1
|
Khirallah J, Bloomer H, Wich D, Huang C, Workman JN, Li Y, Newby GA, Liu DR, Xu Q. In vivo base editing of Angptl3 via lipid nanoparticles to treat cardiovascular disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102486. [PMID: 40129501 PMCID: PMC11932681 DOI: 10.1016/j.omtn.2025.102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 02/12/2025] [Indexed: 03/26/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally and is exacerbated by elevated blood levels of low-density lipoprotein cholesterol (LDL-C) and triglycerides (TGs). Existing approaches for decreasing blood lipid levels rely on daily medications, leading to poor patient adherence. Gene therapy represents a promising "one and done" strategy to durably reduce blood lipid levels. ANGPTL3 has emerged as a potential target for gene therapy, as naturally occurring loss-of-function variants are cardioprotective. Here, we use lipid nanoparticles to package and deliver CRISPR adenine base editors (ABEs), which enable gene knockout without requiring potentially harmful DNA double-strand breaks. We package ABE mRNA and a synthetic guide RNA targeted to disrupt an important splice site in Angptl3, which we administered to mice intravenously. We achieved over 60% base editing in the liver and durable reductions in serum ANGPTL3, LDL-C, and TGs for at least 100 days. Notably, blood lipid levels remained low when mice were challenged with a high-fat high-cholesterol diet up to 191 days after therapy. These results provide a foundation for a potential one-and-done treatment for CVD.
Collapse
Affiliation(s)
- Jennifer Khirallah
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Hanan Bloomer
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- School of Medicine and Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Douglas Wich
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Changfeng Huang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - J. Noah Workman
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Gregory A. Newby
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
| | - David R. Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- School of Medicine and Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
2
|
Tremblay F, Xiong Q, Shah SS, Ko CW, Kelly K, Morrison MS, Giancarlo C, Ramirez RN, Hildebrand EM, Voytek SB, El Sebae GK, Wright SH, Lofgren L, Clarkson S, Waters C, Linder SJ, Liu S, Eom T, Parikh S, Weber Y, Martinez S, Malyala P, Abubucker S, Friedland AE, Maeder ML, Lombardo A, Myer VE, Jaffe AB. A potent epigenetic editor targeting human PCSK9 for durable reduction of low-density lipoprotein cholesterol levels. Nat Med 2025; 31:1329-1338. [PMID: 39930141 PMCID: PMC12003160 DOI: 10.1038/s41591-025-03508-x] [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: 09/13/2024] [Accepted: 01/13/2025] [Indexed: 02/27/2025]
Abstract
Epigenetic editing holds the promise of durable therapeutic effects by silencing disease-causing genes without changing the underlying DNA sequence. In this study, we designed an epigenetic editor to target human PCSK9 and thereby induce DNA methylation at this locus. A single administration of lipid nanoparticles encapsulating mRNA encoding this epigenetic editor was sufficient to drive near-complete silencing of human PCSK9 in transgenic mice. Silencing was durable for at least 1 year and was fully maintained after partial hepatectomy-induced liver regeneration. In addition, we showed reversibility of epigenetic editing in mice with previously silenced PCSK9 upon treatment with a targeted epigenetic activator designed to demethylate the PCSK9 locus. Notably, in cynomolgus monkeys, a single administration of the epigenetic editor potently and durably decreased circulating PCSK9 protein levels by approximately 90% with concomitant reduction in low-density lipoprotein cholesterol levels by approximately 70%. These findings demonstrate the therapeutic potential of durable and reversible epigenetic editing in vivo and support the development of epigenetic editor-based treatment for hypercholesterolemia.
Collapse
Affiliation(s)
| | - Qiang Xiong
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Shrijal S Shah
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Chih-Wei Ko
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | | | | | | | | | - Sarah B Voytek
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | - Shane H Wright
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | | | | | | | - Songlei Liu
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Taesun Eom
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Shefal Parikh
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | - Yuki Weber
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | - Padma Malyala
- Chroma Medicine, Boston, MA, USA
- nChromaBio, Boston, MA, USA
| | | | | | | | - Angelo Lombardo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | | | - Aron B Jaffe
- Chroma Medicine, Boston, MA, USA
- Curie.Bio, Cambridge, MA, USA
| |
Collapse
|
3
|
Inia JA, van Nieuwkoop-van Straalen A, Jukema JW, Rolin B, Staarup EM, Mogensen CK, Princen HMG, van den Hoek AM. Efficacy of a novel PCSK9 inhibitory peptide alone and with evinacumab in a mouse model of atherosclerosis. J Lipid Res 2025; 66:100753. [PMID: 39909173 PMCID: PMC11927713 DOI: 10.1016/j.jlr.2025.100753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/23/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025] Open
Abstract
Atherosclerosis is the major cause of cardiovascular disease. This study evaluated the effect of lipid lowering using a novel peptide inhibiting proprotein convertase subtilisin/kexin type 9 (PCSK9) and a monoclonal antibody against angiopoietin-like 3 (evinacumab), either alone or in combination in APOE∗3-Leiden.CETP mice fed a Western diet. Effects on body weight, plasma lipids, atherosclerotic lesion size, severity, composition, and morphology were assessed. Treatment with PCSK9 inhibitory peptide significantly decreased both cholesterol and triglycerides (-69% and -68%, respectively). Similar reductions were seen in evinacumab-treated mice (-44% and -55%, respectively). The combination of evinacumab and PCSK9 inhibitory peptide lowered these levels to a larger extent than evinacumab alone (cholesterol: -74%; triglycerides: -81%). Reductions occurred in non-HDL-C without changes in HDL-C. Atherosclerotic lesion size was significantly reduced in all treatment groups compared to vehicle controls (evinacumab: -72%; PCSK9 inhibitory peptide: -97%; combination: -98%). Similarly, all interventions improved atherosclerotic lesion severity, with more undiseased segments and fewer severe lesions. Evaluation of the composition of severe atherosclerotic plaques revealed significant improvement in lesion stability in mice treated with both evinacumab and PCSK9 inhibitory peptide, attributable to decreased macrophage content and increased collagen content. Additionally, evaluation of lipid concentrations in cynomolgus monkeys revealed the beneficial effects of the PCSK9 inhibitory peptide on total cholesterol and LDL-C levels. Treatment with a novel PCSK9 inhibitory peptide alone or with evinacumab shows great potential to reduce and stabilize atherosclerotic lesions.
Collapse
Affiliation(s)
- José A Inia
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands; Department of Cardiology, Leiden University Medical Centre, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, the Netherlands.
| | - Anita van Nieuwkoop-van Straalen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Centre, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - Bidda Rolin
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | | | | | - Hans M G Princen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Anita M van den Hoek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| |
Collapse
|
4
|
Inia JA, Attema J, de Ruiter C, Menke AL, Caspers MPM, Verschuren L, Wilson M, Arlantico A, Brightbill HD, Jukema JW, van den Hoek AM, Princen HMG, Chen MZ, Morrison MC. Therapeutic effects of FGF21 mimetic bFKB1 on MASH and atherosclerosis in Ldlr-/-.Leiden mice. FASEB J 2024; 38:e70087. [PMID: 39463193 PMCID: PMC11580715 DOI: 10.1096/fj.202401397r] [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: 06/19/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 10/29/2024]
Abstract
Fibroblast growth factor 21 (FGF21) is a promising target for treatment of obesity-associated diseases including metabolic dysfunction-associated steatohepatitis (MASH) and atherosclerosis. We evaluated the effects of the bispecific anti-FGF21-β klotho (KLB) agonist antibody bFKB1 in a preclinical model of MASH and atherosclerosis. Low-density lipoprotein receptor knockout (Ldlr-/-).Leiden mice received a high-fat diet for 20 weeks, followed by treatment with an isotype control antibody or bFKB1 for 12 weeks. Effects on plasma risk markers and (histo)pathology of liver, adipose tissue, and heart were evaluated alongside hepatic transcriptomics analysis. bFKB1 lowered body weight (-21%) and adipose tissue mass (-22%) without reducing food intake. The treatment also improved plasma insulin (-80%), cholesterol (-48%), triglycerides (-76%), alanine transaminase (ALT: -79%), and liver weight (-43%). Hepatic steatosis and inflammation were strongly reduced (macrovesicular steatosis -34%; microvesicular steatosis -100%; inflammation -74%) and while the total amount of fibrosis was not affected, bFKB1 did decrease new collagen formation (-49%). Correspondingly, hepatic transcriptomics and pathway analysis revealed the mechanistic background underlying these histological improvements, demonstrating broad inactivation of inflammatory and profibrotic transcriptional programs by bFKB1. In epididymal white adipose tissue, bFKB1 reduced adipocyte size (-16%) and inflammation (-52%) and induced browning, signified by increased uncoupling protein-1 (UCP1) protein expression (8.5-fold increase). In the vasculature, bFKB1 had anti-atherogenic effects, lowering total atherosclerotic lesion area (-38%). bFKB1 has strong beneficial metabolic effects associated with a reduction in hepatic steatosis, inflammation, and atherosclerosis. Analysis of new collagen formation and profibrotic transcriptional programs indicate that bFKB1 treatment may have antifibrotic potential in a longer treatment duration as well.
Collapse
Affiliation(s)
- José A. Inia
- Department of Metabolic Health ResearchThe Netherlands Organisation for Applied Scientific Research (TNO)LeidenThe Netherlands
- Department of CardiologyLeiden University Medical Centre (LUMC)LeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
| | - Joline Attema
- Department of Metabolic Health ResearchThe Netherlands Organisation for Applied Scientific Research (TNO)LeidenThe Netherlands
| | - Christa de Ruiter
- Department of Metabolic Health ResearchThe Netherlands Organisation for Applied Scientific Research (TNO)LeidenThe Netherlands
| | - Aswin L. Menke
- Department of Metabolic Health ResearchThe Netherlands Organisation for Applied Scientific Research (TNO)LeidenThe Netherlands
| | | | - Lars Verschuren
- Department of Microbiology and Systems BiologyTNOLeidenThe Netherlands
| | | | | | | | - J. Wouter Jukema
- Department of CardiologyLeiden University Medical Centre (LUMC)LeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLUMCLeidenThe Netherlands
- Netherlands Heart InstituteUtrechtThe Netherlands
| | - Anita M. van den Hoek
- Department of Metabolic Health ResearchThe Netherlands Organisation for Applied Scientific Research (TNO)LeidenThe Netherlands
| | - Hans M. G. Princen
- Department of Metabolic Health ResearchThe Netherlands Organisation for Applied Scientific Research (TNO)LeidenThe Netherlands
| | - Mark Z. Chen
- Translational ImmunologyGenentech Inc.South San FranciscoCaliforniaUSA
| | - Martine C. Morrison
- Department of Metabolic Health ResearchThe Netherlands Organisation for Applied Scientific Research (TNO)LeidenThe Netherlands
| |
Collapse
|
5
|
Chen SJ, Chien HC, Tsai SH, Jheng YS, Chen Y, Hsieh PS, Tsui PF, Chien S, Tsai MC. Melatonin Ameliorates Atherosclerotic Plaque Vulnerability by Regulating PPARδ-Associated Smooth Muscle Cell Phenotypic Switching. J Pineal Res 2024; 76:e12988. [PMID: 38982751 DOI: 10.1111/jpi.12988] [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: 03/19/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/11/2024]
Abstract
Vulnerable atherosclerotic plaque rupture, the leading cause of fatal atherothrombotic events, is associated with an increased risk of mortality worldwide. Peroxisome proliferator-activated receptor delta (PPARδ) has been shown to modulate vascular smooth muscle cell (SMC) phenotypic switching, and, hence, atherosclerotic plaque stability. Melatonin reportedly plays a beneficial role in cardiovascular diseases; however, the mechanisms underlying improvements in atherosclerotic plaque vulnerability remain unknown. In this study, we assessed the role of melatonin in regulating SMC phenotypic switching and its consequential contribution to the amelioration of atherosclerotic plaque vulnerability and explored the mechanisms underlying this process. We analyzed features of atherosclerotic plaque vulnerability and markers of SMC phenotypic transition in high-cholesterol diet (HCD)-fed apolipoprotein E knockout (ApoE-/-) mice and human aortic SMCs (HASMCs). Melatonin reduced atherosclerotic plaque size and necrotic core area while enhancing collagen content, fibrous cap thickness, and smooth muscle alpha-actin positive cell coverage on the plaque cap, which are all known phenotypic characteristics of vulnerable plaques. In atherosclerotic lesions, melatonin significantly decreased the synthetic SMC phenotype and KLF4 expression and increased the expression of PPARδ, but not PPARα and PPARγ, in HCD-fed ApoE-/- mice. These results were subsequently confirmed in the melatonin-treated HASMCs. Further analysis using PPARδ silencing and immunoprecipitation assays revealed that PPARδ plays a role in the melatonin-induced SMC phenotype switching from synthetic to contractile. Collectively, we provided the first evidence that melatonin mediates its protective effect against plaque destabilization by enhancing PPARδ-mediated SMC phenotypic switching, thereby indicating the potential of melatonin in treating atherosclerosis.
