic University HospitalINCLIVA Health Research InstituteValenciaSpain
- Division of Gastroenterology, Hepatology and NutritionCenter for Liver Diseases
| | - Vivian Capilla‐González
- Andalusian Molecular Biology and Regenerative Medicine Centre‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevilleSpain
| | - Alejandro Martín‐Montalvo
- Andalusian Molecular Biology and Regenerative Medicine Centre‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevilleSpain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases‐CIBERDEMInstituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
15
|
Gill D, Dib MJ, Gill R, Bornstein SR, Burgess S, Birkenfeld AL. Effects of ACLY Inhibition on Body Weight Distribution: A Drug Target Mendelian Randomization Study. Genes (Basel) 2024; 15:1059. [PMID: 39202419 PMCID: PMC11353272 DOI: 10.3390/genes15081059] [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: 07/11/2024] [Revised: 08/04/2024] [Accepted: 08/10/2024] [Indexed: 09/03/2024] Open
Abstract
Background: Adenosine triphosphate-citrate lyase (ACLY) inhibition has proven clinically efficacious for low-density lipoprotein cholesterol (LDL-c) lowering and cardiovascular disease (CVD) risk reduction. Clinical and genetic evidence suggests that some LDL-c lowering strategies, such as 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) inhibition with statin therapy increase body weight and the risk of developing type 2 diabetes mellitus (T2DM). However, whether ACLY inhibition affects metabolic risk factors is currently unknown. We aimed to investigate the effects of ACLY inhibition on glycaemic and anthropometric traits using Mendelian randomization (MR). Methods: As genetic instruments for ACLY inhibition, we selected weakly correlated single-nucleotide polymorphisms at the ACLY gene associated with lower ACLY gene expression in the eQTLGen study (N = 31,684) and lower LDL-c levels in the Global Lipid Genetic Consortium study (N = 1.65 million). Two-sample Mendelian randomization was employed to investigate the effects of ACLY inhibition on T2DM risk, and glycaemic and anthropometric traits using summary data from large consortia, with sample sizes ranging from 151,013 to 806,834 individuals. Findings for genetically predicted ACLY inhibition were compared to those obtained for genetically predicted HMGCR inhibition using the same instrument selection strategy and outcome data. Results: Primary MR analyses showed that genetically predicted ACLY inhibition was associated with lower waist-to-hip ratio (β per 1 standard deviation lower LDL-c: -1.17; 95% confidence interval (CI): -1.61 to -0.73; p < 0.001) but not with risk of T2DM (odds ratio (OR) per standard deviation lower LDL-c: 0.74, 95% CI = 0.25 to 2.19, p = 0.59). In contrast, genetically predicted HMGCR inhibition was associated with higher waist-to-hip ratio (β = 0.15; 95%CI = 0.04 to 0.26; p = 0.008) and T2DM risk (OR = 1.73, 95% CI = 1.27 to 2.36, p < 0.001). The MR analyses considering secondary outcomes showed that genetically predicted ACLY inhibition was associated with a lower waist-to-hip ratio adjusted for body mass index (BMI) (β = -1.41; 95%CI = -1.81 to -1.02; p < 0.001). In contrast, genetically predicted HMGCR inhibition was associated with higher HbA1c (β = 0.19; 95%CI = 0.23 to 0.49; p < 0.001) and BMI (β = 0.36; 95%CI = 0.23 to 0.49; p < 0.001). Conclusions: Human genetic evidence supports the metabolically favourable effects of ACLY inhibition on body weight distribution, in contrast to HMGCR inhibition. These findings should be used to guide and prioritize ongoing clinical development efforts.
Collapse
Affiliation(s)
- Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
- Primula Group Ltd., London N8 0RL, UK;
| | - Marie-Joe Dib
- Division of Cardiovascular Medicine, Perelman School of Advanced Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | | | - Stefan R. Bornstein
- Department of Internal Medicine III, University Clinic, Technical University Dresden, D-01062 Dresden, Germany;
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany;
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London WC2R 2LS, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK;
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0SR, UK
| | - Andreas L. Birkenfeld
- German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany;
- Department of Internal Medicine IV, Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, D-72074 Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard Karls University Tübingen, D-72074Tübingen, Germany
| |
Collapse
|
16
|
Kounatidis D, Tentolouris N, Vallianou NG, Mourouzis I, Karampela I, Stratigou T, Rebelos E, Kouveletsou M, Stamatopoulos V, Tsaroucha E, Dalamaga M. The Pleiotropic Effects of Lipid-Modifying Interventions: Exploring Traditional and Emerging Hypolipidemic Therapies. Metabolites 2024; 14:388. [PMID: 39057711 PMCID: PMC11278853 DOI: 10.3390/metabo14070388] [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: 06/18/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerotic cardiovascular disease poses a significant global health issue, with dyslipidemia standing out as a major risk factor. In recent decades, lipid-lowering therapies have evolved significantly, with statins emerging as the cornerstone treatment. These interventions play a crucial role in both primary and secondary prevention by effectively reducing cardiovascular risk through lipid profile enhancements. Beyond their primary lipid-lowering effects, extensive research indicates that these therapies exhibit pleiotropic actions, offering additional health benefits. These include anti-inflammatory properties, improvements in vascular health and glucose metabolism, and potential implications in cancer management. While statins and ezetimibe have been extensively studied, newer lipid-lowering agents also demonstrate similar pleiotropic effects, even in the absence of direct cardiovascular benefits. This narrative review explores the diverse pleiotropic properties of lipid-modifying therapies, emphasizing their non-lipid effects that contribute to reducing cardiovascular burden and exploring emerging benefits for non-cardiovascular conditions. Mechanistic insights into these actions are discussed alongside their potential therapeutic implications.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Eleni Rebelos
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Marina Kouveletsou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | | | - Eleni Tsaroucha
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
17
|
Hu R, Long S, Luo M, Tang B, Tan T, Dong W, Wang Q, Zhang J. Hyperglycemia Inhibits Hepatic SHBG Synthesis Through the NGBR-AMPK-HNF4 Pathway in Rats with Polycystic Ovary Syndrome Induced by Letrozole in Combination with a High-Fat Diet. Mol Nutr Food Res 2024; 68:e2300915. [PMID: 38862276 DOI: 10.1002/mnfr.202300915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/25/2024] [Indexed: 06/13/2024]
Abstract
SCOPE Polycystic ovary syndrome (PCOS) is closely related to non-alcoholic fatty liver disease (NAFLD), and sex hormone-binding globulin (SHBG) is a glycoprotein produced by the liver. Hepatic lipogenesis inhibits hepatic SHBG synthesis, which leads to hyperandrogenemia and ovarian dysfunction in PCOS. Therefore, this study aims to characterize the mechanism whereby liver lipogenesis inhibits SHBG synthesis. METHODS AND RESULTS This study establishes a rat model of PCOS complicated by NAFLD using a high-fat diet in combination with letrozole and performs transcriptomic analysis of the liver. Transcriptomic analysis of the liver shows that the expression of neurite growth inhibitor-B receptor (NgBR), hepatocyte nuclear factor 4α (HNF4α), and SHBG is low. Meantime, HepG2 cells are treated with palmitic acid (PA) to model NAFLD in vitro, which causes decreases in the expression of NgBR, HNF4α, and SHBG. However, the expression of HNF4α and SHBG is restored by treatment with the AMP-activated protein kinase (AMPK) agonist AICAR. CONCLUSIONS NgBR regulates the expression of HNF4α by activating the AMPK signaling pathway, thereby affecting the synthesis of SHBG in the liver. Further mechanistic studies regarding the effect of liver fat on NGBR expression are warranted.
Collapse
Affiliation(s)
- Rao Hu
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Shuanglian Long
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Min Luo
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Bowen Tang
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Tao Tan
- Institute of Applied Anatomy and Reproductive Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Weilei Dong
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qian Wang
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jiaming Zhang
- The First Affiliated Hospital, Gynecology &Obstetrics and Reproductive Medical Center, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
18
|
Chait A, Eckel RH, Vrablik M, Zambon A. Lipid-lowering in diabetes: An update. Atherosclerosis 2024; 394:117313. [PMID: 37945448 DOI: 10.1016/j.atherosclerosis.2023.117313] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/11/2023] [Accepted: 09/22/2023] [Indexed: 11/12/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is accelerated in people with diabetes. Dyslipidemia, hyperglycemia, oxidative stress, and inflammation play a role via a variety of mechanisms operative in the artery wall. In addition, some unique features predispose people with type 1 diabetes to accelerated atherosclerosis. Various organizations have created guidelines that provide advice regarding screening, risk assessment, and roadmaps for treatment to prevent ASCVD in diabetes. Management of dyslipidemia, especially with statins, has proven to be of immense benefit in the prevention of clinical CVD. However, since many patients fail to attain the low levels of low-density lipoproteins (LDL) recommended in these guidelines, supplemental therapy, such as the addition of ezetimibe, bempedoic acid or PCSK9 inhibitors, is often required to reach LDL goals. As a result, the upfront use of combination therapies, particularly a statin plus ezetimibe, is a rational initial approach. The addition to statins of drugs that specifically lower triglyceride levels has not proven beneficial, although the addition of icosapent-ethyl has been shown to be of value, likely by mechanisms independent of triglyceride lowering. Newer treatments in development, including apoC-III and ANGPTL3 inhibitors, seem promising in further reducing apoB-containing lipoproteins.
Collapse
Affiliation(s)
- Alan Chait
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, WA, USA
| | - Robert H Eckel
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Michal Vrablik
- 3rd Department of Internal Medicine, General University Hospital and 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alberto Zambon
- Department of Medicine - DIMED, University of Padova, and IRCCS Multimedica Milan, Italy.
| |
Collapse
|
19
|
Butera E, Termite F, Esposto G, Galasso L, Mignini I, Borriello R, Ainora ME, Miele L, Gasbarrini A, Zocco MA. Exploring the Role of Bempedoic Acid in Metabolic Dysfunction Associated Steatotic Liver Disease: Actual Evidence and Future Perspectives. Int J Mol Sci 2024; 25:6938. [PMID: 39000046 PMCID: PMC11241610 DOI: 10.3390/ijms25136938] [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: 05/25/2024] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/14/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) involves excessive lipid accumulation in hepatocytes, impacting global healthcare due to its high prevalence and risk of progression to severe liver conditions. Its pathogenesis involves genetic, metabolic, and inflammatory factors, with cardiovascular events as the leading cause of mortality. This review examines the role of lipid-lowering therapies in MASLD, with a particular focus on bempedoic acid, a recently approved cholesterol-lowering agent for hypercholesterolemia and high cardiovascular-risk patients. It explores its potential in liver disease by modulating lipid metabolism and inflammatory pathways based on the most recent studies available. Bempedoic acid inhibits ATP-citrate lyase, reducing cholesterol and fatty acid synthesis while activating AMP-activated protein kinase to suppress gluconeogenesis and lipogenesis. Animal studies indicate its efficacy in reducing hepatic steatosis, inflammation, and fibrosis. Bempedoic acid holds promise as a therapeutic for MASLD, offering dual benefits in lipid metabolism and inflammation. Further clinical trials are required to confirm its efficacy and safety in MASLD patients, potentially addressing the multifaceted nature of this disease.
