1
|
Anchouche K, Baass A, Thanassoulis G. Lp(a): A Clinical Review. Clin Biochem 2025; 137:110929. [PMID: 40258460 DOI: 10.1016/j.clinbiochem.2025.110929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
Elevated lipoprotein(a) (Lp[a]) is a genetically determined cardiovascular risk factor, linked to both atherosclerotic cardiovascular disease and aortic stenosis. Elevated Lp(a) is widely prevalent, and consequently, several cardiovascular societies now recommend performing Lp(a) screening at least once in all adults. While there are presently no approved drugs specifically aimed at lowering Lp(a), several promising candidates are currently in the drug development pipeline, and many of these are now undergoing late phase clinical trials. In this comprehensive review, we outline Lp(a) biology and genetics, describe Lp(a)'s relationship to various cardiovascular clinical phenotypes of interest, highlight novel Lp(a)-lowering therapies, and outline what role these may have in future clinical practice.
Collapse
Affiliation(s)
- Khalil Anchouche
- McGill University Health Centre and Research Institute, Montreal, QC, Canada; McGill University, Montreal, QC, Canada
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal, QC, Canada; Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montreal, QC, Canada
| | - George Thanassoulis
- McGill University Health Centre and Research Institute, Montreal, QC, Canada; McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Igarashi R, Oda M, Okada R, Yano T, Takahashi S, Pastuhov S, Matano M, Masuda N, Togasaki K, Ohta Y, Sato S, Hishiki T, Suematsu M, Itoh M, Fujii M, Sato T. Generation of human adult hepatocyte organoids with metabolic functions. Nature 2025:10.1038/s41586-025-08861-y. [PMID: 40240606 DOI: 10.1038/s41586-025-08861-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025]
Abstract
Proliferating hepatocytes often undergo ductal metaplasia to balance the energy trade-off between cellular functions and replication, hindering the expansion of human adult hepatocytes with functional competency1. Here we demonstrate that the combined activation of Wnt and STAT3 signalling enables long-term self-renewal of human adult hepatocyte organoids. YAP activation facilitates hepatocyte proliferation but commits it towards the biliary duct lineage. By contrast, STAT3 activation by oncostatin M induces hepatocyte proliferation while counteracting ductal metaplasia and maintaining the hepatic identity. Xenotransplanted hepatocyte organoids repopulate the recipient mouse liver and reconstitute the metabolic zonation structure. Upon niche factor removal and hormone supplementation, hepatocyte organoids form cord-like structures with bile canalicular networks and exhibit major liver metabolic functions comparable to those of in vivo hepatocytes. Hepatocyte organoids are amenable to gene editing, prompting functional modelling of inherent metabolic liver diseases. The new culture system offers a promising avenue for developing therapeutic strategies against human liver diseases.
Collapse
Affiliation(s)
- Ryo Igarashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mayumi Oda
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Okada
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Tomoki Yano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Strahil Pastuhov
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Norio Masuda
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Kazuhiro Togasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Saeko Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takako Hishiki
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Suematsu
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Manabu Itoh
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
3
|
Wang Y, Choe JYS, Shi Y, Thi TT, Cao X, Hu Y, Cheng KY, Li H, Ji Y, Liu Y, Ackers-Johnson M, Foo RSY, Shen Y, Yu H. Depletion of Hepatic SREBP2 Protects Against Hypercholesterolemia and Atherosclerosis through the ANGPTL3-LPL Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412677. [PMID: 40106311 DOI: 10.1002/advs.202412677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/13/2025] [Indexed: 03/22/2025]
Abstract
Lipolysis of triglyceride-rich lipoproteins by peripheral lipoprotein lipase (LPL) plays an essential role in maintaining systemic cholesterol/lipid homeostasis. Human genetic studies have unequivocally demonstrated that activation of LPL pathway reduces risks for both coronary artery disease (CAD) and type 2 diabetes (T2D). Although sterol regulatory element-binding protein 2 (SREBP2) is well established as the master transcription factor that regulates the hepatic biosynthesis of both cholesterol and fatty acids, whether and how its activity in liver interacts with peripheral LPL pathway remains unknown. Here, it is demonstrated that acute liver-specific depletion of SREBP2 results in divergent effects on the regulation of peripheral LPL activity in mice, depending on the presence or absence of low-density lipoprotein receptors (LDLR). SREBP2 deficiency drastically elevates peripheral LPL activity through downregulation of plasma angiopoietin-related protein 3 (ANGPTL3) levels in LDLR-deficient mice. Moreover, in addition to SREBP2's transcriptional regulation of ANGPTL3, it is found that SREBP2 promotes proteasome-based degradation of ANGPTL3 in the presence of LDLR. Remarkably, acute depletion of hepatic SREBP2 protects against hypercholesterolemia and atherosclerosis, in which atherosclerotic lesions are reduced by 45% compared to control littermates. Taken together, these findings outline a liver-peripheral crosstalk mediated by SREBP2-ANGPTL3-LPL axis and suggest that SREBP2 inhibition can be an effective strategy to tackle homozygous familial hypercholesterolemia (HoFH).