Collapse
MESH Headings
- Animals
- Melatonin/pharmacology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Kruppel-Like Factor 4/metabolism
- Humans
- PPAR delta/metabolism
- PPAR delta/genetics
- Mice, Knockout
- Male
- Mice, Knockout, ApoE
- Phenotype
- Apolipoproteins E/genetics
- Apolipoproteins E/metabolism
- Apolipoproteins E/deficiency
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Sy-Jou Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Che Chien
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hung Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Sin Jheng
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Yi Chen
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Po-Shiuan Hsieh
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan
| | - Pi-Fen Tsui
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Shu Chien
- Department of Bioengineering and Medicine, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, USA
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
6
|
Inia JA, de Jong JCBC, Keijzer N, Menke AL, Princen HMG, Jukema JW, van den Hoek AM. Effects of repeated weight cycling on non-alcoholic steatohepatitis in diet-induced obese mice. FASEB J 2024; 38:e23579. [PMID: 38568838 DOI: 10.1096/fj.202400167r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/08/2024] [Accepted: 03/14/2024] [Indexed: 04/05/2024]
Abstract
Lifestyle interventions remain the treatment of choice for patients with obesity and metabolic complications, yet are difficult to maintain and often lead to cycles of weight loss and regain (weight cycling). Literature on weight cycling remains controversial and we therefore investigated the association between weight cycling and metabolic complications using preexistent obese mice. Ldlr-/-.Leiden mice received a high-fat diet (HFD) for 20 weeks to induce obesity. Subsequently, weight-cycled mice were switched between the healthy chow diet and HFD for four 2-week periods and compared to mice that received HFD for the total study period. Repeated weight cycling tended to decrease body weight and significantly reduced fat mass, whereas adipose tissue inflammation was similar relative to HFD controls. Weight cycling did not significantly affect blood glucose or plasma insulin levels yet significantly reduced plasma free fatty acid and alanine transaminase/aspartate transaminase levels. Hepatic macrovesicular steatosis was similar and microvesicular steatosis tended to be increased upon weight cycling. Weight cycling resulted in a robust decrease in hepatic inflammation compared to HFD controls while hepatic fibrosis and atherosclerosis development were not affected. These results argue against the postulate that repeated weight cycling leads to unfavorable metabolic effects, when compared to a continuous unhealthy lifestyle, and in fact revealed beneficial effects on hepatic inflammation, an important hallmark of non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- José A Inia
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| | - Jelle C B C de Jong
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Nanda Keijzer
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Aswin L Menke
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Hans M G Princen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Anita M van den Hoek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
7
|
Chen Z, Shao W, Li Y, Zhang X, Geng Y, Ma X, Tao B, Ma Y, Yi C, Zhang B, Zhang R, Lin J, Chen J. Inhibition of PCSK9 prevents and alleviates cholesterol gallstones through PPARα-mediated CYP7A1 activation. Metabolism 2024; 152:155774. [PMID: 38191052 DOI: 10.1016/j.metabol.2023.155774] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/28/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND & AIMS Dysregulated cholesterol metabolism is the major factor responsible for cholesterol gallstones (CGS). Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a critical role in cholesterol homeostasis and its inhibitors secure approval for treating various cholesterol metabolic disorders such as hypercholesterolemia and cardiovascular diseases, but its role in CGS remains unclear. Our study aims to clarify mechanisms by which PCSK9 promotes CGS formation and explore the application of the PCSK9 inhibitor, alirocumab, in preventing and treating CGS. APPROACH & RESULTS The expressions of PCSK9 were notably increased in CGS patients' serum, bile, and liver tissues compared to those without gallstones. Moreover, among CGS patients, hepatic PCSK9 was positively correlated with hepatic cholesterol and negatively correlated with hepatic bile acids (BAs), suggesting PCSK9 was involved in disrupted hepatic cholesterol metabolism related to CGS. Mechanistically, in vitro experiments demonstrated that inhibition of PCSK9 enhanced nuclear expression of PPARα by diminishing its lysosomal degradation and subsequently activated CYP7A1 transcription. Finally, inhibition of PCSK9 prevented CGS formation and dissolved the existing stones in CGS mice by elevating the conversion of cholesterol into BAs through PPARα-mediated CYP7A1 activation. Additionally, serum PCSK9 level may function as a prognostic signature to evaluate the therapeutic efficacy of PCSK9 inhibitors. CONCLUSIONS Inhibition of PCSK9 exerts preventive and therapeutic effects on CGS by activating PPARα-mediated CYP7A1 expression and facilitating the conversion of cholesterol into BAs, which highlights the potential of PCSK9 inhibition as a promising candidate for preventing and treating CGS in clinical applications. IMPACT AND IMPLICATIONS PCSK9 plays a pivotal role in cholesterol metabolism and its inhibitors are approved for clinical use in cardiovascular diseases. Our study observes inhibition of PCSK9 prevents and dissolves CGS by activating PPARα-mediated CYP7A1 expression and facilitating the conversion of cholesterol into BAs. Mechanistically, PCSK9 inhibition enhanced the nuclear expression of PPARα by diminishing its lysosomal degradation and subsequently activated CYP7A1 transcription. Our study sheds light on the new function and mechanism of PCSK9 in CGS, providing a novel preventive and therapeutic target with potential clinical applications.
Collapse
Affiliation(s)
- Zhenmei Chen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China; Shanghai Institute of Infectious Disease and Biosecurity, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Weiqing Shao
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Yitong Li
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Xiandi Zhang
- Department of Ultrasound, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Yan Geng
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Xiaochen Ma
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Baorui Tao
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Yue Ma
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Chenhe Yi
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Bo Zhang
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Rui Zhang
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Jing Lin
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China.
| | - Jinhong Chen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China.
| |
Collapse
|
8
|
Ye W, Fan J, Wu W, Chen Z, Huang Q, Qian L. Effects of fecal microbiota transplantation on metabolic health of DBA mice. Front Microbiol 2024; 15:1352555. [PMID: 38444807 PMCID: PMC10912182 DOI: 10.3389/fmicb.2024.1352555] [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: 12/08/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Introduction Numerous studies have demonstrated that C57BL/6 mice exhibit superior growth rates and overall growth performance compared to DBA mice. To investigate whether this discrepancy in growth performance is linked to the composition of gut microorganisms, we conducted fecal microbiome transplantation (FMT) experiments. Methods Specifically, we transplanted fecal fluids from adult C57BL/6 mice, high-fat C57BL/6 mice, and Wistar rats into weaned DBA mice (0.2mL/d), and subsequently analyzed their gut contents and gene expression through 16S rRNA sequencing and transcriptome sequencing. During the test period, C57BL/6 mice and Wistar rats were provided with a normal diet, and high-fat C57BL/6 mice were provided with a high-fat diet. Results The results of our study revealed that mice receiving FMT from all three donor groups exhibited significantly higher daily weight gain and serum triglyceride (TG) levels compared to mice of CK group. 16S rRNA sequensing unveiled substantial differences in the abundance and function of the gut microbiota between the FMT groups and the CK group. Transcriptome analysis revealed a total of 988 differential genes, consisting of 759 up-regulated genes and 187 down-regulated genes, between the three experimental groups and the CK group. Functional Gene Ontology (GO) annotation suggested that these genes were primarily linked to lipid metabolism, coagulation, and immunity. Pearson correlation analysis was performed on the differential genes and clusters, and it revealed significant correlations, mainly related to processes such as fatty acid metabolism, fat digestion and absorption, and cholesterol metabolism. Discussion In summary, FMT from dominant strains improved the growth performance of DBA mice, including body weight gain, institutional growth, and immune performance. This change may be due to the increase of probiotic content in the intestinal tract by FMT and subsequent alteration of intestinal gene expression. However, the effects of cross-species fecal transplantation on the intestinal flora and gene expression of recipient mice were not significant.
Collapse
Affiliation(s)
- Wenxin Ye
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jinghui Fan
- Hangzhou Academy of Agricultural Sciences, Hangzhou, China
| | - Wenzi Wu
- Hainan Institute of Zhejiang University, Sanya, China
| | - Zhuo Chen
- Hainan Institute of Zhejiang University, Sanya, China
| | - Qixin Huang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Lichun Qian
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Parsamanesh N, Poudineh M, Siami H, Butler AE, Almahmeed W, Sahebkar A. RNA interference-based therapies for atherosclerosis: Recent advances and future prospects. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 204:1-43. [PMID: 38458734 DOI: 10.1016/bs.pmbts.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Atherosclerosis represents a pathological state that affects the arterial system of the organism. This chronic, progressive condition is typified by the accumulation of atheroma within arterial walls. Modulation of RNA molecules through RNA-based therapies has expanded the range of therapeutic options available for neurodegenerative diseases, infectious diseases, cancer, and, more recently, cardiovascular disease (CVD). Presently, microRNAs and small interfering RNAs (siRNAs) are the most widely employed therapeutic strategies for targeting RNA molecules, and for regulating gene expression and protein production. Nevertheless, for these agents to be developed into effective medications, various obstacles must be overcome, including inadequate binding affinity, instability, challenges of delivering to the tissues, immunogenicity, and off-target toxicity. In this comprehensive review, we discuss in detail the current state of RNA interference (RNAi)-based therapies.
Collapse
Affiliation(s)
- Negin Parsamanesh
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Haleh Siami
- School of Medicine, Islamic Azad University of Medical Science, Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, Adliya, Bahrain
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Chen L, Wang X, Liu C, Deng P, Pan L, Yang L, Cheng J, Zhang X, Reiter RJ, Yu Z, Pi H, Zhou Z, Hu H. Melatonin ameliorates atherosclerosis by suppressing S100a9-mediated vascular inflammation. Eur J Pharmacol 2023; 957:175965. [PMID: 37625682 DOI: 10.1016/j.ejphar.2023.175965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023]
Abstract
Atherosclerosis (AS)-associated cardiovascular diseases are predominant causes of morbidity and mortality worldwide. Melatonin, a circadian hormone with anti-inflammatory activity, may be a novel therapeutic intervention for AS. However, the exact mechanism is unclear. This research intended to investigate the mechanism of melatonin in treating AS. Melatonin (20 mg/kg/d) was intraperitoneally administered in a high-fat diet (HFD)-induced AS model using apolipoprotein E-deficient (ApoE-/-) mice for 12 weeks. Immunohistochemical and immunofluorescence analyses, data-independent acquisition (DIA)-based protein profiling, ingenuity pathway analysis (IPA), and western blotting were employed to investigate the therapeutic effects of melatonin in treating HFD-induced AS. An adeno-associated virus (AAV) vector was further used to confirm the antiatherosclerotic mechanism of melatonin. Melatonin treatment markedly attenuated atherosclerotic lesions, induced stable phenotypic sclerotic plaques, inhibited macrophage infiltration, and suppressed the production of proinflammatory cytokines in ApoE-/- mice with HFD-induced AS. Notably, DIA-based quantitative proteomics together with IPA identified S100a9 as a pivotal mediator in the protective effects of melatonin. Moreover, melatonin significantly suppressed HFD-induced S100a9 expression at both the mRNA and protein levels. The overexpression of S100a9 significantly activated the NF-κB signaling pathway and markedly abolished the antagonistic effect of melatonin on HFD-induced vascular inflammation during atherogenesis. Melatonin exerts a significant antiatherogenic effect by inhibiting S100a9/NF-κB signaling pathway-mediated vascular inflammation. Our findings reveal a novel antiatherosclerotic mechanism of melatonin and underlie its potential clinical use in modulating AS with good availability and affordability.
Collapse
Affiliation(s)
- Liyuan Chen
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Xue Wang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, 400038, China
| | - Chang Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, 400038, China
| | - Lina Pan
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Lingling Yang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, 400038, China
| | - Juan Cheng
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Xutao Zhang
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, TX, 78229, USA
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, 400038, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, 400038, China.