Collapse
Affiliation(s)
- Elena Butera
- Internal Medicine, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy;
| | - Fabrizio Termite
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy; (F.T.); (G.E.); (L.G.); (I.M.); (R.B.); (M.E.A.); (L.M.); (A.G.)
| | - Giorgio Esposto
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy; (F.T.); (G.E.); (L.G.); (I.M.); (R.B.); (M.E.A.); (L.M.); (A.G.)
| | - Linda Galasso
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy; (F.T.); (G.E.); (L.G.); (I.M.); (R.B.); (M.E.A.); (L.M.); (A.G.)
| | - Irene Mignini
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy; (F.T.); (G.E.); (L.G.); (I.M.); (R.B.); (M.E.A.); (L.M.); (A.G.)
| | - Raffaele Borriello
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy; (F.T.); (G.E.); (L.G.); (I.M.); (R.B.); (M.E.A.); (L.M.); (A.G.)
| | - Maria Elena Ainora
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy; (F.T.); (G.E.); (L.G.); (I.M.); (R.B.); (M.E.A.); (L.M.); (A.G.)
| | - Luca Miele
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy; (F.T.); (G.E.); (L.G.); (I.M.); (R.B.); (M.E.A.); (L.M.); (A.G.)
| | - Antonio Gasbarrini
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy; (F.T.); (G.E.); (L.G.); (I.M.); (R.B.); (M.E.A.); (L.M.); (A.G.)
| | - Maria Assunta Zocco
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A.Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy; (F.T.); (G.E.); (L.G.); (I.M.); (R.B.); (M.E.A.); (L.M.); (A.G.)
| |
Collapse
|
20
|
Nicholls SJ, Nelson AJ. New targets and mechanisms of action for lipid-lowering and anti-inflammatory therapies in atherosclerosis: where does the field stand? Expert Opin Ther Targets 2024; 28:375-384. [PMID: 38815057 DOI: 10.1080/14728222.2024.2362644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/29/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION Atherosclerotic cardiovascular disease remains a leading cause of morbidity and mortality worldwide, despite widespread use of statins. There is a need to develop additional therapeutic strategies that will complement statins to achieve more effective reductions in cardiovascular risk. AREAS COVERED This review provides a comprehensive summary of current areas of therapeutic development targeting both lipid and inflammatory factors implicated in the pathogenesis of atherosclerosis. In addition to develop of novel approaches that will produce more effective lowering of low-density lipoprotein cholesterol, clinical trials are currently evaluating the potential to target other atherogenic lipid parameters such as triglyceride-rich lipoproteins and Lp(a), in addition to promoting the biological properties of high-density lipoproteins. Targeting inflammation within the vascular wall has emerged as a new frontier in cardiovascular prevention, with early evidence that use of anti-inflammatory agents have the potential to reduce cardiovascular risk. EXPERT OPINION Clinical practice has an increasing array of therapeutic tools to achieve more effective lowering of low-density lipoprotein cholesterol for high-risk patients. In addition, clinical trials have the potential to deliver a range of additional agents to the clinic, that target alternative lipid and inflammatory mediators. This will permit the potential to personalize cardiovascular prevention.
Collapse
Affiliation(s)
| | - Adam J Nelson
- Victorian Heart Institute, Monash University, Melbourne, Australia
| |
Collapse
|
21
|
Stroope C, Nettersheim FS, Coon B, Finney AC, Schwartz MA, Ley K, Rom O, Yurdagul A. Dysregulated cellular metabolism in atherosclerosis: mediators and therapeutic opportunities. Nat Metab 2024; 6:617-638. [PMID: 38532071 PMCID: PMC11055680 DOI: 10.1038/s42255-024-01015-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
Accumulating evidence over the past decades has revealed an intricate relationship between dysregulation of cellular metabolism and the progression of atherosclerotic cardiovascular disease. However, an integrated understanding of dysregulated cellular metabolism in atherosclerotic cardiovascular disease and its potential value as a therapeutic target is missing. In this Review, we (1) summarize recent advances concerning the role of metabolic dysregulation during atherosclerosis progression in lesional cells, including endothelial cells, vascular smooth muscle cells, macrophages and T cells; (2) explore the complexity of metabolic cross-talk between these lesional cells; (3) highlight emerging technologies that promise to illuminate unknown aspects of metabolism in atherosclerosis; and (4) suggest strategies for targeting these underexplored metabolic alterations to mitigate atherosclerosis progression and stabilize rupture-prone atheromas with a potential new generation of cardiovascular therapeutics.
Collapse
Affiliation(s)
- Chad Stroope
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Felix Sebastian Nettersheim
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Brian Coon
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Cardiovascular Biology Research Program, OMRF, Oklahoma City, OK, USA
- Department of Cell Biology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
- Immunology Center of Georgia (IMMCG), Augusta University Immunology Center of Georgia, Augusta, GA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
22
|
Xie S, Galimberti F, Olmastroni E, Luscher TF, Carugo S, Catapano AL, Casula M. Effect of lipid-lowering therapies on C-reactive protein levels: a comprehensive meta-analysis of randomized controlled trials. Cardiovasc Res 2024; 120:333-344. [PMID: 38373008 PMCID: PMC10981526 DOI: 10.1093/cvr/cvae034] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/15/2023] [Accepted: 12/14/2023] [Indexed: 02/20/2024] Open
Abstract
Chronic low-degree inflammation is a hallmark of atherosclerotic cardiovascular (CV) disease. To assess the effect of lipid-lowering therapies on C-reactive protein (CRP), a biomarker of inflammation, we conducted a meta-analysis according to the PRISMA guidelines. Databases were searched from inception to July 2023. Inclusion criteria were: (i) randomized controlled trials (RCTs) in human, Phase II, III, or IV; (ii) English language; (iii) comparing the effect of lipid-lowering drugs vs. placebo; (iv) reporting the effects on CRP levels; (v) with intervention duration of more than 3 weeks; (vi) and sample size (for both intervention and control group) over than 100 subjects. The between-group (treatment-placebo) CRP absolute mean differences and 95% confidence intervals were calculated for each drug class separately. A total of 171 668 subjects from 53 RCTs were included. CRP levels (mg/L) were significantly decreased by statins [-0.65 (-0.87 to -0.43), bempedoic acid; -0.43 (-0.67 to -0.20), ezetimibe; -0.28 (-0.48 to -0.08)], and omega-3 fatty acids [omega3FAs, -0.27 (-0.52 to -0.01)]. CRP was reduced by -0.40 (-1.17 to 0.38) with fibrates, although not statistically significant. A slight increase of CRP concentration was observed for proprotein convertase subtilisin/kexin type 9 inhibitors [0.11 (0.07-0.14)] and cholesteryl-ester transfer protein inhibitors [0.10 (0.00-0.21)], the latter being not statistically significant. Meta-regression analysis did not show a significant correlation between changes in CRP and LDL cholesterol (LDL-C) or triglycerides. Statins, bempedoic acid, ezetimibe, and omega3FAs significantly reduce serum CRP concentration, independently of LDL-C reductions. The impact of this anti-inflammatory effect in terms of CV prevention needs further investigation.
Collapse
Affiliation(s)
- Sining Xie
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20033 Milan, Italy
| | - Federica Galimberti
- IRCCS MultiMedica, via Milanese 300, 20099 Sesto San Giovanni (Milan), Italy
| | - Elena Olmastroni
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20033 Milan, Italy
- IRCCS MultiMedica, via Milanese 300, 20099 Sesto San Giovanni (Milan), Italy
| | - Thomas F Luscher
- Center for Molecular Cardiology, University Zurich, Wagistrasse 12, 8952 Schlieren (Zurich), Switzerland
- Cardiac Unit, Royal Brompton and Harefield Hospitals GSTT, Imperial College and King’s College London, Sydney Street, SW3 6NP London, UK
| | - Stefano Carugo
- Department of Clinical Sciences and Community Health, University of Milan, via della Commenda 19, 20122 Milan, Italy
- Cardiology Unit, Department of Internal Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico of Milan, via Francesco Sforza 28, 20122 Milan, Italy
| | - Alberico L Catapano
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20033 Milan, Italy
- IRCCS MultiMedica, via Milanese 300, 20099 Sesto San Giovanni (Milan), Italy
| | - Manuela Casula
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20033 Milan, Italy
- IRCCS MultiMedica, via Milanese 300, 20099 Sesto San Giovanni (Milan), Italy
| |
Collapse
|
23
|
Sharma C, Hamza A, Boyle E, Donu D, Cen Y. Post-Translational Modifications and Diabetes. Biomolecules 2024; 14:310. [PMID: 38540730 PMCID: PMC10968569 DOI: 10.3390/biom14030310] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 09/22/2024] Open
Abstract
Diabetes and its associated complications have increasingly become major challenges for global healthcare. The current therapeutic strategies involve insulin replacement therapy for type 1 diabetes (T1D) and small-molecule drugs for type 2 diabetes (T2D). Despite these advances, the complex nature of diabetes necessitates innovative clinical interventions for effective treatment and complication prevention. Accumulative evidence suggests that protein post-translational modifications (PTMs), including glycosylation, phosphorylation, acetylation, and SUMOylation, play important roles in diabetes and its pathological consequences. Therefore, the investigation of these PTMs not only sheds important light on the mechanistic regulation of diabetes but also opens new avenues for targeted therapies. Here, we offer a comprehensive overview of the role of several PTMs in diabetes, focusing on the most recent advances in understanding their functions and regulatory mechanisms. Additionally, we summarize the pharmacological interventions targeting PTMs that have advanced into clinical trials for the treatment of diabetes. Current challenges and future perspectives are also provided.