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400030, China.
| | - Houyuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
11
|
Morinaga J, Kashiwabara K, Torigoe D, Okadome Y, Aizawa K, Uemura K, Kurashima A, Matsunaga E, Fukami H, Horiguchi H, Sato M, Sugizaki T, Miyata K, Kadomatsu T, Mukoyama M, Miyauchi K, Hokimoto S, Fukumoto Y, Hiro T, Hibi K, Nakagawa Y, Sakuma I, Ozaki Y, Iwata H, Iimuro S, Daida H, Shimokawa H, Kimura T, Matsuzaki M, Saito Y, Matsuyama Y, Nagai R, Oike Y. Plasma ANGPTL8 Levels and Risk for Secondary Cardiovascular Events in Japanese Patients With Stable Coronary Artery Disease Receiving Statin Therapy. Arterioscler Thromb Vasc Biol 2023; 43:1549-1559. [PMID: 37259862 DOI: 10.1161/atvbaha.122.318880] [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: 12/12/2022] [Accepted: 05/19/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND The ability to predict secondary cardiovascular events could improve health of patients undergoing statin treatment. Circulating ANGPTL8 (angiopoietin-like protein 8) levels, which positively correlate with proatherosclerotic lipid profiles, activate the pivotal proatherosclerotic factor ANGPTL3. Here, we assessed potential association between circulating ANGPTL8 levels and risk of secondary cardiovascular events in statin-treated patients. METHODS We conducted a biomarker study with a case-cohort design, using samples from a 2018 randomized control trial known as randomized evaluation of high-dose (4 mg/day) or low-dose (1 mg/day) lipid-lowering therapy with pitavastatin in coronary artery disease (REAL-CAD [Randomized Evaluation of Aggressive or Moderate Lipid-Lowering Therapy With Pitavastatin in Coronary Artery Disease])." From that study's full analysis set (n=12 413), we selected 2250 patients with stable coronary artery disease (582 with the primary outcome, 1745 randomly chosen, and 77 overlapping subjects). A composite end point including cardiovascular-related death, nonfatal myocardial infarction, nonfatal ischemic stroke, or unstable angina requiring emergent admission was set as a primary end point. Circulating ANGPTL8 levels were measured at baseline and 6 months after randomization. RESULTS Over a 6-month period, ANGPTL8 level changes significantly decreased in the high-dose pitavastatin group, which showed 19% risk reduction of secondary cardiovascular events compared with the low-dose group in the REAL-CAD [Randomized Evaluation of Aggressive or Moderate Lipid-Lowering Therapy With Pitavastatin in Coronary Artery Disease] study. In the highest quartiles, relative increases in ANGPTL8 levels were significantly associated with increased risk for secondary cardiovascular events, after adjustment for several cardiovascular disease risk factors and pitavastatin treatment (hazard ratio in Q4, 1.67 [95% CI, 1.17-2.39). Subgroup analyses showed relatively strong relationships between relative ANGPTL8 increases and secondary cardiovascular events in the high-dose pitavastatin group (hazard ratio in Q4, 2.07 [95% CI, 1.21-3.55]) and in the low ANGPTL8 group at baseline (166 CONCLUSIONS Monitoring ANGPTL8 levels over time might be useful to assess residual risk of cardiovascular secondary events in patients with cardiovascular disease undergoing statin therapy.
Collapse
Affiliation(s)
- Jun Morinaga
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
- Department of Nephrology (J.M., A.K., E.M., H.F., M.M.), Graduate School of Medical Sciences, Kumamoto University, Japan
- Department of Clinical Investigation, Kumamoto University Hospital, Japan (J.M.)
| | - Kosuke Kashiwabara
- Data Science Office, Clinical Research Promotion Center, The University of Tokyo Hospital, Japan (K.K.)
| | - Daisuke Torigoe
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Yusuke Okadome
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Kenichi Aizawa
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Tochigi, Japan (K.A.)
| | - Kohei Uemura
- Department of Biostatistics and Bioinformatics, Interfaculty Initiative in Information Studies (K.U.), The University of Tokyo, Japan
| | - Ai Kurashima
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
- Department of Nephrology (J.M., A.K., E.M., H.F., M.M.), Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Eiji Matsunaga
- Department of Nephrology (J.M., A.K., E.M., H.F., M.M.), Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Hirotaka Fukami
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
- Department of Nephrology (J.M., A.K., E.M., H.F., M.M.), Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Haruki Horiguchi
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Michio Sato
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Taichi Sugizaki
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Keishi Miyata
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Masashi Mukoyama
- Department of Nephrology (J.M., A.K., E.M., H.F., M.M.), Graduate School of Medical Sciences, Kumamoto University, Japan
| | - Katsumi Miyauchi
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan (K.M., H.I., H.D.)
| | | | - Yoshihiro Fukumoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, Japan (Y.F.)
| | - Takafumi Hiro
- Division of Cardiology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan (T.H.)
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, Japan (K.H.)
| | - Yoshihisa Nakagawa
- Department of Cardiovascular Medicine, Shiga University of Medical Science, Otsu, Japan (Y.N.)
| | | | - Yukio Ozaki
- Department of Cardiology, Fujita Health University School of Medicine, Toyoake, Japan (Y.O.)
| | - Hiroshi Iwata
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan (K.M., H.I., H.D.)
| | - Satoshi Iimuro
- Innovation and Research Support Center, International University of Health and Welfare, Tokyo, Japan (S.I.)
| | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan (K.M., H.I., H.D.)
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (H.S.)
- International University of Health and Welfare, Narita, Japan (H.S.)
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Japan (T.K.)
| | | | | | - Yutaka Matsuyama
- Department of Biostatistics, School of Public Health, Graduate School of Medicine (Y.M.), The University of Tokyo, Japan
| | - Ryozo Nagai
- Jichi Medical University, Shimotsuke, Japan (R.N.)
| | - Yuichi Oike
- Department of Molecular Genetics (J.M., D.T., Y. Okadome., A.K., E.M., H.F., H.H., M.S., T.S., K.M., T.K. Y. Oike),, Graduate School of Medical Sciences, Kumamoto University, Japan
| |
Collapse
|
12
|
In Het Panhuis W, Schönke M, Modder M, Tom HE, Lalai RA, Pronk ACM, Streefland TCM, van Kerkhof LWM, Dollé MET, Depuydt MAC, Bot I, Vos WG, Bosmans LA, van Os BW, Lutgens E, Rensen PCN, Kooijman S. Time-restricted feeding attenuates hypercholesterolaemia and atherosclerosis development during circadian disturbance in APOE∗3-Leiden.CETP mice. EBioMedicine 2023; 93:104680. [PMID: 37356205 DOI: 10.1016/j.ebiom.2023.104680] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Circadian disturbance (CD) is the consequence of a mismatch between endogenous circadian rhythms, behaviour, and/or environmental cycles, and frequently occurs during shift work. Shift work has been associated with elevated risk for atherosclerotic cardiovascular disease (asCVD) in humans, but evidence for the effectiveness of prevention strategies is lacking. METHODS Here, we applied time-restricted feeding (TRF) as a strategy to counteract atherosclerosis development during CD in female APOE∗3-Leiden.CETP mice, a well-established model for humanized lipoprotein metabolism. Control groups were subjected to a fixed 12:12 h light-dark cycle, while CD groups were subjected to 6-h phase advancement every 3 days. Groups had either ad libitum (AL) access to food or were subjected to TRF with restricted food access to the dark phase. FINDINGS TRF did not prevent the increase in the relative abundance of circulating inflammatory monocytes and elevation of (postprandial) plasma triglycerides during CD. Nonetheless, TRF reduced atherosclerotic lesion size and prevented an elevation in macrophage content of atherosclerotic lesions during CD, while it increased the relative abundance of anti-inflammatory monocytes, prevented activation of T cells, and lowered plasma total cholesterol levels and markers of hepatic cholesterol synthesis. These effects were independent of total food intake. INTERPRETATION We propose that time restricted eating could be a promising strategy for the primary prevention of asCVD risk in shift workers, which warrants future study in humans. FUNDING This work was funded by the Novo Nordisk Foundation, the Netherlands Ministry of Social Affairs and Employment, Amsterdam Cardiovascular Sciences, and the Dutch Heart Foundation.
Collapse
Affiliation(s)
- Wietse In Het Panhuis
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Melanie Modder
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Hannah E Tom
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Reshma A Lalai
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Amanda C M Pronk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Trea C M Streefland
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Linda W M van Kerkhof
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Martijn E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Marie A C Depuydt
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, the Netherlands
| | - Ilze Bot
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, the Netherlands
| | - Winnie G Vos
- Department of Medical Biochemistry, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Immunity and Infection, Amsterdam, the Netherlands
| | - Laura A Bosmans
- Department of Medical Biochemistry, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Immunity and Infection, Amsterdam, the Netherlands
| | - Bram W van Os
- Department of Medical Biochemistry, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Immunity and Infection, Amsterdam, the Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Immunity and Infection, Amsterdam, the Netherlands; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
13
|
Inia JA, Stokman G, Morrison MC, Worms N, Verschuren L, Caspers MPM, Menke AL, Petitjean L, Chen L, Petitjean M, Jukema JW, Princen HMG, van den Hoek AM. Semaglutide Has Beneficial Effects on Non-Alcoholic Steatohepatitis in Ldlr-/-.Leiden Mice. Int J Mol Sci 2023; 24:ijms24108494. [PMID: 37239841 DOI: 10.3390/ijms24108494] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
Semaglutide, a glucagon-like peptide-1 receptor agonist, is an antidiabetic medication that has recently been approved for the treatment of obesity as well. Semaglutide is postulated to be a promising candidate for the treatment of non-alcoholic steatohepatitis (NASH). Here, Ldlr-/-.Leiden mice received a fast-food diet (FFD) for 25 weeks, followed by another 12 weeks on FFD with daily subcutaneous injections of semaglutide or vehicle (control). Plasma parameters were evaluated, livers and hearts were examined, and hepatic transcriptome analysis was performed. In the liver, semaglutide significantly reduced macrovesicular steatosis (-74%, p < 0.001) and inflammation (-73%, p < 0.001) and completely abolished microvesicular steatosis (-100%, p < 0.001). Histological and biochemical assessment of hepatic fibrosis showed no significant effects of semaglutide. However, digital pathology revealed significant improvements in the degree of collagen fiber reticulation (-12%, p < 0.001). Semaglutide did not affect atherosclerosis relative to controls. Additionally, we compared the transcriptome profile of FFD-fed Ldlr-/-.Leiden mice with a human gene set that differentiates human NASH patients with severe fibrosis from those with mild fibrosis. In FFD-fed Ldlr-/-.Leiden control mice, this gene set was upregulated as well, while semaglutide predominantly reversed this gene expression. Using a translational model with advanced NASH, we demonstrated that semaglutide is a promising candidate with particular potential for the treatment of hepatic steatosis and inflammation, while for the reversal of advanced fibrosis, combinations with other NASH agents may be necessary.
Collapse
Affiliation(s)
- José A Inia
- Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
- Cardiology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| | - Geurt Stokman
- Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Martine C Morrison
- Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Nicole Worms
- Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Lars Verschuren
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Martien P M Caspers
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Aswin L Menke
- Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | | | - Li Chen
- PharmaNest Inc., Princeton, NJ 08540, USA
| | | | - J Wouter Jukema
- Cardiology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
- Netherlands Heart Institute, 3511 EP Utrecht, The Netherlands
| | - Hans M G Princen
- Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Anita M van den Hoek
- Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| |
Collapse
|
14
|
Inia JA, Stokman G, Pieterman EJ, Morrison MC, Menke AL, Verschuren L, Caspers MPM, Giera M, Jukema JW, van den Hoek AM, Princen HMG. Atorvastatin Attenuates Diet-Induced Non-Alcoholic Steatohepatitis in APOE*3-Leiden Mice by Reducing Hepatic Inflammation. Int J Mol Sci 2023; 24:ijms24097818. [PMID: 37175538 PMCID: PMC10178767 DOI: 10.3390/ijms24097818] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Patients with metabolic syndrome are often prescribed statins to prevent the development of cardiovascular disease. Conversely, data on their effects on non-alcoholic steatohepatitis (NASH) are lacking. We evaluated these effects by feeding APOE*3-Leiden mice a Western-type diet (WTD) with or without atorvastatin to induce NASH and hepatic fibrosis. Besides the well-known plasma cholesterol lowering (-30%) and anti-atherogenic effects (severe lesion size -48%), atorvastatin significantly reduced hepatic steatosis (-22%), the number of aggregated inflammatory cells in the liver (-80%) and hepatic fibrosis (-92%) compared to WTD-fed mice. Furthermore, atorvastatin-treated mice showed less immunohistochemically stained areas of inflammation markers. Atorvastatin prevented accumulation of free cholesterol in the form of cholesterol crystals (-78%). Cholesterol crystals are potent inducers of the NLRP3 inflammasome pathway and atorvastatin prevented its activation, which resulted in reduced expression of the pro-inflammatory cytokines interleukin (IL)-1β (-61%) and IL-18 (-26%). Transcriptome analysis confirmed strong reducing effects of atorvastatin on inflammatory mediators, including NLRP3, NFκB and TLR4. The present study demonstrates that atorvastatin reduces hepatic steatosis, inflammation and fibrosis and prevents cholesterol crystal formation, thereby precluding NLRP3 inflammasome activation. This may render atorvastatin treatment as an attractive approach to reduce NAFLD and prevent progression into NASH in dyslipidemic patients.
Collapse
Affiliation(s)
- José A Inia
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| | - Geurt Stokman
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Elsbet J Pieterman
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Martine C Morrison
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Aswin L Menke
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Martien P M Caspers
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
- Netherlands Heart Institute, 3511 EP Utrecht, The Netherlands
| | - Anita M van den Hoek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| | - Hans M G Princen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands
| |
Collapse
|
15
|
Kallapur A, Sallam T. Pharmacotherapy in familial hypercholesterolemia - Current state and emerging paradigms. Trends Cardiovasc Med 2023; 33:170-179. [PMID: 34968676 DOI: 10.1016/j.tcm.2021.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/01/2022]
Abstract
Familial hypercholesterolemia is a highly prevalent but underdiagnosed disease marked by increased risk of cardiovascular morbidity and mortality. Aggressive reduction of LDL-cholesterol is a hallmark of cardiovascular risk mitigation in familial hypercholesterolemia. More recently, we have witnessed an expanded repertoire of pharmacologic agents that directly target LDL-cholesterol and/or reduce heart disease burden. In this state-of-the-art review, we explore the development, clinical efficacy and limitations of existing and potential future therapeutics in familial hypercholesterolemia.
Collapse
Affiliation(s)
- Aneesh Kallapur
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, CA, United States; Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, United States
| | - Tamer Sallam
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, CA, United States; Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, United States.
| |
Collapse
|
16
|
van Gemert Y, Blom AB, Di Ceglie I, Walgreen B, Helsen M, Sloetjes A, Vogl T, Roth J, Kruisbergen NNL, Pieterman EJ, Princen HMG, van der Kraan PM, van Lent PLEM, van den Bosch MHJ. Intensive cholesterol-lowering treatment reduces synovial inflammation during early collagenase-induced osteoarthritis, but not pathology at end-stage disease in female dyslipidemic E3L.CETP mice. Osteoarthritis Cartilage 2023:S1063-4584(23)00703-3. [PMID: 36898656 DOI: 10.1016/j.joca.2023.01.577] [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: 07/28/2022] [Revised: 01/12/2023] [Accepted: 01/31/2023] [Indexed: 03/12/2023]
Abstract
INTRODUCTION The association between metabolic syndrome (MetS) and osteoarthritis (OA) development has become increasingly recognized. In this context, the exact role of cholesterol and cholesterol-lowering therapies in OA development has remained elusive. Recently, we did not observe beneficial effects of intensive cholesterol-lowering treatments on spontaneous OA development in E3L.CETP mice. We postulated that in the presence of local inflammation caused by a joint lesion, cholesterol-lowering therapies may ameliorate OA pathology. MATERIALS AND METHODS Female ApoE3∗Leiden.CETP mice were fed a cholesterol-supplemented Western type diet. After 3 weeks, half of the mice received intensive cholesterol-lowering treatment consisting of atorvastatin and the anti-PCSK9 antibody alirocumab. Three weeks after the start of the treatment, OA was induced via intra-articular injections of collagenase. Serum levels of cholesterol and triglycerides were monitored throughout the study. Knee joints were analyzed for synovial inflammation, cartilage degeneration, subchondral bone sclerosis and ectopic bone formation using histology. Inflammatory cytokines were determined in serum and synovial washouts. RESULTS Cholesterol-lowering treatment strongly reduced serum cholesterol and triglyceride levels. Mice receiving cholesterol-lowering treatment showed a significant reduction in synovial inflammation (P = 0.008, WTD: 95% CI: 1.4- 2.3; WTD + AA: 95% CI: 0.8- 1.5) and synovial lining thickness (WTD: 95% CI: 3.0-4.6, WTD + AA: 95% CI: 2.1-3.2) during early-stage collagenase-induced OA. Serum levels of S100A8/A9, MCP-1 and KC were significantly reduced after cholesterol-lowering treatment (P = 0.0005, 95% CI: -46.0 to -12.0; P = 2.8 × 10-10, 95% CI: -398.3 to -152.1; P = 2.1 × 10-9, -66.8 to -30.4, respectively). However, this reduction did not reduce OA pathology, determined by ectopic bone formation, subchondral bone sclerosis and cartilage damage at end-stage disease. CONCLUSION This study shows that intensive cholesterol-lowering treatment reduces joint inflammation after induction of collagenase-induced OA, but this did not reduce end stage pathology in female mice.
Collapse
Affiliation(s)
- Y van Gemert
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - I Di Ceglie
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - B Walgreen
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M Helsen
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A Sloetjes
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - T Vogl
- Institute of Immunology, University of Münster, Germany
| | - J Roth
- Institute of Immunology, University of Münster, Germany
| | - N N L Kruisbergen
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - E J Pieterman
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - H M G Princen
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - P M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - P L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M H J van den Bosch
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
17
|
van Gemert Y, Kruisbergen NNL, Blom AB, van den Bosch MHJ, van der Kraan PM, Pieterman EJ, Princen HMG, van Lent PLEM. IL-1β inhibition combined with cholesterol-lowering therapies decreases synovial lining thickness and spontaneous cartilage degeneration in a humanized dyslipidemia mouse model. Osteoarthritis Cartilage 2023; 31:340-350. [PMID: 36442605 DOI: 10.1016/j.joca.2022.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Both systemic inflammation and dyslipidemia contribute to osteoarthritis (OA) development and have been suggested as a possible link between metabolic disease and OA development. Recently, the CANTOS trial showed a reduction in knee and hip replacements after inhibition of IL-1β in patients with a history of cardiovascular disease and high inflammatory risk. In this light, we investigated whether inhibition of IL-1β combined with cholesterol-lowering therapies can reduce OA development in dyslipidemic APOE∗3Leiden mice under pro-inflammatory dietary conditions. MATERIALS AND METHODS Female ApoE3∗Leiden mice were fed a cholesterol-supplemented Western-Type diet (WTD) for 38 weeks. After 14 weeks, cholesterol-lowering and anti-inflammatory treatments were started. Treatments included atorvastatin alone or with an anti-IL1β antibody, and atorvastatin combined with proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitor alirocumab without or with the anti-IL1β antibody. Knee joints were analyzed for cartilage degradation, synovial inflammation and ectopic bone formation using histology at end point. RESULTS Cholesterol-lowering treatment successfully decreased systemic inflammation in dyslipidemic mice, which was not further affected by inhibition of IL-1β. Synovial thickening and cartilage degeneration were significantly decreased in mice that received cholesterol-lowering treatment combined with inhibition of IL-1β (P < 0.01, P < 0.05, respectively) compared to mice fed a WTD alone. Ectopic bone formation was comparable between all groups. CONCLUSION These results indicate that inhibition of IL-1β combined with cholesterol-lowering therapy diminishes synovial thickening and cartilage degeneration in mice and may imply that this combination therapy could be beneficial in patients with metabolic inflammation.
Collapse
Affiliation(s)
- Y van Gemert
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - N N L Kruisbergen
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M H J van den Bosch
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - P M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - E J Pieterman
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - H M G Princen
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - P L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
18
|
Wu Z, Gao L, Lin Z. Can proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors regress coronary atherosclerotic plaque? A systematic review and meta-analysis. Am J Transl Res 2023; 15:452-465. [PMID: 36777825 PMCID: PMC9908469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/16/2022] [Indexed: 02/14/2023]
Abstract
OBJECTIVE Whether inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes the regression of coronary atherosclerotic plaque in statin-treated individuals remains unclear. This study examined whether PCSK9 inhibitors combined with statin therapy could increase atherosclerotic plaque regression compared with statin therapy alone. METHODS PubMed, the Cochrane Central Register of Controlled Trials (CENTRAL), the database Clinical trials, and the Web of Science were searched to report the coronary atherosclerotic plaque of PCSK9 inhibitors using intravascular ultrasonography (IVUS) or optical coherence tomography (OCT) in statin patients. The weighted mean difference (WMD) of the random-effects/fixed-effects model was used to pool data that satisfied our inclusion criteria obtained from the included studies. RESULTS When compared with statin therapy alone, pooled studies revealed that PCSK9 inhibitors combined with statin therapy significantly decreased percent atheroma volume (PAV) (WMD: -1.06%, 95% confidence interval [CI]: -1.39 to -0.73; P<0.001) and total atheroma volume (TAV) (WMD: -6.38 mm3, 95% CI: -10.12 to -2.64; P=0.001). Moreover, the fibrous cap thickness (FCT) of the coronary atherosclerotic plaque increases to 21.31 um (WMD: 21.31, 95% CI: 7.08 to 35.53, P<0.001), and the maximum lipid arc decreases 10.9° (WMD: -10.9, 95% CI: -15.24 to -5.34, P<0.001). CONCLUSION In our systematic review and meta-analysis, PCSK9 inhibitors combined with statin therapy were found to be more effective than statin therapy alone for slowing coronary plaque progression by decreasing PAV, TAV, and increasing FCT, maximum lipid arc.
Collapse
Affiliation(s)
- Zijia Wu
- Department of Cardiology, Yulin First People’s Hospital, The Sixth Affiliated Hospital of Guangxi Medical UniversityYulin 537000, Guangxi, China
| | - Lulan Gao
- Department of Laboratory, Yulin First People’s Hospital, The Sixth Affiliated Hospital of Guangxi Medical UniversityYulin 537000, Guangxi, China
| | - Zhihai Lin
- Department of Cardiology, Yulin First People’s Hospital, The Sixth Affiliated Hospital of Guangxi Medical UniversityYulin 537000, Guangxi, China
| |
Collapse
|
19
|
Surma S, Romańczyk M, Filipiak KJ. Angiopoietin-like proteins inhibitors: New horizons in the treatment of atherogenic dyslipidemia and familial hypercholesterolemia. Cardiol J 2023; 30:131-142. [PMID: 33470417 PMCID: PMC9987553 DOI: 10.5603/cj.a2021.0006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/10/2020] [Accepted: 09/25/2020] [Indexed: 11/25/2022] Open
Abstract
Angiopoietin-like proteins (ANGPTL) are involved in the regulation of numerous physiological and biochemical processes. ANGPTL3, 4 and 8, which are involved in the regulation of lipoprotein metabolism, are particularly important. ANGPTL3, 4 and 8 have been shown to regulate triglyceride availability depending on the nutritional status of the body. In addition, a deficiency of these proteins has been found to cause hypolipidemia (reduction of all lipid fractions). Increases in ANGPTL3, 4 and 8 appear to be associated with cardiovascular risk. Animal studies indicate that the use of ANGPTL3 (evinacumab) inhibitors significantly reduces plasma total cholesterol, triglycerides and low-density lipoprotein concentrations. The use of evinacumab in clinical trials also led to the normalization of plasma lipid concentrations in patients with atherogenic dyslipidemia and homozygous familial hypercholesterolemia. The results of these studies indicate that evinacumab may in the future be used in the treatment of lipid disorders, especially those with hypertriglyceridemia.
Collapse
Affiliation(s)
- Stanisław Surma
- Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Monika Romańczyk
- Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | | |
Collapse
|
20
|
Beltran RA, Zemeir KJ, Kimberling CR, Kneer MS, Mifflin MD, Broderick TL. Is a PCSK9 Inhibitor Right for Your Patient? A Review of Treatment Data for Individualized Therapy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16899. [PMID: 36554779 PMCID: PMC9779535 DOI: 10.3390/ijerph192416899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
INTRODUCTION In the United States, a significant amount of the population is affected by hyperlipidemia, which is associated with increased levels of serum low-density lipoprotein (LDL-C) and risk of cardiovascular disease. As of 2019, the guidelines set by the American College of Cardiology/American Heart Association advocate for the use of statins as the major contributor to lowering serum LDL-C. While proven to be effective, side effects, including muscle-related symptoms and new-onset diabetes mellitus, can make patients unable to tolerate statin therapy. Additionally, there is a subset of the population which does not approach a recommended LDL-C goal on statin treatment. Due to these findings, it was deemed necessary to review the literature of current statin-alternative lipid-lowering therapies. METHODS A systematic review of preclinical and clinical papers, and a current meta-analysis, was performed using PubMed and Google Scholar. Following the literature review, a meta-analysis was conducted using ProMeta 3. RESULTS Through systematic review and meta-analysis of the current literature, it is suggested that newer lipid-lowering therapies such as proprotein convertase subtilsin-kixen type 9 (PCSK9) inhibitors are a safe and effective statin alternative for the population with statin intolerance. PCSK9 inhibitors were shown to have no significant effect in causing myalgia in patients and showed no increase in adverse cardiovascular outcomes compared to a control of a current antilipemic medication regimen. DISCUSSION There are many statin-alternative therapies that should be investigated further as a potential replacement for patients with statin intolerance or as an addition for patients with statin resistance.