Collapse
Affiliation(s)
- Chiranjeev Sharma
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Abu Hamza
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Emily Boyle
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Dickson Donu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Yana Cen
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
24
|
Abrahams T, Nelson AJ, Nicholls SJ. How Will Our Practice Change After the CLEAR Outcomes Trial? Curr Atheroscler Rep 2024; 26:83-89. [PMID: 38294660 PMCID: PMC10881600 DOI: 10.1007/s11883-024-01188-5] [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] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
PURPOSE OF REVIEW Bempedoic acid is a novel therapeutic agent that is designed to reduce levels of low-density lipoprotein cholesterol (LDL-C). The purpose of this review is to provide the background for development of bempedoic acid, findings from clinical trials and to discuss clinical implications. RECENT FINDINGS Bempedoic acid inhibits ATP citrate lyase within the liver and reduces cholesterol synthesis, with the potential to avoid muscle symptoms experienced by patients treated with statins. Early clinical studies demonstrated that administration of bempedoic acid resulted in lowering of LDL-C by 20-30% as monotherapy and by 40-50% when combined with ezetimibe, in addition to lowering of high sensitivity C-reactive protein by 20-30%. The CLEAR Outcomes trial of high cardiovascular risk patients, with elevated LDL-C levels and either unable or unwilling to take statins demonstrated that bempedoic acid reduced the rate of major adverse cardiovascular events. A greater incidence of elevation of hepatic transaminase and creatinine, gout, and cholelithiasis were consistently observed in bempedoic acid-treated patients. Bempedoic acid presents an additional therapeutic option to achieve more effective lowering of LDL-C levels and reduction in cardiovascular risk.
Collapse
Affiliation(s)
- Timothy Abrahams
- Victorian Heart Institute, MonashUniversity, 631 Blackburn Road, Clayton, VIC, 3168, Australia
| | - Adam J Nelson
- Victorian Heart Institute, MonashUniversity, 631 Blackburn Road, Clayton, VIC, 3168, Australia
| | - Stephen J Nicholls
- Victorian Heart Institute, MonashUniversity, 631 Blackburn Road, Clayton, VIC, 3168, Australia.
| |
Collapse
|
25
|
Mormone A, Tortorella G, Esposito F, Caturano A, Marrone A, Cozzolino D, Galiero R, Marfella R, Sasso FC, Rinaldi L. Advances in Pharmacological Approaches for Managing Hypercholesterolemia: A Comprehensive Overview of Novel Treatments. Biomedicines 2024; 12:432. [PMID: 38398034 PMCID: PMC10887105 DOI: 10.3390/biomedicines12020432] [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: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Hypercholesterolemia plays a crucial role in the formation of lipid plaques, particularly with elevated low-density lipoprotein (LDL-C) levels, which are linked to increased risks of cardiovascular disease, cerebrovascular disease, and peripheral arterial disease. Controlling blood cholesterol values, specifically reducing LDL-C, is widely recognized as a key modifiable risk factor for decreasing the morbidity and mortality associated with cardiovascular diseases. Historically, statins, by inhibiting the enzyme β-hydroxy β-methylglutaryl-coenzyme A (HMG)-CoA reductase, have been among the most effective drugs. However, newer non-statin agents have since been introduced into hypercholesterolemia therapy, providing a viable alternative with a favorable cost-benefit ratio. This paper aims to delve into the latest therapies, shedding light on their mechanisms of action and therapeutic benefits.
Collapse
Affiliation(s)
- Andrea Mormone
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Giovanni Tortorella
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Francesca Esposito
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
- Department of Experimental Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy
| | - Aldo Marrone
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Domenico Cozzolino
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (A.M.); (G.T.); (F.E.); (A.C.); (A.M.); (D.C.); (R.G.); (R.M.); (F.C.S.)
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
| |
Collapse
|
26
|
Paponja K, Pećin I, Reiner Ž, Banach M. Bempedoic acid: new evidence and recommendations on use. Curr Opin Lipidol 2024; 35:41-50. [PMID: 38085172 DOI: 10.1097/mol.0000000000000911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
PURPOSES OF REVIEW Cardiovascular diseases (CVDs) are the leading cause of mortality and morbidity worldwide. Recognizing the importance of dyslipidemia treatment in the prevention of cardiovascular events has become a part of standard clinical practice. Desired values of LDL cholesterol (LDL-C) have become lower and lower in the last few decades, as evidenced by the most recent guidelines. Therefore, efforts to lower LDL cholesterol concentrations with conventional therapies and combinations of lipid-lowering therapy may not be successful in a high proportion of patients. RECENT FINDINGS Bempedoic acid is a novel agent, first in-class ATP Citrate Lyase (ACL) inhibitor, which targets biosynthesis of the cholesterol in the liver. Considering the results of phase 3 studies, it has been approved for sole use for dyslipidemia treatment for patients who are statin-intolerant or in combination with statin-ezetimibe for those suffering from familial hypercholesterolemia or ASCVD and unable to reach targeted LDL-C values. SUMMARY Bempedoic acid has proven beneficial for further reduction of LDL cholesterol for targeted groups of patients. It is not only efficient but also a well tolerated, affordable, and available agent whose place in lipid-lowering management is yet to be fully understood with new data collected from ongoing clinical research. In this review we suggest the place of bempedoic acid in lipid-lowering management.
Collapse
Affiliation(s)
- Kristina Paponja
- University Hospital Center Zagreb, Department of Internal Medicine, Division of Metabolic Diseases
| | - Ivan Pećin
- University Hospital Center Zagreb, Department of Internal Medicine, Division of Metabolic Diseases
- Zagreb School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Željko Reiner
- University Hospital Center Zagreb, Department of Internal Medicine, Division of Metabolic Diseases
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother's Memorial Hospital Research Institute
| | - Maciej Banach
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother's Memorial Hospital Research Institute
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Wang Q, Liu J, Chen Z, Zheng J, Wang Y, Dong J. Targeting metabolic reprogramming in hepatocellular carcinoma to overcome therapeutic resistance: A comprehensive review. Biomed Pharmacother 2024; 170:116021. [PMID: 38128187 DOI: 10.1016/j.biopha.2023.116021] [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: 09/18/2023] [Revised: 11/23/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Hepatocellular carcinoma (HCC) poses a heavy burden on human health with high morbidity and mortality rates. Systematic therapy is crucial for advanced and mid-term HCC, but faces a significant challenge from therapeutic resistance, weakening drug effectiveness. Metabolic reprogramming has gained attention as a key contributor to therapeutic resistance. Cells change their metabolism to meet energy demands, adapt to growth needs, or resist environmental pressures. Understanding key enzyme expression patterns and metabolic pathway interactions is vital to comprehend HCC occurrence, development, and treatment resistance. Exploring metabolic enzyme reprogramming and pathways is essential to identify breakthrough points for HCC treatment. Targeting metabolic enzymes with inhibitors is key to addressing these points. Inhibitors, combined with systemic therapeutic drugs, can alleviate resistance, prolong overall survival for advanced HCC, and offer mid-term HCC patients a chance for radical resection. Advances in metabolic research methods, from genomics to metabolomics and cells to organoids, help build the HCC metabolic reprogramming network. Recent progress in biomaterials and nanotechnology impacts drug targeting and effectiveness, providing new solutions for systemic therapeutic drug resistance. This review focuses on metabolic enzyme changes, pathway interactions, enzyme inhibitors, research methods, and drug delivery targeting metabolic reprogramming, offering valuable references for metabolic approaches to HCC treatment.
Collapse
Affiliation(s)
- Qi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Juan Liu
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 100021, China; Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China; Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Ziye Chen
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Jingjing Zheng
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yunfang Wang
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 100021, China; Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China; Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Jiahong Dong
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, China; Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 100021, China; Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China; Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, China.
| |
Collapse
|
28
|
Xiang W, Lv H, Xing F, Sun X, Ma Y, Wu L, Lv G, Zong Q, Wang L, Wu Z, Feng Q, Yang W, Wang H. Inhibition of ACLY overcomes cancer immunotherapy resistance via polyunsaturated fatty acids peroxidation and cGAS-STING activation. SCIENCE ADVANCES 2023; 9:eadi2465. [PMID: 38055816 DOI: 10.1126/sciadv.adi2465] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/27/2023] [Indexed: 12/08/2023]
Abstract
Adenosine 5'-triphosphate citrate lyase (ACLY) is a cytosolic enzyme that converts citrate into acetyl-coenzyme A for fatty acid and cholesterol biosynthesis. ACLY is up-regulated or activated in many cancers, and targeting ACLY by inhibitors holds promise as potential cancer therapy. However, the role of ACLY in cancer immunity regulation remains poorly understood. Here, we show that ACLY inhibition up-regulates PD-L1 immune checkpoint expression in cancer cells and induces T cell dysfunction to drive immunosuppression and compromise its antitumor effect in immunocompetent mice. Mechanistically, ACLY inhibition causes polyunsaturated fatty acid (PUFA) peroxidation and mitochondrial damage, which triggers mitochondrial DNA leakage to activate the cGAS-STING innate immune pathway. Pharmacological and genetic inhibition of ACLY overcomes cancer resistance to anti-PD-L1 therapy in a cGAS-dependent manner. Furthermore, dietary PUFA supplementation mirrors the enhanced efficacy of PD-L1 blockade by ACLY inhibition. These findings reveal an immunomodulatory role of ACLY and provide combinatorial strategies to overcome immunotherapy resistance in tumors.
Collapse
Affiliation(s)
- Wei Xiang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Hongwei Lv
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University (Second Military Medical University), Shanghai 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai 201805, China
| | - Fuxue Xing
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiaoyan Sun
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yue Ma
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Lu Wu
- Fourth Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai 200438, China
| | - Guishuai Lv
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University (Second Military Medical University), Shanghai 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai 201805, China
| | - Qianni Zong
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University (Second Military Medical University), Shanghai 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai 201805, China
| | - Liang Wang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University (Second Military Medical University), Shanghai 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai 201805, China
| | - Zixin Wu
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Qiyu Feng
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Wen Yang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University (Second Military Medical University), Shanghai 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai 201805, China
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology, Shanghai 200438, China
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai 200438, China
| | - Hongyang Wang
- Cancer Research Center, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University (Second Military Medical University), Shanghai 200438, China
- National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai 201805, China
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Shanghai Key Laboratory of Hepatobiliary Tumor Biology, Shanghai 200438, China
- Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai 200438, China
| |
Collapse
|
29
|
Srivastava RAK. New opportunities in the management and treatment of refractory hypercholesterolemia using in vivo CRISPR-mediated genome/base editing. Nutr Metab Cardiovasc Dis 2023; 33:2317-2325. [PMID: 37805309 DOI: 10.1016/j.numecd.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 10/09/2023]
Abstract
AIMS Refractory hypercholesterolemia (RH), caused primarily by the loss-of-function mutation of LDL receptor (LDLR) gene seen in HoFH and HeFH patients, remains a major risk factor for atherosclerotic cardiovascular disease (ASCVD). Statin and ezetimibe combination therapy lower circulating LDL by 30% in HoFH patients. PCSK9 mAB, being an LDLR-dependent therapy, is not effective in HoFH, but lowers LDL by 25% in HeFH patients. A maximum reduction of 50% was noted in HoFH patients treated with ANGPTL3 mAB, which was not enough to achieve therapeutic goal of LDL. Therefore, new approaches are warranted to offer hopes to individuals intolerant to higher dose statins and not able to achieve recommended LDL level. DATA SYNTHESIS New approaches to lower LDL include gene therapy and gene editing. AAV-based gene therapy has shown encouraging results in animal models. Using CRISPR/Cas9-mediated genome/base editing, gain of function and loss of function have been successfully done in animal models. Recent progress in the refinement of genome/base editing has overcome the issues of off-target mutagenesis with ∼1% mutagenesis in case of PCSK9 and almost no off-target mutagenesis in inactivating ANGPTL3 in animal models showing 50% reduction in cholesterol. Current approaches using CRISPR-Cas9 genome/base editing targeting LDLR-dependent and LDLR-independent pathways are underway. CONCLUSIONS The new information on gain of LDLR function and inactivation of ANGPTL3 together with developments in genome/base editing technology to overcome off-target insertion and deletion mutagenesis offer hope to refractory hypercholesterolemic individuals who are at a higher risk of developing ASCVD.
Collapse
|
30
|
Hasan I, Rainsford KD, Ross JS. Salsalate: a pleotropic anti-inflammatory drug in the treatment of diabetes, obesity, and metabolic diseases. Inflammopharmacology 2023; 31:2781-2797. [PMID: 37758933 DOI: 10.1007/s10787-023-01242-9] [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: 03/17/2023] [Accepted: 04/12/2023] [Indexed: 09/29/2023]
Abstract
Type two Diabetes Mellitus (T2DM) is a rising epidemic. Available therapeutic strategies have provided glycaemic control via HbA1c reduction but fail to provide clinically meaningful reduction in microvascular and macrovascular (cardiac, renal, ophthalmological, and neurological) complications. Inflammation is strongly linked to the pathogenesis of T2DM. Underlying inflammatory mechanisms include oxidative stress, endoplasmic reticulum stress amyloid deposition in the pancreas, lipotoxicity, and glucotoxicity. Molecular signalling mechanisms in chronic inflammation linked to obesity and diabetes include JANK, NF-kB, and AMPK pathways. These activated pathways lead to a production of various inflammatory cytokines, such as Interleukin (IL-6), tumor necrosis factor (TNF)-alpha, and C-reactive protein (CRP), which create a chronic low-grade inflammation and ultimately dysregulation of glucose homeostasis in the liver, skeletal muscle, and smooth muscle. Anti-inflammatory agents are being tested as anti-diabetic agents such as the IL-1b antagonist, Anakinra, the IL-1b inhibitor, Canakinuma, the IL-6 antagonists such as Tocilizumab, Rapamycin (Everolimus), and the IKK-beta kinase inhibitor, Salsalate. Salsalate is a century old safe anti-inflammatory drug used in the treatment of arthritis. Long-term safety and efficacy of Salsalate in the treatment of T2DM have been evaluated, which showed improved fasting plasma glucose and reduced HbA1C levels as well as reduced pro-inflammatory markers in T2DM patients. Current publication summarizes the literature review of pathophysiology of role of inflammation in T2DM and clinical efficacy and safety of Salsalate in the treatment of T2DM.
Collapse
Affiliation(s)
- I Hasan
- RH Nanopharmaceuticals LLC, 140 Ocean Ave, Monmouth Beach, New Jersey, 07750, USA.
| | - K D Rainsford
- Emeritus Professor of Biomedical Sciences, Department of Biosciences and Chemistry, BMRC, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK
| | - Joel S Ross
- RH Nanopharmaceuticals LLC, 140 Ocean Ave, Monmouth Beach, New Jersey, 07750, USA
- J & D Pharmaceuticals LLC, Monmouth County, USA
| |
Collapse
|
31
|
Sun Q, Guo Y, Hu W, Zhang M, Wang S, Lei Y, Meng H, Li N, Xu P, Li Z, Lin H, Huang F, Qiu Z. Bempedoic Acid Unveils Therapeutic Potential in Non-Alcoholic Fatty Liver Disease: Suppression of the Hepatic PXR-SLC13A5/ACLY Signaling Axis. Drug Metab Dispos 2023; 51:1628-1641. [PMID: 37684055 DOI: 10.1124/dmd.123.001449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
The hepatic SLC13A5/SLC25A1-ATP-dependent citrate lyase (ACLY) signaling pathway, responsible for maintaining the citrate homeostasis, plays a crucial role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Bempedoic acid (BA), an ACLY inhibitor commonly used for managing hypercholesterolemia, has shown promising results in addressing hepatic steatosis. This study aimed to elucidate the intricate relationships in processes of hepatic lipogenesis among SLC13A5, SLC25A1, and ACLY and to examine the therapeutic potential of BA in NAFLD, providing insights into its underlying mechanism. In murine primary hepatocytes and HepG2 cells, the silencing or pharmacological inhibition of SLC25A1/ACLY resulted in significant upregulation of SLC13A5 transcription and activity. This increase in SLC13A5 activity subsequently led to enhanced lipogenesis, indicating a compensatory role of SLC13A5 when the SLC25A1/ACLY pathway was inhibited. However, BA effectively counteracted this upregulation, reduced lipid accumulation, and ameliorated various biomarkers of NAFLD. The disease-modifying effects of BA were further confirmed in NAFLD mice. Mechanistic investigations revealed that BA could reverse the elevated transcription levels of SLC13A5 and ACLY, and the subsequent lipogenesis induced by PXR activation in vitro and in vivo. Importantly, this effect was diminished when PXR was knocked down, suggesting the involvement of the hepatic PXR-SLC13A5/ACLY signaling axis in the mechanism of BA action. In conclusion, SLC13A5-mediated extracellular citrate influx emerges as an alternative pathway to SLC25A1/ACLY in the regulation of lipogenesis in hepatocytes, BA exhibits therapeutic potential in NAFLD by suppressing the hepatic PXR-SLC13A5/ACLY signaling axis, while PXR, a key regulator in drug metabolism may be involved in the pathogenesis of NAFLD. SIGNIFICANCE STATEMENT: This work describes that bempedoic acid, an ATP-dependent citrate lyase (ACLY) inhibitor, ameliorates hepatic lipid accumulation and various hallmarks of non-alcoholic fatty liver disease. Suppression of hepatic SLC25A1-ACLY pathway upregulates SLC13A5 transcription, which in turn activates extracellular citrate influx and the subsequent DNL. Whereas in hepatocytes or the liver tissue challenged with high energy intake, bempedoic acid reverses compensatory activation of SLC13A5 via modulating the hepatic PXR-SLC13A5/ACLY axis, thereby simultaneously downregulating SLC13A5 and ACLY.
Collapse
Affiliation(s)
- Qiushuang Sun
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Yating Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Wenjun Hu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Mengdi Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Shijiao Wang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Yuanyuan Lei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Haitao Meng
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Ning Li
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Pengfei Xu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Zhiyu Li
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Haishu Lin
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Fang Huang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| | - Zhixia Qiu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy (Q.S., Y.G., F.H.), Departments of Pharmacology (W.H., S.W., Y.L., Z.Q.) and Medicinal Chemistry, School of Pharmacy (P.X., Z.L.), School of Basic Medical Sciences and Clinical Pharmacy (M.Z.), and National Experimental Teaching Demonstration Center of Pharmacy, School of Pharmacy (N.L.), China Pharmaceutical University, Nanjing, China; Shimadzu (China) Co., LTD., Nanjing Branch, Nanjing, China (H.M.); and College of Pharmacy, Shenzhen Technology University, Shenzhen, China (H.L.)
| |
Collapse
|
32
|
Albosta M, Grant JK, Michos ED. Bempedoic Acid: Lipid Lowering for Cardiovascular Disease Prevention. Heart Int 2023; 17:27-34. [PMID: 38419721 PMCID: PMC10900064 DOI: 10.17925/hi.2023.17.2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 08/22/2023] [Indexed: 03/02/2024] Open
Abstract
The management of low-density lipoprotein cholesterol (LDL-C) levels is a central strategy for the prevention of atherosclerotic cardiovascular disease. Current United States (2018 American Heart Association/American College of Cardiology/Multisociety) and European (2019 European Society of Cardiology/European Atherosclerosis Society) guidelines endorse statin therapy as the first-line therapy for pharmacologic LDL-C lowering. However, in clinical practice up to 30% of patients report partial or complete intolerance to statin therapy. While the nocebo effect with statins is well described, perceived statin intolerance prevents many patients from achieving LDL-C thresholds associated with clinical benefit. Bempedoic acid is a novel, oral, non-statin lipid-l owering therapy that works by inhibiting adenosine triphosphate-citrate lyase, an enzymatic reaction upstream of 3-hydroxy-3-methylglutaryl coenzyme A reductase in the hepatic cholesterol synthesis pathway. Bempedoic acid confers reduction in LDL-C of ~18% on background statin therapy,~21% in patients with statin intolerance, and ~38% when given in fixed-dose combination with ezetimibe. The CLEAR Outcomes trial, which enrolled high-risk primary and secondary prevention patients with reported statin intolerance and LDL-C levels ≥100 mg/dL, showed that bempedoic acid compared with placebo reduced 4-component major adverse cardiovascular events (MACE) by 13% (hazard ratio 0.87, 95% confidence interval 0.79-0.96). Bempedoic acid also reduced 3-component MACE by 15%, myocardial infarction by 23% and coronary revascularization by 19%. The benefit was even greater in the primary prevention cohort (hazard ratio 0.70, 95% confidence interval 0.55-0.89) for 4-component MACE. Bempedoic acid was associated with increases in uric acid levels and cholelithiasis, but numerically fewer events of myalgia and new-onset diabetes. These findings confirm that bempedoic acid is an effective approach to reduce cardiovascular outcomes in high-risk patients with statin intolerance who require further reduction in LDL-C.