Collapse
Affiliation(s)
- Roman A. Beltran
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Kyle J. Zemeir
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Chase R. Kimberling
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Mary S. Kneer
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Michelle D. Mifflin
- College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Tom L. Broderick
- Laboratory of Diabetes and Exercise Metabolism, Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| |
Collapse
|
21
|
Kong N, Xu Q, Cui W, Feng X, Gao H. PCSK9 inhibitor inclisiran for treating atherosclerosis via regulation of endothelial cell pyroptosis. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1205. [PMID: 36544639 PMCID: PMC9761140 DOI: 10.21037/atm-22-4652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/25/2022] [Indexed: 11/21/2022]
Abstract
Background Proprotein convertase subtilisin/kexin type 9 (PCSK9) belongs to an intracellular invertase or decarboxylase and is an independent risk factor for atherosclerosis (AS). This study aimed to investigate the therapeutic potential of the PCSK9 inhibitor, inclisiran, and its underlying mechanism in AS. Methods ApoE-/- mice were fed with a high-fat diet (HFD) and intraperitoneally injected with 1, 5, or 10 mg/kg inclisiran. Low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC), triglyceride (TG), and high-density lipoprotein cholesterol (HDL-C) levels were determined using commercially available kits. Oil Red O staining was applied to detect the aortic plaque area and oil formation. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to induce cell injuries. Cell death was determined using a Hoechst 33342/propidium iodide (PI) dual-staining assay. Cytotoxicity was measured by lactate dehydrogenase (LDH) activity analysis. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot analyses were performed to examine the pyroptosis-related factors. Results Inclisiran inhibited the levels of LDL-C, TC, and TG, but increased the HDL-C level in the AS animal model. It also significantly inhibited plaque and oil droplet formation in a dose-dependent manner. Moreover, inclisiran markedly inhibited pyroptosis, as evidenced by the decreased levels of cleaved-caspase-1, NOD-like receptor family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase-1 recruitment domain (ASC), gasdermin-D (GSDMD)-N, interleukin (IL)-1β, and IL-18. Furthermore, inclisiran substantially inhibited cell death and cytotoxicity induced by ox-LDL in HUVECs. Conclusions Inclisiran exerted an anti-atherosclerotic effect by inhibiting pyroptosis. This study provides a theoretical basis for the therapeutic potential of inclisiran in AS.
Collapse
Affiliation(s)
- Ni Kong
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Qin Xu
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Wei Cui
- Basic Medical School, Qingdao University, Qingdao, China
| | - Xiaoying Feng
- School of Pharmacy, Guangzhou Medical University, Guangzhou, China
| | - Huijie Gao
- Department of Immunopharmacology, Jining Medical University, Rizhao, China
| |
Collapse
|
22
|
Duan Y, Gong K, Xu S, Zhang F, Meng X, Han J. Regulation of cholesterol homeostasis in health and diseases: from mechanisms to targeted therapeutics. Signal Transduct Target Ther 2022; 7:265. [PMID: 35918332 PMCID: PMC9344793 DOI: 10.1038/s41392-022-01125-5] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Disturbed cholesterol homeostasis plays critical roles in the development of multiple diseases, such as cardiovascular diseases (CVD), neurodegenerative diseases and cancers, particularly the CVD in which the accumulation of lipids (mainly the cholesteryl esters) within macrophage/foam cells underneath the endothelial layer drives the formation of atherosclerotic lesions eventually. More and more studies have shown that lowering cholesterol level, especially low-density lipoprotein cholesterol level, protects cardiovascular system and prevents cardiovascular events effectively. Maintaining cholesterol homeostasis is determined by cholesterol biosynthesis, uptake, efflux, transport, storage, utilization, and/or excretion. All the processes should be precisely controlled by the multiple regulatory pathways. Based on the regulation of cholesterol homeostasis, many interventions have been developed to lower cholesterol by inhibiting cholesterol biosynthesis and uptake or enhancing cholesterol utilization and excretion. Herein, we summarize the historical review and research events, the current understandings of the molecular pathways playing key roles in regulating cholesterol homeostasis, and the cholesterol-lowering interventions in clinics or in preclinical studies as well as new cholesterol-lowering targets and their clinical advances. More importantly, we review and discuss the benefits of those interventions for the treatment of multiple diseases including atherosclerotic cardiovascular diseases, obesity, diabetes, nonalcoholic fatty liver disease, cancer, neurodegenerative diseases, osteoporosis and virus infection.
Collapse
Affiliation(s)
- Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Suowen Xu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Feng Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xianshe Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China. .,College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.
| |
Collapse
|
23
|
Recent Updates in Hypertriglyceridemia Management for Cardiovascular Disease Prevention. Curr Atheroscler Rep 2022; 24:767-778. [PMID: 35895246 DOI: 10.1007/s11883-022-01052-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Mounting evidence continues to support the causal role of triglyceride-rich lipoproteins (TRL) in the development of atherosclerotic cardiovascular disease (ASCVD). Substantial residual ASCVD risk remains among high-risk patients who have elevated triglycerides despite reduction in low-density lipoprotein cholesterol (LDL-C) with statin therapy. Ongoing research efforts have focused on evaluating triglyceride-lowering therapies among patients with hypertriglyceridemia. RECENT FINDINGS The REDUCE-IT trial showed that the addition of icosapent ethyl, a highly purified form of eicosapentaenoic acid (EPA), can reduce vascular events among statin-treated individuals with elevated triglycerides who have either clinical ASCVD or diabetes plus another risk factor. Although additional evidence for EPA has emerged from other trials, conflicting results have been reported by subsequent trials that tested different omega-3 fatty acid formulations. Randomized clinical trials have not demonstrated incremental ASCVD benefit of fibrates on background of statin therapy, but fibrates are used to help prevent pancreatitis in patients with severe hypertriglyceridemia. Selective inhibitors of apolipoprotein C-III (apoC3) and angiopoietin-like protein 3 (ANGPTL3), proteins that are involved in metabolism of TRLs by regulating lipoprotein lipase, have been tested in selected patient populations and showed significant reduction in triglyceride and LDL-C levels. Statin therapy continues to be the cornerstone of pharmacologic reduction of cardiovascular risk. High-dose EPA in the form of icosapent ethyl has been demonstrated to have cardiovascular benefit on top of statins in persons with elevated triglycerides at high ASCVD risk. Ongoing clinical trials are evaluating novel selective therapies such as apoC3 and ANGPTL3 inhibitors.
Collapse
|
24
|
Aprotosoaie AC, Costache AD, Costache II. Therapeutic Strategies and Chemoprevention of Atherosclerosis: What Do We Know and Where Do We Go? Pharmaceutics 2022; 14:722. [PMID: 35456556 PMCID: PMC9025701 DOI: 10.3390/pharmaceutics14040722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 12/15/2022] Open
Abstract
Despite progress in understanding the pathogenesis of atherosclerosis, the development of effective therapeutic strategies is a challenging task that requires more research to attain its full potential. This review discusses current pharmacotherapy in atherosclerosis and explores the potential of some important emerging therapies (antibody-based therapeutics, cytokine-targeting therapy, antisense oligonucleotides, photodynamic therapy and theranostics) in terms of clinical translation. A chemopreventive approach based on modern research of plant-derived products is also presented. Future perspectives on preventive and therapeutic management of atherosclerosis and the design of tailored treatments are outlined.
Collapse
Affiliation(s)
- Ana Clara Aprotosoaie
- Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania;
| | - Alexandru-Dan Costache
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania;
| | - Irina-Iuliana Costache
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania;
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| |
Collapse
|
25
|
PCSK9 acts as a key regulator of Aβ clearance across the blood-brain barrier. Cell Mol Life Sci 2022; 79:212. [PMID: 35344086 PMCID: PMC8960591 DOI: 10.1007/s00018-022-04237-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 12/18/2022]
Abstract
Despite the neurodegenerative disorder Alzheimer's disease (AD) is the most common form of dementia in late adult life, there is currently no therapy available to prevent the onset or slow down the progression of AD. The progressive cognitive decline in AD correlates with a successive accumulation of cerebral amyloid-β (Aβ) due to impaired clearance mechanisms. A significant percentage is removed by low-density lipoprotein receptor-related protein 1 (LRP1)-mediated transport across the blood-brain barrier (BBB) into the periphery. Circulating proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to members of the low-density lipoprotein receptor protein family at the cell surface and targets them for lysosomal degradation, which reduces the number of functional receptors. However, the adverse impact of PCSK9 on LRP1-mediated brain Aβ clearance remains elusive. By using an established BBB model, we identified reduced LRP1-mediated brain-to-blood Aβ clearance due to PCSK9 across different endothelial monolayer in vitro. Consequently, the repetitive application of FDA-approved monoclonal anti-PCSK9 antibodies into 5xFAD mice decreased the cerebral Aβ burden across variants and aggregation state, which was not reproducible in brain endothelial-specific LRP1-/- 5xFAD mice. The peripheral PCSK9 inhibition reduced Aβ pathology in prefrontal cortex and hippocampus-brain areas critically involved in memory processing-and prevented disease-related impairment in hippocampus-dependent memory formation. Our data suggest that peripheral inhibition of PCSK9 by already available therapeutic antibodies may be a novel and easily applicable potential AD treatment.
Collapse
|
26
|
Gao Y, Zhang B, Yang J. Evinacumab for the treatment of homozygous familial hypercholesterolemia. Expert Rev Clin Pharmacol 2022; 15:139-145. [PMID: 35220876 DOI: 10.1080/17512433.2022.2047934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Yanli Gao
- Department of Clinical Pharmacy, Linyi Central Hospital, Linyi, Shandong, China
| | - Baoqi Zhang
- Department of Surgery, Linyi Central Hospital, Linyi, Shandong, China
| | - Junyi Yang
- Department of Clinical Pharmacy, Linyi Central Hospital, Linyi, Shandong, China
| |
Collapse
|
27
|
Cholesterol Lowering Biotechnological Strategies: From Monoclonal Antibodies to Antisense Therapies. A Pre-Clinical Perspective Review. Cardiovasc Drugs Ther 2022; 37:585-598. [PMID: 35022949 DOI: 10.1007/s10557-021-07293-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2021] [Indexed: 12/17/2022]
Abstract
In recent years, the increase in available genetic information and a better understanding of the genetic bases of dyslipidemias has led to the identification of potential new avenues for therapies. Additionally, the development of new technologies has presented the key for developing novel therapeutic strategies targeting not only proteins (e.g., the monoclonal antibodies and vaccines) but also the transcripts (from antisense oligonucleotides (ASOs) to small interfering RNAs) or the genomic sequence (gene therapies). These pharmacological advances have led to successful therapeutic improvements, particularly in the cardiovascular arena because we are now able to treat rare, genetically driven, and previously untreatable conditions (e.g, familial hypertriglyceridemia or hyperchylomicronemia). In this review, the pre-clinical pharmacological development of the major biotechnological cholesterol lowering advances were discussed, describing facts, gaps, potential future steps forward, and therapeutic opportunities.
Collapse
|
28
|
Pulipati VP, Alenghat FJ. The impact of lipid-lowering medications on coronary artery plaque characteristics. Am J Prev Cardiol 2021; 8:100294. [PMID: 34877559 PMCID: PMC8627965 DOI: 10.1016/j.ajpc.2021.100294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/29/2021] [Accepted: 11/06/2021] [Indexed: 11/28/2022] Open
Abstract
Atherosclerosis is the predominant cause of coronary artery disease. The last several decades have witnessed significant advances in lipid-lowering therapies, which comprise a central component of atherosclerotic cardiovascular disease prevention. In addition to cardiovascular risk reduction with dyslipidemia management, some lipid-based therapies show promise at the level of the atherosclerotic plaque itself through mechanisms governing lipid accumulation, plaque stability, local inflammation, endothelial dysfunction, and thrombogenicity. The capacity of lipid-lowering therapies to modify atherosclerotic plaque burden, size, composition, and vulnerability should correlate with their ability to reduce disease progression. This review discusses plaque characteristics, diagnostic modalities to evaluate these characteristics, and how they are altered by current and emerging lipid-lowering therapies, all in human coronary artery disease.
Collapse
Affiliation(s)
- Vishnu Priya Pulipati
- Section of Cardiology, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 6080, Chicago, IL 60637, United States
| | - Francis J. Alenghat
- Section of Cardiology, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 6080, Chicago, IL 60637, United States
- Pritzker School of Medicine, University of Chicago, Chicago, United States
| |
Collapse
|
29
|
Emerging Anti-Atherosclerotic Therapies. Int J Mol Sci 2021; 22:ijms222212109. [PMID: 34829992 PMCID: PMC8624828 DOI: 10.3390/ijms222212109] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/24/2021] [Accepted: 11/02/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CAD) is the main cause of morbidity and deaths in the western world. The development of atherosclerosis underlying CAD development begins early in human life. There are numerous genetic and environmental risk factors accelerating its progression which then leads to the occurrence of acute events. Despite considerable progress in determining risk factors, there is still a lot of work ahead since identified determinants are responsible only for a part of overall CAD risk. Current therapies are insufficient to successfully reduce the risk of atherosclerosis development. Therefore, there is a need for effective preventive measures of clinical manifestations of atherosclerosis since the currently available drugs cannot prevent the occurrence of even 70% of clinical events. The shift of the target from lipid metabolism has opened the door to many new therapeutic targets. Currently, the majority of known targets for anti-atherosclerotic drugs focus also on inflammation (a common mediator of many risk factors), mechanisms of innate and adaptive immunity in atherosclerosis, molecule scavengers, etc. The therapeutic potential of cyclodextrins, protein kinase inhibitors, colchicine, inhibitors of p38 mitogen-activated protein kinase (MAPK), lipid dicarbonyl scavengers, a monoclonal antibody targeting interleukin-1β, and P-selectin inhibitors is still not fully confirmed and requires confirmation in large clinical trials. The preliminary results look promising.