Collapse
Affiliation(s)
- Michael Albosta
- Internal Medicine Department, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, FL, USA
| | - Jelani K Grant
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Erin D Michos
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
33
|
Zhang J, Guan X, Zhang B, Wang J, Jin X, Zhao Y, Li B. Bempedoic Acid can Reduce Cardiovascular Events in Combination with Statins or As Monotherapy: A Systematic Review and Meta-analysis. Am J Cardiovasc Drugs 2023; 23:695-708. [PMID: 37672202 DOI: 10.1007/s40256-023-00606-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 09/07/2023]
Abstract
AIM Bempedoic acid has shown noteworthy progress in the prevention and management of atherosclerotic cardiovascular disease (ASCVD) in recent years. However, there has been a lack of high-quality evidence regarding the risk reduction of clinical events with bempedoic acid. Therefore, the aim of this article is to conduct a comprehensive evaluation of the impact of bempedoic acid on the incidence of cardiovascular events. METHODS A systematic review and meta-analysis of randomized controlled trials pertaining to bempedoic acid was carried out. We conducted a systematic search across the Pubmed, Embase, and Cochrane Central Register of Controlled Trials databases to identify relevant studies published from inception to 23 April 2023. A total of four trials comparing the clinical benefit achieved with bempedoic acid versus placebo were included. RESULTS Our analysis comprised four trials that encompassed a total of 17,323 patients. In comparison to the placebo, bempedoic acid showed a significant reduction in the risk of major adverse cardiovascular events (MACE) [relative risk (RR), 0.86, 95% confidence interval (CI) 0.87-0.94]. Additionally, bempedoic acid substantially lowered the occurrence of fatal or nonfatal myocardial infarction (RR 0.76, 95% CI 0.66-0.89), hospitalization for unstable angina (RR 0.70, 95% CI 0.55-0.89), and coronary revascularization (RR 0.82, 95% CI 0.73-0.92). There was also a similar reduction in MACE in patients on the maximally tolerated statin therapy. CONCLUSION Bempedoic acid may reduce the risk of cardiovascular events regardless of whether the patient is taking stains or not. REGISTRATION PROSPERO registration number CRD42023422932.
Collapse
Affiliation(s)
- Ju Zhang
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Xiangfeng Guan
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Baixue Zhang
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Jia Wang
- Department of Cardiology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou City, 256603, Shandong Province, People's Republic of China
| | - Xiaodong Jin
- Department of Geriatrics, Zibo Central Hospital, No. 10, South Shanghai Road, Zibo, People's Republic of China.
| | - Yunhe Zhao
- Department of Cardiology, Zibo Central Hospital Affiliated to Binzhou Medical College, NO.10, South Shanghai Road, Zibo, People's Republic of China.
| | - Bo Li
- Department of Cardiology, Zibo Central Hospital Affiliated to Binzhou Medical College, NO.10, South Shanghai Road, Zibo, People's Republic of China.
| |
Collapse
|
34
|
Alunno A, Carubbi F, Campanozzi E, Bellisario F, Schoones JW, Mariani FM, Di Ruscio E, Altieri P, Ferri C. Untangling the relationship between bempedoic acid and gout: results from a systematic literature review. Front Cardiovasc Med 2023; 10:1234601. [PMID: 37953764 PMCID: PMC10634504 DOI: 10.3389/fcvm.2023.1234601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Background Bempedoic acid (BA) is a small-molecule first-in-class of inhibitor of ATP citrate lyase that significantly lowers low-density lipoproteins cholesterol (LDL-c) in statin-intolerant and inadequate responders. Increased serum uric acid (SUA) levels and gout incidence have been described in BA-treated patients. The aim of this systematic review was to investigate the safety of BA regarding SUA levels and gout in randomised controlled trials (RCTs). Methods A search on 7 databases was performed from inception to May 4, 2023. RCTs of BA monotherapy or combination with other lipid-lowering treatment (LLT) in patients with increased LDL-c were included. Dual data extraction was performed with disagreements resolved through consensus. Due to the methodological purpose of this review risk-of-bias assessment of studies was not performed. Results 6 Phase 3 RCTs (N = 17,975 patients of which 9,635 received BA) 9 Phase 2 RCTs (N = 362 patients of which 170 received BA) and an open-label extension of a Phase 3 RCT were included. Gout and/or hyperuricemia were not mentioned as exclusion criteria, previous/current use of urate-lowering therapies (ULT) and/or colchicine and/or dietary patterns were not reported. Phase 3 RCTs: 2 studies specified the number of patients experiencing hyperuricemia over the study period (BA: 4.9%-11%; placebo: 1.9%-5.6%) and the effect size was significant only in 1 study (OR = 2.0, 95% CI 1.8-2.3). Four RCTs reported a higher incidence of gout in the BA arm however, when we calculated the effect size, it was small and often not significant. Two studies reported 0 cases of gout. The paucity of information about SUA levels at baseline and/or at the end of follow-up do not allow us to quantify the effect sizes for BA-induced SUA elevation. Data on gout from Phase 2 RCTs is scant. Conclusions Data from phase 2 and 3 RCTs do not allow for confirming a clear association between BA and gout. It is conceivable that a careful assessment of SUA levels/history of gout at baseline and the concomitant use of urate-lowering agents may be instrumental to minimise the risk of new-onset gout/gout flares in patients treated with BA.
Collapse
Affiliation(s)
- Alessia Alunno
- Department of Clinical Medicine, Life, Health & Environmental Sciences, Internal Medicine and Nephrology Division, ASL1 Avezzano-Sulmona-L'Aquila, San Salvatore Hospital, University of L’Aquila, L'Aquila, Italy
| | - Francesco Carubbi
- Department of Clinical Medicine, Life, Health & Environmental Sciences, Internal Medicine and Nephrology Division, ASL1 Avezzano-Sulmona-L'Aquila, San Salvatore Hospital, University of L’Aquila, L'Aquila, Italy
| | - Elena Campanozzi
- Department of Clinical Medicine, Life, Health & Environmental Sciences, Internal Medicine and Nephrology Division, ASL1 Avezzano-Sulmona-L'Aquila, San Salvatore Hospital, University of L’Aquila, L'Aquila, Italy
| | - Federico Bellisario
- Department of Clinical Medicine, Life, Health & Environmental Sciences, Internal Medicine and Nephrology Division, ASL1 Avezzano-Sulmona-L'Aquila, San Salvatore Hospital, University of L’Aquila, L'Aquila, Italy
| | - Jan W. Schoones
- Directorate of Research Policy, Leiden University Medical Center, Leiden, Netherlands
| | - Francesco Maria Mariani
- Department of Clinical Medicine, Life, Health & Environmental Sciences, Internal Medicine and Nephrology Division, ASL1 Avezzano-Sulmona-L'Aquila, San Salvatore Hospital, University of L’Aquila, L'Aquila, Italy
| | - Evy Di Ruscio
- Department of Clinical Medicine, Life, Health & Environmental Sciences, Internal Medicine and Nephrology Division, ASL1 Avezzano-Sulmona-L'Aquila, San Salvatore Hospital, University of L’Aquila, L'Aquila, Italy
| | - Piera Altieri
- Department of Clinical Medicine, Life, Health & Environmental Sciences, Internal Medicine and Nephrology Division, ASL1 Avezzano-Sulmona-L'Aquila, San Salvatore Hospital, University of L’Aquila, L'Aquila, Italy
| | - Claudio Ferri
- Department of Clinical Medicine, Life, Health & Environmental Sciences, Internal Medicine and Nephrology Division, ASL1 Avezzano-Sulmona-L'Aquila, San Salvatore Hospital, University of L’Aquila, L'Aquila, Italy
| |
Collapse
|
35
|
Amore BM, MacDougall DE, Hanselman JC, Emery MG. Phase 1, Single- and Multiple-Ascending-Dose, Food-Effect, and East Asian Subject Studies to Assess the Pharmacokinetics, Safety, and Tolerability of Bempedoic Acid, a Selective Inhibitor of Adenosine Triphosphate Citrate Lyase. Clin Pharmacol Drug Dev 2023; 12:1022-1035. [PMID: 37477389 DOI: 10.1002/cpdd.1297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/23/2023] [Indexed: 07/22/2023]
Abstract
Bempedoic acid is an adenosine triphosphate citrate lyase inhibitor that lowers low-density lipoprotein cholesterol by inhibiting cholesterol synthesis and upregulating hepatic low-density lipoprotein receptor expression. After oral dosing, bempedoic acid was readily absorbed, attaining maximum concentrations with a median time of 3.5 hours, and may be taken without regard to food. Steady-state oral pharmacokinetics in healthy adults receiving bempedoic acid at the approved 180 mg/day dose were characterized by mean maximum concentration of 20.6 µg/mL, area under the concentration-time curve over 24 hours of 289 µg·h/mL, and elimination half-life of 21.1 hours. Multiple-dose pharmacokinetics were linear at bempedoic acid doses of 120-220 mg/day. Circulating concentrations of the active metabolite ESP15228 were 18.0% of bempedoic acid concentrations on average. Comparisons of bempedoic acid 180 mg/day pharmacokinetics after single and multiple dosing revealed no clinically meaningful differences between Japanese, Chinese, and Western subjects. Mean estimates of bempedoic acid elimination half-life in Japanese (25.2 hours) and Chinese (20.0 hours) subjects were comparable to Western subjects (23.9 hours) following 14 days of once-daily dosing. Bempedoic acid was generally safe and well tolerated up to a dose of 220 mg/day across the study populations described herein.
Collapse
|
36
|
Desjardins EM, Wu J, Lavoie DCT, Ahmadi E, Townsend LK, Morrow MR, Wang D, Tsakiridis EE, Batchuluun B, Fayyazi R, Kwiecien JM, Tsakiridis T, Lally JSV, Paré G, Pinkosky SL, Steinberg GR. Combination of an ACLY inhibitor with a GLP-1R agonist exerts additive benefits on nonalcoholic steatohepatitis and hepatic fibrosis in mice. Cell Rep Med 2023; 4:101193. [PMID: 37729871 PMCID: PMC10518624 DOI: 10.1016/j.xcrm.2023.101193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/17/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023]
Abstract
Increased liver de novo lipogenesis (DNL) is a hallmark of nonalcoholic steatohepatitis (NASH). A key enzyme controlling DNL upregulated in NASH is ATP citrate lyase (ACLY). In mice, inhibition of ACLY reduces liver steatosis, ballooning, and fibrosis and inhibits activation of hepatic stellate cells. Glucagon-like peptide-1 receptor (GLP-1R) agonists lower body mass, insulin resistance, and steatosis without improving fibrosis. Here, we find that combining an inhibitor of liver ACLY, bempedoic acid, and the GLP-1R agonist liraglutide reduces liver steatosis, hepatocellular ballooning, and hepatic fibrosis in a mouse model of NASH. Liver RNA analyses revealed additive downregulation of pathways that are predictive of NASH resolution, reductions in the expression of prognostically significant genes compared with clinical NASH samples, and a predicted gene signature profile that supports fibrosis resolution. These findings support further investigation of this combinatorial therapy to treat obesity, insulin resistance, hypercholesterolemia, steatohepatitis, and fibrosis in people with NASH.
Collapse
Affiliation(s)
- Eric M Desjardins
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jianhan Wu
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada; Population Health Research Institute, McMaster University, Hamilton, ON L8L 2X2, Canada
| | - Declan C T Lavoie
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Elham Ahmadi
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Logan K Townsend
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Marisa R Morrow
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Dongdong Wang
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Evangelia E Tsakiridis
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Battsetseg Batchuluun
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Russta Fayyazi
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jacek M Kwiecien
- Department of Pathology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Theodoros Tsakiridis
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Department of Oncology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - James S V Lally
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Guillaume Paré
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Population Health Research Institute, McMaster University, Hamilton, ON L8L 2X2, Canada; Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON L8L 2X2, Canada
| | | | - Gregory R Steinberg
- Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton ON L8S 4L8, Canada; Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada.
| |
Collapse
|
37
|
Park JK, Balarbar N, Agarwala A. Bempedoic Acid: A Contemporary Review of Its Pharmacology, Efficacy, and Safety Profile, Including Recent Data from the CLEAR Outcomes Clinical Trial. Curr Cardiol Rep 2023; 25:969-978. [PMID: 37405598 DOI: 10.1007/s11886-023-01911-9] [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] [Accepted: 06/18/2023] [Indexed: 07/06/2023]
Abstract
PURPOSE OF REVIEW To provide an updated review of bempedoic acid's clinical application in lowering LDL-C in the setting of statin intolerance and the recent findings in the CLEAR Outcomes trial as well as summarize and synthesize the current state of knowledge regarding bempedoic acid while providing an in-depth analysis of the drug's pharmacological properties, mechanism of action, clinical trials, safety, and efficacy. RECENT FINDINGS The CLEAR Outcomes trial has provided evidence to support bempedoic acid as a viable alternative to statins for the primary and secondary prevention of cardiovascular disease. Bempedoic acid is a promising treatment option for patients with hypercholesterolemia who are unable to tolerate statin therapy or require additional LDL-C reduction in the treatment of cardiovascular disease, with the newest lipid-lowering cardiovascular outcomes trials expanding on their generalizability particularly in the inclusion of women.
Collapse
Affiliation(s)
- Jong Kun Park
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Noah Balarbar
- Center for Cardiovascular Disease Prevention, Baylor Scott and White Health Heart Hospital Baylor Plano, Plano, TX, USA
| | - Anandita Agarwala
- Center for Cardiovascular Disease Prevention, Baylor Scott and White Health Heart Hospital Baylor Plano, Plano, TX, USA.
| |
Collapse
|
38
|
Uehara K, Santoleri D, Whitlock AEG, Titchenell PM. Insulin Regulation of Hepatic Lipid Homeostasis. Compr Physiol 2023; 13:4785-4809. [PMID: 37358513 PMCID: PMC10760932 DOI: 10.1002/cphy.c220015] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
The incidence of obesity, insulin resistance, and type II diabetes (T2DM) continues to rise worldwide. The liver is a central insulin-responsive metabolic organ that governs whole-body metabolic homeostasis. Therefore, defining the mechanisms underlying insulin action in the liver is essential to our understanding of the pathogenesis of insulin resistance. During periods of fasting, the liver catabolizes fatty acids and stored glycogen to meet the metabolic demands of the body. In postprandial conditions, insulin signals to the liver to store excess nutrients into triglycerides, cholesterol, and glycogen. In insulin-resistant states, such as T2DM, hepatic insulin signaling continues to promote lipid synthesis but fails to suppress glucose production, leading to hypertriglyceridemia and hyperglycemia. Insulin resistance is associated with the development of metabolic disorders such as cardiovascular and kidney disease, atherosclerosis, stroke, and cancer. Of note, nonalcoholic fatty liver disease (NAFLD), a spectrum of diseases encompassing fatty liver, inflammation, fibrosis, and cirrhosis, is linked to abnormalities in insulin-mediated lipid metabolism. Therefore, understanding the role of insulin signaling under normal and pathologic states may provide insights into preventative and therapeutic opportunities for the treatment of metabolic diseases. Here, we provide a review of the field of hepatic insulin signaling and lipid regulation, including providing historical context, detailed molecular mechanisms, and address gaps in our understanding of hepatic lipid regulation and the derangements under insulin-resistant conditions. © 2023 American Physiological Society. Compr Physiol 13:4785-4809, 2023.
Collapse
Affiliation(s)
- Kahealani Uehara
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dominic Santoleri
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna E. Garcia Whitlock
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul M. Titchenell
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Srivastava RAK. A Review of Progress on Targeting LDL Receptor-Dependent and -Independent Pathways for the Treatment of Hypercholesterolemia, a Major Risk Factor of ASCVD. Cells 2023; 12:1648. [PMID: 37371118 DOI: 10.3390/cells12121648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Since the discovery of the LDL receptor in 1973 by Brown and Goldstein as a causative protein in hypercholesterolemia, tremendous amounts of effort have gone into finding ways to manage high LDL cholesterol in familial hypercholesterolemic (HoFH and HeFH) individuals with loss-of-function mutations in the LDL receptor (LDLR) gene. Statins proved to be the first blockbuster drug, helping both HoFH and HeFH individuals by inhibiting the cholesterol synthesis pathway rate-limiting enzyme HMG-CoA reductase and inducing the LDL receptor. However, statins could not achieve the therapeutic goal of LDL. Other therapies targeting LDLR include PCSK9, which lowers LDLR by promoting LDLR degradation. Inducible degrader of LDLR (IDOL) also controls the LDLR protein, but an IDOL-based therapy is yet to be developed. Among the LDLR-independent pathways, such as angiopoietin-like 3 (ANGPTL3), apolipoprotein (apo) B, apoC-III and CETP, only ANGPTL3 offers the advantage of treating both HoFH and HeFH patients and showing relatively better preclinical and clinical efficacy in animal models and hypercholesterolemic individuals, respectively. While loss-of-LDLR-function mutations have been known for decades, gain-of-LDLR-function mutations have recently been identified in some individuals. The new information on gain of LDLR function, together with CRISPR-Cas9 genome/base editing technology to target LDLR and ANGPTL3, offers promise to HoFH and HeFH individuals who are at a higher risk of developing atherosclerotic cardiovascular disease (ASCVD).
Collapse
Affiliation(s)
- Rai Ajit K Srivastava
- Integrated Pharma Solutions LLC, Boston, MA 02101-02117, USA
- College of Professional Studies, Northeastern University, Boston, MA 02101-02117, USA
| |
Collapse
|
40
|
Piccirillo F, Mastroberardino S, Nusca A, Frau L, Guarino L, Napoli N, Ussia GP, Grigioni F. Novel Antidiabetic Agents and Their Effects on Lipid Profile: A Single Shot for Several Cardiovascular Targets. Int J Mol Sci 2023; 24:10164. [PMID: 37373310 PMCID: PMC10299555 DOI: 10.3390/ijms241210164] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Type-2 diabetes mellitus (DM) represents one of the most important risk factors for cardiovascular diseases (CVD). Hyperglycemia and glycemic variability are not the only determinant of the increased cardiovascular (CV) risk in diabetic patients, as a frequent metabolic disorder associated with DM is dyslipidemia, characterized by hypertriglyceridemia, decreased high-density lipoprotein (HDL) cholesterol levels and a shift towards small dense low-density lipoprotein (LDL) cholesterol. This pathological alteration, also called diabetic dyslipidemia, represents a relevant factor which could promotes atherosclerosis and subsequently an increased CV morbidity and mortality. Recently, the introduction of novel antidiabetic agents, such as sodium glucose transporter-2 inhibitors (SGLT2i), dipeptidyl peptidase-4 inhibitors (DPP4i) and glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1 RAs), has been associated with a significant improvement in CV outcomes. Beyond their known action on glycemia, their positive effects on the CV system also seems to be related to an ameliorated lipidic profile. In this context, this narrative review summarizes the current knowledge regarding these novel anti-diabetic drugs and their effects on diabetic dyslipidemia, which could explain the provided global benefit to the cardiovascular system.
Collapse
Affiliation(s)
- Francesco Piccirillo
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (S.M.); (L.F.); (L.G.); (N.N.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Sara Mastroberardino
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (S.M.); (L.F.); (L.G.); (N.N.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Annunziata Nusca
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (S.M.); (L.F.); (L.G.); (N.N.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Lorenzo Frau
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (S.M.); (L.F.); (L.G.); (N.N.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Lorenzo Guarino
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (S.M.); (L.F.); (L.G.); (N.N.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Nicola Napoli
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (S.M.); (L.F.); (L.G.); (N.N.); (G.P.U.); (F.G.)
- Research Unit of Endocrinology and Diabetes Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Gian Paolo Ussia
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (S.M.); (L.F.); (L.G.); (N.N.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Francesco Grigioni
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (F.P.); (S.M.); (L.F.); (L.G.); (N.N.); (G.P.U.); (F.G.)