Collapse
|
30
|
Pieterman EJ, Princen HMG, Jarke A, Nilsson R, Cavallin A, Bergenholm L, Henricsson M, Gopaul VS, Agrawal R, Nissen SE, Hurt-Camejo E. Chronic Oral Administration of Mineral Oil Compared With Corn Oil: Effects on Gut Permeability and Plasma Inflammatory and Lipid Biomarkers. Front Pharmacol 2021; 12:681455. [PMID: 34483899 PMCID: PMC8415260 DOI: 10.3389/fphar.2021.681455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/02/2021] [Indexed: 12/02/2022] Open
Abstract
We investigated the effects of chronic oral administration of mineral oil, versus corn oil as control, on intestinal permeability, inflammatory markers, and plasma lipids in APOE*3-Leiden.CETP mice. Mice received mineral oil or corn oil 15 or 30 μL/mouse/day for 16 weeks (15 mice/group). Intestinal permeability was increased with mineral versus corn oil 30 µL/day, shown by increased mean plasma FITC-dextran concentrations 2 h post-administration (11 weeks: 1.5 versus 1.1 μg/ml, p = 0.02; 15 weeks: 1.7 versus 1.3 μg/ml, p = 0.08). Mean plasma lipopolysaccharide-binding protein levels were raised with mineral versus corn oil 30 µL/day (12 weeks: 5.8 versus 4.4 μg/ml, p = 0.03; 16 weeks: 5.8 versus 4.5 μg/ml, p = 0.09), indicating increased intestinal bacterial endotoxin absorption and potential pro-inflammatory effects. Plasma cholesterol and triglyceride concentrations were decreased with mineral oil, without affecting liver lipids among treated groups. Fecal neutral sterol measurements indicated increased fecal cholesterol excretion with mineral oil 30 µL/day (+16%; p = 0.04). Chronic oral administration of mineral oil in APOE*3-Leiden.CETP mice increased intestinal permeability, with potential pro-inflammatory effects, and decreased plasma cholesterol and triglyceride levels. Our findings may raise concerns about the use of mineral oil as a placebo in clinical studies.
Collapse
Affiliation(s)
- Elsbet J Pieterman
- The Netherlands Organisation for Applied Scientific Research (TNO), Metabolic Health Research, Leiden, Netherlands
| | - Hans M G Princen
- The Netherlands Organisation for Applied Scientific Research (TNO), Metabolic Health Research, Leiden, Netherlands
| | - Annica Jarke
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ralf Nilsson
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Cavallin
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Linnéa Bergenholm
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marcus Henricsson
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - V Sashi Gopaul
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rahul Agrawal
- Global Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Steven E Nissen
- Department of Cardiovascular Medicine and Cleveland Clinic Coordinating Center for Clinical Research, Cleveland Clinic, Cleveland, OH, United States
| | - Eva Hurt-Camejo
- Early Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
31
|
van Gemert Y, Kozijn AE, Pouwer MG, Kruisbergen NNL, van den Bosch MHJ, Blom AB, Pieterman EJ, Weinans H, Stoop R, Princen HMG, van Lent PLEM. Novel high-intensive cholesterol-lowering therapies do not ameliorate knee OA development in humanized dyslipidemic mice. Osteoarthritis Cartilage 2021; 29:1314-1323. [PMID: 33722697 DOI: 10.1016/j.joca.2021.02.570] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 02/03/2021] [Accepted: 02/25/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE High systemic cholesterol levels have been associated with osteoarthritis (OA) development. Therefore, cholesterol lowering by statins has been suggested as a potential treatment for OA. We investigated whether therapeutic high-intensive cholesterol-lowering attenuated OA development in dyslipidemic APOE∗3Leiden.CETP mice. METHODS Female mice (n = 13-16 per group) were fed a Western-type diet (WTD) for 38 weeks. After 13 weeks, mice were divided into a baseline group and five groups receiving WTD alone or with treatment: atorvastatin alone, combined with PCSK9 inhibitor alirocumab and/or ANGPTL3 inhibitor evinacumab. Knee joints were analysed for cartilage degradation, synovial inflammation and ectopic bone formation using histology. Aggrecanase activity in articular cartilage and synovial S100A8 expression were determined as markers of cartilage degradation/regeneration and inflammation. RESULTS Cartilage degradation and active repair were significantly increased in WTD-fed mice, but cholesterol-lowering strategies did not ameliorate cartilage destruction. This was supported by comparable aggrecanase activity and S100A8 expression in all treatment groups. Ectopic bone formation was comparable between groups and independent of cholesterol levels. CONCLUSIONS Intensive therapeutic cholesterol lowering per se did not attenuate progression of cartilage degradation in dyslipidemic APOE∗3Leiden.CETP mice, with minor joint inflammation. We propose that inflammation is a key feature in the disease and therapeutic cholesterol-lowering strategies may still be promising for OA patients presenting both dyslipidemia and inflammation.
Collapse
Affiliation(s)
- Y van Gemert
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A E Kozijn
- Metabolic Health Research, TNO, Leiden, the Netherlands; Department of Orthopaedics, UMC Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - M G Pouwer
- Metabolic Health Research, TNO, Leiden, the Netherlands; Department of Cardiology, Leiden UMC, Leiden, the Netherlands
| | - N N L Kruisbergen
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M H J van den Bosch
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - E J Pieterman
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - H Weinans
- Department of Orthopaedics, UMC Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, the Netherlands
| | - R Stoop
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - H M G Princen
- Metabolic Health Research, TNO, Leiden, the Netherlands
| | - P L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
32
|
Ling P, Zheng X, Luo S, Ge J, Xu S, Weng J. Targeting angiopoietin-like 3 in atherosclerosis: From bench to bedside. Diabetes Obes Metab 2021; 23:2020-2034. [PMID: 34047441 DOI: 10.1111/dom.14450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/10/2021] [Accepted: 05/23/2021] [Indexed: 12/13/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the largest cause of morbidity and mortality worldwide. Lipid-lowering therapies are the current major cornerstone of ASCVD management. Statins, ezetimibe, fibrates and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors effectively reduce the plasma low-density lipoprotein cholesterol (LDL-C) level in most individuals at risk of atherosclerosis. Still, some patients (such as those with homozygous familial hypercholesterolaemia), who do not respond to standard therapies, and other patients who cannot take these agents, remain at a high risk of ASCVD. In recent years there has been tremendous progress in understanding the mechanism and efficacy of lipid-lowering strategies. Apart from the recently approved PCSK9 and ATP citrate lyase inhibitors, angiopoietin-like 3 (ANGPTL3) is another potential target for the treatment of dyslipidaemia and its clinical sequalae of atherosclerosis. ANGPTL3 is a pivotal modulator of plasma triglycerides (TG), LDL-C and high-density lipoprotein cholesterol (HDL-C) levels, achieved by inhibiting the activities of lipoprotein lipase and endothelial lipase. Familial combined hypolipidaemia is derived from the Angptl3 loss-of-function mutations, which leads to low levels of LDL-C, HDL-C and TG, and has a 34% decreased risk of ASCVD compared with non-carriers. To date, monoclonal antibodies (evinacumab) and antisense oligonucleotides against ANGPTL3 have been investigated in clinical trials for dyslipidaemia therapy. Herein, we review the biology and function of ANGPTL3, as well as the latest developments of ANGPTL3-targeted therapies. We also summarize evidence from basic research to clinical trials, with the aim of providing novel insights into the biological functions of ANGPTL3 and related targeted therapies.
Collapse
Affiliation(s)
- Ping Ling
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xueying Zheng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sihui Luo
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Junbo Ge
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Cardiology, Zhong Shan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
33
|
Yeoh SG, Sum JS, Lai JY, W Isa WYH, Lim TS. Potential of Phage Display Antibody Technology for Cardiovascular Disease Immunotherapy. J Cardiovasc Transl Res 2021; 15:360-380. [PMID: 34467463 DOI: 10.1007/s12265-021-10169-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/22/2021] [Indexed: 11/26/2022]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. CVD includes coronary artery diseases such as angina, myocardial infarction, and stroke. "Lipid hypothesis" which is also known as the cholesterol hypothesis proposes the linkage of plasma cholesterol level with the risk of developing CVD. Conventional management involves the use of statins to reduce the serum cholesterol levels as means for CVD prevention or treatment. The regulation of serum cholesterol levels can potentially be regulated with biological interventions like monoclonal antibodies. Phage display is a powerful tool for the development of therapeutic antibodies with successes over the recent decade. Although mainly for oncology, the application of monoclonal antibodies as immunotherapeutic agents could potentially be expanded to CVD. This review focuses on the concept of phage display for antibody development and discusses the potential target antigens that could potentially be beneficial for serum cholesterol management.
Collapse
Affiliation(s)
- Soo Ghee Yeoh
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Jia Siang Sum
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Jing Yi Lai
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - W Y Haniff W Isa
- School of Medical Sciences, Department of Medicine, Universiti Sains Malaysia, Kubang Kerian, 16150, Kelantan, Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800, Penang, Malaysia.
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800, Penang, Malaysia.
| |
Collapse
|
34
|
Fortini F, Vieceli Dalla Sega F, Marracino L, Severi P, Rapezzi C, Rizzo P, Ferrari R. Well-Known and Novel Players in Endothelial Dysfunction: Updates on a Notch(ed) Landscape. Biomedicines 2021; 9:biomedicines9080997. [PMID: 34440201 PMCID: PMC8393382 DOI: 10.3390/biomedicines9080997] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
Endothelial dysfunction characterizes every aspect of the so-called cardiovascular continuum, a series of events ranging from hypertension to the development of atherosclerosis and, finally, to coronary heart disease, thrombus formation, myocardial infarction, and heart failure. Endothelial dysfunction is the main prognostic factor for the progression of vascular disorders, which responds to drug intervention and lifestyle changes. Virtually all of the drugs used to prevent cardiovascular disorders, such as long-used and new antilipidemic agents and inhibitors of angiotensin enzyme (ACEi), exert an important effect on the endothelium. Endothelial dysfunction is a central feature of coronavirus disease -19 (COVID-19), and it is now clear that life-risk complications of the disease are prompted by alterations of the endothelium induced by viral infection. As a consequence, the progression of COVID-19 is worse in the subjects in whom endothelial dysfunction is already present, such as elderly, diabetic, obese, and hypertensive patients. Importantly, circulating biomarkers of endothelial activation and injury predict the severity and mortality of the disease and can be used to evaluate the efficacy of treatments. The purpose of this review is to provide updates on endothelial function by discussing its clinical relevance in the cardiovascular continuum, the latest insights from molecular and cellular biology, and their implications for clinical practice, with a focus on new actors, such as the Notch signaling and emerging therapies for cardiovascular disease.
Collapse
Affiliation(s)
- Francesca Fortini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
| | | | - Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Paolo Severi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Claudio Rapezzi
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
- Correspondence: ; Tel.: +39-053-229-3707
| |
Collapse
|
35
|
Thompson GR. The scientific basis and future of lipoprotein apheresis. Ther Apher Dial 2021; 26:32-36. [PMID: 34331508 DOI: 10.1111/1744-9987.13716] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 01/27/2023]
Abstract
Lipoprotein apheresis plays a vital role in the management of the severe hyperlipidemias that predispose to atherosclerosis. Determinants of efficacy are the acute reduction in lipoproteins achieved by each apheresis procedure, their frequency, and the fractional catabolic rates and hence pool sizes of low-density lipoprotein (LDL) or lipoprotein (a) (Lp(a)) of the patient being treated. A useful criterion of the efficacy of apheresis plus lipid-lowering drug therapy is the decrease in the interval (time-averaged) mean of serum total or LDL cholesterol or Lp(a) between procedures, expressed as the percent decrease in the interval means below the maximal levels of these lipoproteins when off all treatment. Recent advances in lipid-lowering drug therapy may diminish the use of lipoprotein apheresis but will not abolish its unique role as a therapeutic "last chance saloon," especially for children and pregnant women with homozygous familial hypercholesterolemia.
Collapse
Affiliation(s)
- Gilbert R Thompson
- Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London, UK
| |
Collapse
|
36
|
Abstract
Introduction: Familial hypercholesterolemia (FH) is characterized by lifelong elevation of low-density lipoprotein cholesterol (LDL-C), early onset coronary atherosclerosis, and premature death. FH is underdiagnosed and undertreated, but requires aggressive LDL-C-lowering to prevent complications. Current treatment strategies such as lifestyle modification and numerous LDL-C-lowering medications are often insufficient to achieve lipid goals in FH.Areas covered: Angiopoietin-like 3 protein (ANGPTL3) is intricately involved in lipid metabolism. Loss-of-function mutations in ANGPTL3 are associated with panhypolipidemia and reduced coronary atherosclerosis. Evinacumab, a fully human monoclonal antibody, inhibits ANGPTL3 and reduces multiple lipoprotein fractions ~50%, including LDL-C. The use of evinacumab within the FH population is described as well as its regulatory journey to an approved therapeutic.Expert opinion: Evinacumab, with its capacity to lower multiple lipoprotein fractions, particularly LDL-C, independently of LDLR function has potential to revolutionize treatment for FH patients. Current FDA-approval is only for homozygous FH (HoFH), arguably the most impactful indication, but use in other lipid disorders is under investigation. The short-term tolerability of evinacumab is very good, with infrequent, mild, and transient adverse events; however, long-term safety data are needed. The high cost and requirement for intravenous administration may limit adoption of evinacumab, but dramatic LDL-C-lowering and need for new therapeutic options for HoFH will drive interest.
Collapse
Affiliation(s)
- Bruce A Warden
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, USA
| | - P Barton Duell
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, USA.,Division of Endocrinology, Diabetes and Clinical Nutrition, Oregon Health & Science University, Portland, USA
| |
Collapse
|
37
|
The Importance of Lipoprotein Lipase Regulation in Atherosclerosis. Biomedicines 2021; 9:biomedicines9070782. [PMID: 34356847 PMCID: PMC8301479 DOI: 10.3390/biomedicines9070782] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 02/07/2023] Open
Abstract
Lipoprotein lipase (LPL) plays a major role in the lipid homeostasis mainly by mediating the intravascular lipolysis of triglyceride rich lipoproteins. Impaired LPL activity leads to the accumulation of chylomicrons and very low-density lipoproteins (VLDL) in plasma, resulting in hypertriglyceridemia. While low-density lipoprotein cholesterol (LDL-C) is recognized as a primary risk factor for atherosclerosis, hypertriglyceridemia has been shown to be an independent risk factor for cardiovascular disease (CVD) and a residual risk factor in atherosclerosis development. In this review, we focus on the lipolysis machinery and discuss the potential role of triglycerides, remnant particles, and lipolysis mediators in the onset and progression of atherosclerotic cardiovascular disease (ASCVD). This review details a number of important factors involved in the maturation and transportation of LPL to the capillaries, where the triglycerides are hydrolyzed, generating remnant lipoproteins. Moreover, LPL and other factors involved in intravascular lipolysis are also reported to impact the clearance of remnant lipoproteins from plasma and promote lipoprotein retention in capillaries. Apolipoproteins (Apo) and angiopoietin-like proteins (ANGPTLs) play a crucial role in regulating LPL activity and recent insights into LPL regulation may elucidate new pharmacological means to address the challenge of hypertriglyceridemia in atherosclerosis development.
Collapse
|
38
|
Ruotsalainen AK, Mäkinen P, Ylä-Herttuala S. Novel RNAi-Based Therapies for Atherosclerosis. Curr Atheroscler Rep 2021; 23:45. [PMID: 34146172 PMCID: PMC8214045 DOI: 10.1007/s11883-021-00938-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Atherosclerosis, defined by inflammation and accumulation of cholesterol, extracellular matrix, and cell debris into the arteries is a common factor behind cardiovascular diseases (CVD), such as coronary artery disease, peripheral artery disease, and stroke. In this review, we discuss and describe novel RNA interference (RNAi)-based therapies in clinical trials and on the market. RECENT FINDINGS The first RNAi-based therapies have entered clinical use for the control of atherosclerosis risk factors, i.e., blood cholesterol levels. The most advanced treatment is silencing of proprotein convertase subtilisin/kexin type 9 (PCSK9) with a drug called inclisiran, which has been approved for the treatment of hypercholesterolemia in late 2020, and results in a robust decrease in plasma cholesterol levels. As the new RNAi therapies for atherosclerosis are now entering markets, the usefulness of these therapies will be further evaluated in larger patient cohorts. Thus, it remains to be seen how fast, effectively and eminently these new drugs consolidate their niche within the cardiovascular disease drug palette.
Collapse
Affiliation(s)
- Anna-Kaisa Ruotsalainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
- Kuopio Center for Gene and Cell Therapy, FIN-70210 Kuopio, Finland
| | - Petri Mäkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, P.O. Box 1777, FIN-70211 Kuopio, Finland
| |
Collapse
|
39
|
Effects of mineral oil administration on the pharmacokinetics, metabolism and pharmacodynamics of atorvastatin and pravastatin in mice and dogs. Eur J Pharm Sci 2021; 161:105776. [PMID: 33667667 DOI: 10.1016/j.ejps.2021.105776] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/16/2022]
Abstract
We investigated the effects of mineral oil on statin pharmacokinetics and inflammatory markers in animal models. A new synthesis strategy produced regioisomers that facilitated the characterization of the main metabolite (M1) of atorvastatin, a lipophilic statin, in C57BL/6NCrl mice. The chemical structure of M1 in mice was confirmed as ortho-hydroxy β-oxidized atorvastatin. Atorvastatin and M1 pharmacokinetics and inflammatory markers were assessed in C57BL6/J mice given atorvastatin 5 mg/kg/day or 10 mg/kg/day, as a single dose or for 21 days, with or without 10 µL or 30 µL mineral oil. No consistent differences in plasma exposure of atorvastatin or M1 were observed in mice after single or repeat dosing of atorvastatin with or without mineral oil. However, mice administered atorvastatin 10 mg/kg with 30 µL mineral oil for 21 days had significantly increased plasma levels of serum amyloid A (mean 9.6 µg/mL vs 7.9 µg/mL without mineral oil; p < 0.01) and significantly increased proportions of C62Lhigh B cells (mean 18% vs 12% without mineral oil; p = 0.04). There were no statistically significant differences for other inflammatory markers assessed. In dogs, pharmacokinetics of atorvastatin, its two hydroxy metabolites and pravastatin (a hydrophilic statin) were evaluated after single administration of atorvastatin 10 mg plus pravastatin 40 mg with or without 2 g mineral oil. Pharmacokinetics of atorvastatin, hydroxylated atorvastatin metabolites or pravastatin were not significantly different after single dosing with or without mineral oil in dogs. Collectively, the results in mice and dogs indicate that mineral oil does not affect atorvastatin or pravastatin pharmacokinetics, but could cause low-grade inflammation with chronic oral administration, which warrants further investigation.
Collapse
|
40
|
Fragki S, Dirven H, Fletcher T, Grasl-Kraupp B, Bjerve Gützkow K, Hoogenboom R, Kersten S, Lindeman B, Louisse J, Peijnenburg A, Piersma AH, Princen HMG, Uhl M, Westerhout J, Zeilmaker MJ, Luijten M. Systemic PFOS and PFOA exposure and disturbed lipid homeostasis in humans: what do we know and what not? Crit Rev Toxicol 2021; 51:141-164. [PMID: 33853480 DOI: 10.1080/10408444.2021.1888073] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Associations between per- and polyfluoroalkyl substances (PFASs) and increased blood lipids have been repeatedly observed in humans, but a causal relation has been debated. Rodent studies show reverse effects, i.e. decreased blood cholesterol and triglycerides, occurring however at PFAS serum levels at least 100-fold higher than those in humans. This paper aims to present the main issues regarding the modulation of lipid homeostasis by the two most common PFASs, PFOS and PFOA, with emphasis on the underlying mechanisms relevant for humans. Overall, the apparent contrast between human and animal data may be an artifact of dose, with different molecular pathways coming into play upon exposure to PFASs at very low versus high levels. Altogether, the interpretation of existing rodent data on PFOS/PFOA-induced lipid perturbations with respect to the human situation is complex. From a mechanistic perspective, research on human liver cells shows that PFOS/PFOA activate the PPARα pathway, whereas studies on the involvement of other nuclear receptors, like PXR, are less conclusive. Other data indicate that suppression of the nuclear receptor HNF4α signaling pathway, as well as perturbations of bile acid metabolism and transport might be important cellular events that require further investigation. Future studies with human-relevant test systems would help to obtain more insight into the mechanistic pathways pertinent for humans. These studies shall be designed with a careful consideration of appropriate dosing and toxicokinetics, so as to enable biologically plausible quantitative extrapolations. Such research will increase the understanding of possible perturbed lipid homeostasis related to PFOS/ PFOA exposure and the potential implications for human health.
Collapse
Affiliation(s)
- Styliani Fragki
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Hubert Dirven
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Tony Fletcher
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England (PHE), Chilton, UK
| | - Bettina Grasl-Kraupp
- Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, Vienna, Austria
| | | | - Ron Hoogenboom
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Birgitte Lindeman
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Jochem Louisse
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Ad Peijnenburg
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Aldert H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.,Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Hans M G Princen
- Metabolic Health Research, The Netherlands Organization of Applied Scientific Research (TNO), Gaubius Laboratory, Leiden, The Netherlands
| | - Maria Uhl
- Environment Agency Austria (EAA), Vienna, Austria
| | - Joost Westerhout
- Risk Analysis for Products In Development, The Netherlands Organization of Applied Scientific Research (TNO), Utrecht, The Netherlands
| | - Marco J Zeilmaker
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
41
|
Rosenson RS. Existing and emerging therapies for the treatment of familial hypercholesterolemia. J Lipid Res 2021; 62:100060. [PMID: 33716107 PMCID: PMC8065289 DOI: 10.1016/j.jlr.2021.100060] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/12/2021] [Accepted: 02/21/2021] [Indexed: 12/30/2022] Open
Abstract
Familial hypercholesterolemia (FH), an autosomal dominant disorder of LDL metabolism that is characterized by elevated LDL-cholesterol, is commonly encountered in patients with atherosclerotic coronary heart disease. Combinations of cholesterol-lowering therapies are often used to lower LDL-cholesterol in patients with FH; however, current treatment goals for LDL-cholesterol are rarely achieved in patients with homozygous FH (HoFH) and are difficult to achieve in patients with heterozygous FH (HeFH). Therapies that lower LDL-cholesterol through LDL receptor-mediated mechanisms have thus far been largely ineffective in patients with HoFH, particularly in those with negligible (<2%) LDL receptor activity. Among patients with HeFH who were at very high risk for atherosclerotic cardiovascular disease events, combined therapy consisting of a high dose of high-intensity statin, ezetimibe, and proprotein convertase subtilisin Kexin type 9 inhibitor failed to lower LDL-cholesterol to minimal acceptable goals in more than 50%. This article provides a framework for the use of available and emerging treatments that lower LDL-cholesterol in adult patients with HoFH and HeFH. A framework is provided for the use of angiopoietin-like protein 3 inhibitors in the treatment of HoFH and HeFH.
Collapse
Affiliation(s)
- Robert S Rosenson
- Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health. Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
42
|
Akoumianakis I, Zvintzou E, Kypreos K, Filippatos TD. ANGPTL3 and Apolipoprotein C-III as Novel Lipid-Lowering Targets. Curr Atheroscler Rep 2021; 23:20. [PMID: 33694000 DOI: 10.1007/s11883-021-00914-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Despite significant progress in plasma lipid lowering strategies, recent clinical trials highlight the existence of residual cardiovascular risk. Angiopoietin-like protein 3 (ANGPTL3) and apolipoprotein C-III (Apo C-III) have been identified as novel lipid-lowering targets. RECENT FINDINGS Apo C-III and ANGPTL3 have emerged as novel regulators of triglyceride (TG) and low-density lipoprotein-cholesterol (LDL-C) levels. ANGPTL3 is an inhibitor of lipoprotein lipase (LPL), reducing lipolysis of Apo B-containing lipoproteins. Loss-of-function ANGPLT3 mutations are associated with reduced plasma cholesterol and TG, while novel ANGPLT3 inhibition strategies, including monoclonal antibodies (evinacumab), ANGPLT3 antisense oligonucleotides (IONIS-ANGPTL3-LRx), and small interfering RNA (siRNA) silencing techniques (ARO-ANG3), result in increased lipolysis and significant reductions of LDL-C and TG levels in phase I and II clinical trials. Similarly, Apo C-III inhibits LPL while promoting the hepatic secretion of TG-rich lipoproteins and preventing their clearance. Loss-of-function APOC3 mutations have been associated with reduced TG levels. Targeting of Apo C-III with volanesorsen, an APOC3 siRNA, results in significant reduction in plasma TG levels but possibly also increased risk for thrombocytopenia, as recently demonstrated in phase I, II, and III clinical trials. ARO-APOC3 is a novel siRNA-based agent targeting Apo C-III which is currently under investigation with regard to its lipid-lowering efficiency. ANGPTL3 and Apo C-III targeting agents have demonstrated striking lipid-lowering effects in recent clinical trials; however, more thorough safety and efficacy data are required. Here, we evaluate the role of ANGPLT3 and Apo C-III in lipid metabolism, present the latest clinical advances targeting those molecules, and outline the remaining scientific challenges on residual lipid-associated cardiovascular risk.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Department of Internal Medicine, School of Medicine, University Hospital of Heraklion, University of Crete, Heraklion, Crete, Greece.,Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Evangelia Zvintzou
- Department of Medicine, Pharmacology Laboratory, School of Health Sciences, University of Patras, Achaias, Rio, Greece
| | - Kyriakos Kypreos
- Department of Medicine, Pharmacology Laboratory, School of Health Sciences, University of Patras, Achaias, Rio, Greece.,Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Theodosios D Filippatos
- Department of Internal Medicine, School of Medicine, University Hospital of Heraklion, University of Crete, Heraklion, Crete, Greece. .,Metabolic Diseases Research Unit, Internal Medicine Laboratory, School of Sciences, Faculty of Medicine, University of Crete, P.O. Box 2208, Heraklion, Crete, Greece.
| |
Collapse
|
43
|
Abstract
After training as a gastroenterologist in the UK, the author became interested in lipidology while he was a research fellow in the USA and switched careers after returning home. Together with Nick Myant, he introduced the use of plasma exchange to treat familial hypercholesterolemia (FH) homozygotes and undertook non-steady state studies of LDL kinetics, which showed that the fractional catabolic rate of LDL remained constant irrespective of pool size. Subsequent steady-state turnover studies showed that FH homozygotes had an almost complete lack of receptor-mediated LDL catabolism, providing in vivo confirmation of the Nobel Prize-winning discovery by Goldstein and Brown that LDL receptor dysfunction was the cause of FH. Further investigation of metabolic defects in FH revealed that a significant proportion of LDL in homozygotes and heterozygotes was produced directly via a VLDL-independent pathway. Management of heterozygous FH has been greatly facilitated by statins and proprotein convertase subtilisin/kexin type 9 inhibitors but remains dependent upon lipoprotein apheresis in homozygotes. In a recent analysis of a large cohort treated with a combination of lipid-lowering measures, survival was markedly enhanced in homozygotes in the lowest quartile of on-treatment serum cholesterol. Emerging therapies could further improve the prognosis of homozygous FH; whereas in heterozygotes, the current need is better detection.