- Research Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
41
|
Bai R, Meng Y, Cui J. Therapeutic strategies targeting metabolic characteristics of cancer cells. Crit Rev Oncol Hematol 2023:104037. [PMID: 37236409 DOI: 10.1016/j.critrevonc.2023.104037] [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: 01/07/2023] [Revised: 04/26/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023] Open
Abstract
Metabolic reprogramming is one of the important characteristics of cancer and is a key process leading to malignant proliferation, tumor development and treatment resistance. A variety of therapeutic drugs targeting metabolic reaction enzymes, transport receptors, and special metabolic processes have been developed. In this review, we investigate the characteristics of multiple metabolic changes in cancer cells, including glycolytic pathways, lipid metabolism, and glutamine metabolism changes, describe how these changes promote tumor development and tumor resistance, and summarize the progress and challenges of therapeutic strategies targeting various links of tumor metabolism in combination with current study data.
Collapse
Affiliation(s)
- Rilan Bai
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
| | - Ying Meng
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
42
|
Kuna RS, Kumar A, Wessendorf-Rodriguez KA, Galvez H, Green CR, McGregor GH, Cordes T, Shaw RJ, Svensson RU, Metallo CM. Inter-organelle cross-talk supports acetyl-coenzyme A homeostasis and lipogenesis under metabolic stress. SCIENCE ADVANCES 2023; 9:eadf0138. [PMID: 37134162 PMCID: PMC10156121 DOI: 10.1126/sciadv.adf0138] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 04/03/2023] [Indexed: 05/05/2023]
Abstract
Proliferating cells rely on acetyl-CoA to support membrane biogenesis and acetylation. Several organelle-specific pathways are available for provision of acetyl-CoA as nutrient availability fluctuates, so understanding how cells maintain acetyl-CoA homeostasis under such stresses is critically important. To this end, we applied 13C isotope tracing cell lines deficient in these mitochondrial [ATP-citrate lyase (ACLY)]-, cytosolic [acetyl-CoA synthetase (ACSS2)]-, and peroxisomal [peroxisomal biogenesis factor 5 (PEX5)]-dependent pathways. ACLY knockout in multiple cell lines reduced fatty acid synthesis and increased reliance on extracellular lipids or acetate. Knockout of both ACLY and ACSS2 (DKO) severely stunted but did not entirely block proliferation, suggesting that alternate pathways can support acetyl-CoA homeostasis. Metabolic tracing and PEX5 knockout studies link peroxisomal oxidation of exogenous lipids as a major source of acetyl-CoA for lipogenesis and histone acetylation in cells lacking ACLY, highlighting a role for inter-organelle cross-talk in supporting cell survival in response to nutrient fluctuations.
Collapse
Affiliation(s)
- Ramya S. Kuna
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Avi Kumar
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Karl A. Wessendorf-Rodriguez
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hector Galvez
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Courtney R. Green
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Grace H. McGregor
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Thekla Cordes
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig 38106, Germany
| | - Reuben J. Shaw
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | - Christian M. Metallo
- Department of Molecular and Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
43
|
Kumric M, Urlic H, Bozic J, Vilovic M, Ticinovic Kurir T, Glavas D, Miric D, Zanchi J, Bradaric-Slujo A, Lozo M, Borovac JA. Emerging Therapies for the Treatment of Atherosclerotic Cardiovascular Disease: From Bench to Bedside. Int J Mol Sci 2023; 24:8062. [PMID: 37175766 PMCID: PMC10178593 DOI: 10.3390/ijms24098062] [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: 03/31/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Primarily a consequence of sedentary lifestyle, atherosclerosis has already reached pandemic proportions, and with every year the burden of it is only increasing. As low-density lipoprotein cholesterol (LDL-C) represents a crucial factor in atherosclerosis formation and progression, stringent lipid-lowering therapy could conceivably be the key to preventing the unfavorable outcomes that arise as a consequence of atherosclerosis. The use of statins in lipid-lowering is often burdened by adverse events or is insufficient to prevent cardiovascular events as a monotherapy. Therefore, in the present review, the authors aimed to discuss the underlying mechanisms of dyslipidemia and associated atherosclerotic cardiovascular disease (ASCVD) and preclinical and clinical trials of novel therapeutic approaches to its treatment, some of which are still in the early stages of development. Apart from novel therapies, a novel change in perspective is needed. Specifically, the critical objective in the future management of ASCVD is to embrace emerging evidence in the field of atherosclerosis, because clinicians are often burden by common practice and personal experience, both of which have so far been shown to be futile in the setting of atherosclerosis.
Collapse
Affiliation(s)
- Marko Kumric
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia; (M.K.)
| | - Hrvoje Urlic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia; (M.K.)
| | - Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia; (M.K.)
| | - Marino Vilovic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia; (M.K.)
| | - Tina Ticinovic Kurir
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia; (M.K.)
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Hospital of Split, 21000 Split, Croatia
| | - Duska Glavas
- Cardiovascular Diseases Department, University Hospital of Split, 21000 Split, Croatia
| | - Dino Miric
- Cardiovascular Diseases Department, University Hospital of Split, 21000 Split, Croatia
| | - Jaksa Zanchi
- Cardiovascular Diseases Department, University Hospital of Split, 21000 Split, Croatia
| | - Anteo Bradaric-Slujo
- Cardiovascular Diseases Department, University Hospital of Split, 21000 Split, Croatia
| | - Mislav Lozo
- Cardiovascular Diseases Department, University Hospital of Split, 21000 Split, Croatia
| | - Josip A. Borovac
- Cardiovascular Diseases Department, University Hospital of Split, 21000 Split, Croatia
| |
Collapse
|
44
|
Stroes ESG, Bays HE, Banach M, Catapano AL, Duell PB, Laufs U, Mancini GBJ, Ray KK, Sasiela WJ, Zhang Y, Gotto AM. Bempedoic acid lowers high-sensitivity C-reactive protein and low-density lipoprotein cholesterol: Analysis of pooled data from four phase 3 clinical trials. Atherosclerosis 2023; 373:1-9. [PMID: 37075696 DOI: 10.1016/j.atherosclerosis.2023.03.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND AND AIMS High-sensitivity C-reactive protein (hsCRP), a marker for atherosclerotic cardiovascular disease risk, is reduced by bempedoic acid. We assessed the relationship between changes in low-density lipoprotein cholesterol (LDL-C) and hsCRP in relation to baseline statin use. METHODS Pooled data from four phase 3 trials (patients on maximally tolerated statins [Pool 1] and patients receiving no or low-dose statins [Pool 2]) were used to determine the proportion of patients with baseline hsCRP ≥2 mg/L who achieved hsCRP <2 mg/L at week 12. The percentage of patients who achieved hsCRP <2 mg/L and guideline-recommended LDL-C (Pool 1, <70 mg/dL; Pool 2, <100 mg/dL) was determined for patients on statins in Pool 1 and those not on statins in Pool 2, as was the correlation between percent changes in hsCRP and LDL-C. RESULTS Overall, 38.7% in Pool 1 and 40.7% in Pool 2 with baseline hsCRP ≥2 mg/L achieved hsCRP <2 mg/L with bempedoic acid, with little effect from background statin. Among patients taking a statin in Pool 1 or not taking a statin in Pool 2, 68.6% and 62.4% achieved hsCRP <2 mg/L. Both hsCRP <2 mg/L and United States guideline-recommended LDL-C were achieved more often with bempedoic acid vs. placebo (20.8% vs. 4.3%, respectively, in Pool 1 and 32.0% vs. 5.3%, in Pool 2). Changes in hsCRP and LDL-C were only weakly correlated (Pool 1, r = 0.112; Pool 2, r = 0.173). CONCLUSIONS Bempedoic acid significantly reduced hsCRP irrespective of background statin therapy; the effect was largely independent of LDL-C lowering.
Collapse
Affiliation(s)
- Erik S G Stroes
- Department of Vascular Medicine, Academic Medical Centre, Amsterdam, the Netherlands.
| | - Harold E Bays
- Louisville Metabolic and Atherosclerosis Research Center, Louisville, KY, USA
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Łódź and Polish Mother's Memorial Hospital Research Institute (PMMHRI), Łódź, Poland
| | | | - P Barton Duell
- Knight Cardiovascular Institute, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - G B John Mancini
- Division of Cardiology, Centre for Cardiovascular Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College London, London, UK
| | | | - Yang Zhang
- Esperion Therapeutics, Inc, Ann Arbor, MI, USA
| | - Antonio M Gotto
- Houston Methodist Research Institute, Houston, TX, USA; Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
45
|
Roglans N, Laguna JC, Alegret M. Bempedoic acid for nonalcoholic fatty liver disease: evidence and mechanisms of action. Curr Opin Lipidol 2023:00041433-990000000-00034. [PMID: 36942869 DOI: 10.1097/mol.0000000000000878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
PURPOSE OF REVIEW Nonalcoholic fatty liver disease (NAFLD) is a highly prevalent progressive condition that lacks a specific pharmacological treatment. ATP-citrate lyase (ACLY) is one of the emergent targets for the treatment of NAFLD. This review aims to summarize the role of ACLY in NAFLD, provide evidences of the beneficial effects of the ACLY inhibitor bempedoic acid (BemA) in NAFLD and discuss the mechanisms involved. RECENT FINDINGS BemA is effective in reducing hepatic steatosis in several animal models that recapitulate different stages of the disease. Thus, in a dietary model of simple hepatic steatosis in female rats, BemA abrogates the accumulation of liver fat. Apart from ACLY inhibition, BemA has several functions in the liver that contribute to the antisteatotic effect: inhibition of ketohexokinase, induction of patatin-like phospholipase domain-containing protein 3 and increases in both fatty acid β-oxidation activity and hepatic H2S production. In models of the advanced phases of NAFLD, BemA reduces not only steatosis, but also ballooning, lobular inflammation and hepatic fibrosis, by mechanisms involving both hepatocytes and hepatic stellate cells. SUMMARY BemA, an ACLY inhibitor currently approved for the treatment of hypercholesterolemia, may be a useful drug to treat NAFLD through its antisteatotic, anti-inflammatory and antifibrotic effects.