Collapse
Affiliation(s)
- Gilbert R Thompson
- Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom.
| |
Collapse
|
44
|
Viedma-Guiard E, Guidoux C, Amarenco P, Meseguer E. Aortic Sources of Embolism. Front Neurol 2021; 11:606663. [PMID: 33519684 PMCID: PMC7843443 DOI: 10.3389/fneur.2020.606663] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022] Open
Abstract
Aortic arch atheroma is a frequent finding in ischemic stroke patients. Its role as a source of cerebral emboli or a marker of atherosclerosis is unclear. Transesophageal echography is considered the gold standard for its detection, whereas computed tomography angiography is a good alternative; magnetic resonance and positron emission tomography could be proposed to better analyze plaque vulnerability. Despite the interest in this condition, the optimal antithrombotic treatment remains uncertain, while intensive lipid-lowering therapy should be recommended. This review aims to offer guidance on patients with aortic arch atheroma, about its causal role in stroke, diagnosis, and treatment based on current available evidence.
Collapse
Affiliation(s)
- Elena Viedma-Guiard
- Department of Neurology and Stroke Center, APHP Bichat Hospital, Paris, France Université de Paris, LVTS, Inserm U1148, Paris, France
| | - Celine Guidoux
- Department of Neurology and Stroke Center, APHP Bichat Hospital, Paris, France Université de Paris, LVTS, Inserm U1148, Paris, France
| | - Pierre Amarenco
- Department of Neurology and Stroke Center, APHP Bichat Hospital, Paris, France Université de Paris, LVTS, Inserm U1148, Paris, France
| | - Elena Meseguer
- Department of Neurology and Stroke Center, APHP Bichat Hospital, Paris, France Université de Paris, LVTS, Inserm U1148, Paris, France
| |
Collapse
|
45
|
Rakipovski G, Hovingh GK, Nyberg M. Proprotein convertase subtilisin/kexin type 9 inhibition as the next statin? Curr Opin Lipidol 2020; 31:340-346. [PMID: 33060383 DOI: 10.1097/mol.0000000000000718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Despite the wide use of statins and other LDL-cholesterol (LDL-C)-lowering therapies, atherosclerotic cardiovascular disease remains an important cause of mortality and morbidity. Here, we discuss efficacy, side effects and convenience of current and future therapies inhibiting proprotein convertase subtilisin/kexin type 9 (PCSK9). RECENT FINDINGS Clinical trials with mAbs administered every 2-4 weeks and small interfering RNAs given two to four times per year have consistently demonstrated substantial LDL-C-lowering (40-60%) and improved outcome when added to existing lipid-lowering therapies. Pleiotropic effects of PCSK9 inhibition are somewhat different from those observed with statin treatment as evidenced by reduced levels of triglycerides and lipoprotein(a) with no apparent effect on inflammatory markers in patients treated with PCSK9 inhibitors. Treatment with mAb and small interfering RNA are associated with a high-cost, however, small molecules and vaccines may improve cost and convenience if development of these are successful. SUMMARY PCSK9 inhibitors are currently considered to be an add-on therapy and whether these drugs will be used as stand-alone and/or as a first choice is dependent on clinical readouts from ongoing and future trials, real-world evidence, convenience and treatment costs.
Collapse
Affiliation(s)
| | - G Kees Hovingh
- Novo Nordisk A/S, Copenhagen, Denmark
- Department of Vascular Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| | | |
Collapse
|
46
|
Ke LY, Law SH, Mishra VK, Parveen F, Chan HC, Lu YH, Chu CS. Molecular and Cellular Mechanisms of Electronegative Lipoproteins in Cardiovascular Diseases. Biomedicines 2020; 8:biomedicines8120550. [PMID: 33260304 PMCID: PMC7760527 DOI: 10.3390/biomedicines8120550] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of glucose and lipid metabolism increases plasma levels of lipoproteins and triglycerides, resulting in vascular endothelial damage. Remarkably, the oxidation of lipid and lipoprotein particles generates electronegative lipoproteins that mediate cellular deterioration of atherosclerosis. In this review, we examined the core of atherosclerotic plaque, which is enriched by byproducts of lipid metabolism and lipoproteins, such as oxidized low-density lipoproteins (oxLDL) and electronegative subfraction of LDL (LDL(−)). We also summarized the chemical properties, receptors, and molecular mechanisms of LDL(−). In combination with other well-known markers of inflammation, namely metabolic diseases, we concluded that LDL(−) can be used as a novel prognostic tool for these lipid disorders. In addition, through understanding the underlying pathophysiological molecular routes for endothelial dysfunction and inflammation, we may reassess current therapeutics and might gain a new direction to treat atherosclerotic cardiovascular diseases, mainly targeting LDL(−) clearance.
Collapse
Affiliation(s)
- Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
- Graduate Institute of Medicine, College of Medicine and Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
| | - Shi Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
| | - Vineet Kumar Mishra
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
| | - Farzana Parveen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (L.-Y.K.); (S.H.L.); (V.K.M.); (F.P.)
| | - Hua-Chen Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
| | - Ye-Hsu Lu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
| | - Chih-Sheng Chu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (H.-C.C.); (Y.-H.L.)
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Correspondence: ; Tel.: +886-73121101 (ext. 2297); Fax: +886-73111996
| |
Collapse
|
47
|
Stokman G, van den Hoek AM, Denker Thorbekk D, Pieterman EJ, Skovgård Veidal S, Basta B, Iruarrizaga‐Lejarreta M, van der Hoorn JW, Verschuren L, Berbée JFP, Rensen PCN, Skjæret T, Alonso C, Feigh M, Kastelein JJP, Friedman SL, Princen HMG, Fraser DA. Dual targeting of hepatic fibrosis and atherogenesis by icosabutate, an engineered eicosapentaenoic acid derivative. Liver Int 2020; 40:2860-2876. [PMID: 32841505 PMCID: PMC7702170 DOI: 10.1111/liv.14643] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/01/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS While fibrosis stage predicts liver-associated mortality, cardiovascular disease (CVD) is still the major overall cause of mortality in patients with NASH. Novel NASH drugs should thus ideally reduce both liver fibrosis and CVD. Icosabutate is a semi-synthetic, liver-targeted eicosapentaenoic acid (EPA) derivative in clinical development for NASH. The primary aims of the current studies were to establish both the anti-fibrotic and anti-atherogenic efficacy of icosabutate in conjunction with changes in lipotoxic and atherogenic lipids in liver and plasma respectively. METHODS The effects of icosabutate on fibrosis progression and lipotoxicity were investigated in amylin liver NASH (AMLN) diet (high fat, cholesterol and fructose) fed ob/ob mice with biopsy-confirmed steatohepatitis and fibrosis and compared with the activity of obeticholic acid. APOE*3Leiden.CETP mice, a translational model for hyperlipidaemia and atherosclerosis, were used to evaluate the mechanisms underlying the lipid-lowering effect of icosabutate and its effect on atherosclerosis. RESULTS In AMLN ob/ob mice, icosabutate significantly reduced hepatic fibrosis and myofibroblast content in association with downregulation of the arachidonic acid cascade and a reduction in both hepatic oxidised phospholipids and apoptosis. In APOE*3Leiden.CETP mice, icosabutate reduced plasma cholesterol and TAG levels via increased hepatic uptake, upregulated hepatic lipid metabolism and downregulated inflammation pathways, and effectively decreased atherosclerosis development. CONCLUSIONS Icosabutate, a structurally engineered EPA derivative, effectively attenuates both hepatic fibrosis and atherogenesis and offers an attractive therapeutic approach to both liver- and CV-related morbidity and mortality in NASH patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Brittany Basta
- Division of Liver DiseasesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | | | | | - Jimmy F. P. Berbée
- Department. of MedicineDivision of EndocrinologyEinthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Patrick C. N. Rensen
- Department. of MedicineDivision of EndocrinologyEinthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Tore Skjæret
- NorthSea Therapeutics BVAmsterdamThe Netherlands
| | - Cristina Alonso
- Division of Liver DiseasesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - John J. P. Kastelein
- Department of Vascular MedicineAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Scott L. Friedman
- Division of Liver DiseasesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | | |
Collapse
|
48
|
Adam RC, Mintah IJ, Alexa-Braun CA, Shihanian LM, Lee JS, Banerjee P, Hamon SC, Kim HI, Cohen JC, Hobbs HH, Van Hout C, Gromada J, Murphy AJ, Yancopoulos GD, Sleeman MW, Gusarova V. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance. J Lipid Res 2020; 61:1271-1286. [PMID: 32646941 PMCID: PMC7469887 DOI: 10.1194/jlr.ra120000888] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
Angiopoietin-like protein (ANGPTL)3 regulates plasma lipids by inhibiting LPL and endothelial lipase (EL). ANGPTL3 inactivation lowers LDL-C independently of the classical LDLR-mediated pathway and represents a promising therapeutic approach for individuals with homozygous familial hypercholesterolemia due to LDLR mutations. Yet, how ANGPTL3 regulates LDL-C levels is unknown. Here, we demonstrate in hyperlipidemic humans and mice that ANGPTL3 controls VLDL catabolism upstream of LDL. Using kinetic, lipidomic, and biophysical studies, we show that ANGPTL3 inhibition reduces VLDL-lipid content and size, generating remnant particles that are efficiently removed from the circulation. This suggests that ANGPTL3 inhibition lowers LDL-C by limiting LDL particle production. Mechanistically, we discovered that EL is a key mediator of ANGPTL3's novel pathway. Our experiments revealed that, although dispensable in the presence of LDLR, EL-mediated processing of VLDL becomes critical for LDLR-independent particle clearance. In the absence of EL and LDLR, ANGPTL3 inhibition perturbed VLDL catabolism, promoted accumulation of atypical remnants, and failed to reduce LDL-C. Taken together, we uncover ANGPTL3 at the helm of a novel EL-dependent pathway that lowers LDL-C in the absence of LDLR.
Collapse
Affiliation(s)
- Rene C Adam
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | | | | | | | | | - Hye In Kim
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Jonathan C Cohen
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Helen H Hobbs
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Ruhanen H, Haridas PAN, Jauhiainen M, Olkkonen VM. Angiopoietin-like protein 3, an emerging cardiometabolic therapy target with systemic and cell-autonomous functions. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158791. [PMID: 32777482 DOI: 10.1016/j.bbalip.2020.158791] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/23/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022]
Abstract
Angiopoietin like protein 3 (ANGPTL3) is best known for its function as an inhibitor of lipoprotein and endothelial lipases. Due to the capacity of genetic or pharmacologic ANGPTL3 suppression to markedly reduce circulating lipoproteins, and the documented cardioprotection upon such suppression, ANGPTL3 has become an emerging therapy target for which both antibody and antisense oligonucleotide (ASO) therapeutics are being clinically tested. While the antibody is relatively selective for circulating ANGPTL3, the ASO also depletes the intra-hepatocellular protein, and there is emerging evidence for cell-autonomous functions of ANGPTL3 in the liver. These include regulation of hepatocyte glucose and fatty acid uptake, insulin sensitivity, LDL/VLDL remnant uptake, VLDL assembly/secretion, polyunsaturated fatty acid (PUFA) and PUFA-derived lipid mediator content, and gene expression. In this review we elaborate on (i) why ANGPTL3 is considered one of the most promising new cardiometabolic therapy targets, and (ii) the present evidences for its intra-hepatocellular or cell-autonomous functions.
Collapse
Affiliation(s)
- Hanna Ruhanen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Molecular and Integrative Biosciences, University of Helsinki, Finland
| | | | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
50
|
Stock JK. Homozygous familial hypercholesterolaemia: New hope for getting patients to goal? Atherosclerosis 2020; 304:55-56. [DOI: 10.1016/j.atherosclerosis.2020.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 11/15/2022]
|