Collapse
Affiliation(s)
- Núria Roglans
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science
- Institute of Biomedicine, University of Barcelona, Barcelona
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Juan Carlos Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science
- Institute of Biomedicine, University of Barcelona, Barcelona
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science
- Institute of Biomedicine, University of Barcelona, Barcelona
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
46
|
Ahmed AA, Mohamed SK, Nofal S, El Morsy EM, Ahmed AAE. Effect of bempedoic acid on angiotensin-II induced hypertension and vascular tissue remodelling in renal hypertensive rats through AMPK multiple signalling pathways modulation. Life Sci 2023; 320:121573. [PMID: 36931497 DOI: 10.1016/j.lfs.2023.121573] [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: 11/23/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Angiotensin II (Ang II), the effector of the renin-angiotensin system (RAS), is a key player in the pathogenesis of chronic hypertension, accompanied by vascular tissue resistance, remodelling, and damage. Chronic activation of Ang II receptor 1 (AT-1R) impairs multiple cellular targets implicated in cellular protection and survival, including adenosine Monophosphate-activated protein kinase (AMPK) signalling. In addition, it induces oxidative damage, endoplasmic reticulum (ER) stress, and fibrotic changes in resistance vessels. Our study investigated the antihypertensive and antifibrotic effects of bempedoic acid, a first-in-class antihyperlipidemic drug that targets adenosine triphosphate-citrate lyase enzyme to inhibit cholesterol synthesis. We also studied the modulation of multiple AMPK signalling pathways by bempedoic acid in a chronic hypertension model in rats. Sixty male Sprague-Dawley rats were divided into four groups: sham group, hypertensive group, standard captopril group, and bempedoic treated group. All groups underwent left renal artery ligation except the sham group. Fourteen days post-surgery, captopril and bempedoic acid were administered with a dose of 30 mg/kg/day orally to captopril-standard and bempedoic acid-treated groups for two weeks, respectively. In mesenteric resistance arteries, bempedoic acid activated AMPK energy independently and augmented AMPK multiple cellular targets to adapt to Ang II-induced cellular stress. It exerted antioxidant activity, increased endothelial nitric oxide synthase, and reversed the ER stress. Bempedoic acid maintained vascular integrity and prevented vascular remodelling by inhibiting extracellular signal-regulated kinase (ERK)/transforming growth factor-β fibrotic pathway. These effects were reflected in the improved hemodynamic measurements.
Collapse
Affiliation(s)
- Asmaa A Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt.
| | - Shimaa K Mohamed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt
| | - Shahira Nofal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt
| | - Engy M El Morsy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt
| | - Amany A E Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Egypt
| |
Collapse
|
47
|
Sola-García A, Cáliz-Molina MÁ, Espadas I, Petr M, Panadero-Morón C, González-Morán D, Martín-Vázquez ME, Narbona-Pérez ÁJ, López-Noriega L, Martínez-Corrales G, López-Fernández-Sobrino R, Castillo-Peña A, Carmona-Marin LM, Martínez-Force E, Yanes O, Vinaixa M, López-López D, Reyes JC, Dopazo J, Martín F, Gauthier BR, Scheibye-Knudsen M, Capilla-González V, Martín-Montalvo A. Metabolic reprogramming by Acly inhibition using SB-204990 alters glucoregulation and modulates molecular mechanisms associated with aging. Commun Biol 2023; 6:250. [PMID: 36890357 PMCID: PMC9995519 DOI: 10.1038/s42003-023-04625-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 02/23/2023] [Indexed: 03/10/2023] Open
Abstract
ATP-citrate lyase is a central integrator of cellular metabolism in the interface of protein, carbohydrate, and lipid metabolism. The physiological consequences as well as the molecular mechanisms orchestrating the response to long-term pharmacologically induced Acly inhibition are unknown. We report here that the Acly inhibitor SB-204990 improves metabolic health and physical strength in wild-type mice when fed with a high-fat diet, while in mice fed with healthy diet results in metabolic imbalance and moderated insulin resistance. By applying a multiomic approach using untargeted metabolomics, transcriptomics, and proteomics, we determined that, in vivo, SB-204990 plays a role in the regulation of molecular mechanisms associated with aging, such as energy metabolism, mitochondrial function, mTOR signaling, and folate cycle, while global alterations on histone acetylation are absent. Our findings indicate a mechanism for regulating molecular pathways of aging that prevents the development of metabolic abnormalities associated with unhealthy dieting. This strategy might be explored for devising therapeutic approaches to prevent metabolic diseases.
Collapse
Affiliation(s)
- Alejandro Sola-García
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - María Ángeles Cáliz-Molina
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Isabel Espadas
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Michael Petr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Tracked.bio, Copenhagen, Denmark
| | - Concepción Panadero-Morón
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Daniel González-Morán
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - María Eugenia Martín-Vázquez
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Álvaro Jesús Narbona-Pérez
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Livia López-Noriega
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Guillermo Martínez-Corrales
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Raúl López-Fernández-Sobrino
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Alejandro Castillo-Peña
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Lina M Carmona-Marin
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Oscar Yanes
- Universitat Rovira i Virgili, Department of electronic Engineering & IISPV, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Vinaixa
- Universitat Rovira i Virgili, Department of electronic Engineering & IISPV, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel López-López
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocio, c/Manuel Siurot s/n, 41013, Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocio, Sevilla, 41013, Spain
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocío, Sevilla, 41013, Spain
| | - José Carlos Reyes
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Joaquín Dopazo
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocio, c/Manuel Siurot s/n, 41013, Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocio, Sevilla, 41013, Spain
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocío, Sevilla, 41013, Spain
- FPS/ELIXIR-es, Hospital Virgen del Rocío, Sevilla, 42013, Spain
| | - Franz Martín
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
- CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Benoit R Gauthier
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
- CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Tracked.bio, Copenhagen, Denmark
| | - Vivian Capilla-González
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain
| | - Alejandro Martín-Montalvo
- Andalusian Molecular Biology and Regenerative Medicine Centre-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, 41092, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
48
|
Abstract
Few metabolites can claim a more central and versatile role in cell metabolism than acetyl coenzyme A (acetyl-CoA). Acetyl-CoA is produced during nutrient catabolism to fuel the tricarboxylic acid cycle and is the essential building block for fatty acid and isoprenoid biosynthesis. It also functions as a signalling metabolite as the substrate for lysine acetylation reactions, enabling the modulation of protein functions in response to acetyl-CoA availability. Recent years have seen exciting advances in our understanding of acetyl-CoA metabolism in normal physiology and in cancer, buoyed by new mouse models, in vivo stable-isotope tracing approaches and improved methods for measuring acetyl-CoA, including in specific subcellular compartments. Efforts to target acetyl-CoA metabolic enzymes are also advancing, with one therapeutic agent targeting acetyl-CoA synthesis receiving approval from the US Food and Drug Administration. In this Review, we give an overview of the regulation and cancer relevance of major metabolic pathways in which acetyl-CoA participates. We further discuss recent advances in understanding acetyl-CoA metabolism in normal tissues and tumours and the potential for targeting these pathways therapeutically. We conclude with a commentary on emerging nodes of acetyl-CoA metabolism that may impact cancer biology.
Collapse
Affiliation(s)
- David A Guertin
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA.
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
49
|
Use of bempedoic acid for LDL cholesterol lowering and cardiovascular risk reduction: a consensus document from the Italian study group on atherosclerosis, thrombosis and vascular biology. Vascul Pharmacol 2023; 148:107137. [PMID: 36464086 DOI: 10.1016/j.vph.2022.107137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
The clinical benefit of LDL cholesterol (LDL-C) lowering for cardiovascular disease prevention is well documented. This paper from the Italian Study Group on Atherosclerosis, Thrombosis and Vascular Biology summarizes current recommendations for treatment of hypercholesterolemia, barriers to lipid-lowering therapy implementation and tips to overcome them, as well as available evidence on the efficacy and safety of bempedoic acid. We also report an updated therapeutic algorithm for pharmacological LDL-C lowering in view of the introduction of bempedoic acid in clinical practice.
Collapse
|
50
|
Xie Z, Zhang M, Song Q, Cheng L, Zhang X, Song G, Sun X, Gu M, Zhou C, Zhang Y, Zhu K, Yin J, Chen X, Li J, Nan F. Development of the novel ACLY inhibitor 326E as a promising treatment for hypercholesterolemia. Acta Pharm Sin B 2023; 13:739-753. [PMID: 36873173 PMCID: PMC9979192 DOI: 10.1016/j.apsb.2022.06.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/24/2022] [Accepted: 05/30/2022] [Indexed: 11/27/2022] Open
Abstract
Hepatic cholesterol accumulation is an important contributor to hypercholesterolemia, which results in atherosclerosis and cardiovascular disease (CVD). ATP-citrate lyase (ACLY) is a key lipogenic enzyme that converts cytosolic citrate derived from tricarboxylic acid cycle (TCA cycle) to acetyl-CoA in the cytoplasm. Therefore, ACLY represents a link between mitochondria oxidative phosphorylation and cytosolic de novo lipogenesis. In this study, we developed the small molecule 326E with an enedioic acid structural moiety as a novel ACLY inhibitor, and its CoA-conjugated form 326E-CoA inhibited ACLY activity with an IC50 = 5.31 ± 1.2 μmol/L in vitro. 326E treatment reduced de novo lipogenesis, and increased cholesterol efflux in vitro and in vivo. 326E was rapidly absorbed after oral administration, exhibited a higher blood exposure than that of the approved ACLY inhibitor bempedoic acid (BA) used for hypercholesterolemia. Chronic 326E treatment in hamsters and rhesus monkeys resulted in remarkable improvement of hyperlipidemia. Once daily oral administration of 326E for 24 weeks prevented the occurrence of atherosclerosis in ApoE-/- mice to a greater extent than that of BA treatment. Taken together, our data suggest that inhibition of ACLY by 326E represents a promising strategy for the treatment of hypercholesterolemia.
Collapse
Affiliation(s)
- Zhifu Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qian Song
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Long Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinwen Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Gaolei Song
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyu Sun
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Min Gu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chendong Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yangming Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,Burgeon Therapeutics Co., Ltd., Shanghai 201203, China
| | - Kexin Zhu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianpeng Yin
- Drug Discovery Shandong Laboratory, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Xiaoyan Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingya Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fajun Nan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Drug Discovery Shandong Laboratory, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| |
Collapse
|