Collapse
Affiliation(s)
- Yifan Wang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Jia You Sarafina Choe
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Yu Shi
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Thi Tun Thi
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Xiaoyun Cao
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Yang Hu
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Kai Yan Cheng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Hui Li
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Yang Ji
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Yan Liu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Matthew Ackers-Johnson
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Roger S Y Foo
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Yujia Shen
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Haojie Yu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| |
Collapse
|
4
|
Chen YH, Dettipponpong P, Sin MY, Chang LC, Cheng CY, Huang SY, Walzem RL, Cheng HC, Chen SE. Ovarian expression of functional MTTP and apoB for VLDL assembly and secretion in chickens. Poult Sci 2025; 104:104993. [PMID: 40073639 PMCID: PMC11951013 DOI: 10.1016/j.psj.2025.104993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
In mammals, tissues other than liver and intestine are known to possess functional MTTP (microsomal triglyceride transfer protein) and apoB (apolipoprotein B) capable of VLDL (very low-density lipoprotein) assembly. Birds are oviparous and possess unique capabilities in lipid biology to accommodate yolk formation through massive deposition of hepatically assembled yolk-targeted VLDLy into ovarian follicles. Following identifications of MTTP and ApoB expression within chicken ovarian stroma, granulosa, theca, and epithelial cells of various classes of follicles, we sought to define the functionality of ovarian MTTP and ApoB in VLDL assembly. In situ hybridization analysis found that ApoB transcripts are most abundant in thecal layers, whereas immunohistochemistry showed that MTTP predominates in the granulosa layers. MTTP lipid transfer activity was greater in small yellow follicles than in hierarchical follicles. Metabolic labeling, electron microscopy, and Western blot studies confirmed the functionality of ovarian apoB and MTTP as newly assembled VLDL around 50-200 nm in diameter and lacking ApoVLDL-II dissimilar to VLDLy, were secreted from cultured follicular cells. Lomitapide and the ApoB-antisense oligonucleotide Mipomersen dose-dependently decreased MTTP activity and VLDL-apoB secretion from cultured follicular cells, while oleate addition or acute heat stress enhanced VLDL-apoB secretion. Ultrastructural images showed VLDL assembly and trafficking toward the secretion route. The findings support the notion that VLDL assembly and secretion within avian ovarian tissues functions as a protective mechanism against fuel and physical stressors to secure follicle development and/or nutritional quality control of yolk for embryo development.
Collapse
Affiliation(s)
- Yu-Hui Chen
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan; Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | | | - Mei-Ying Sin
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404327, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 40402, Taiwan
| | - Ling-Chu Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404327, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 40402, Taiwan
| | - Chuen-Yu Cheng
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407224, Taiwan
| | - San-Yuan Huang
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan; The iEGG and Animal Biotechnology Center and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan; i-Center for Advanced Science and Technology (iCAST), National Chung Hsing University, Taichung 40227, Taiwan
| | - Rosemary L Walzem
- Department of Poultry Science, Texas A&M University, College Station, TX 77843, USA
| | - Hsu-Chen Cheng
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan; i-Center for Advanced Science and Technology (iCAST), National Chung Hsing University, Taichung 40227, Taiwan.
| | - Shuen-Ei Chen
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan; The iEGG and Animal Biotechnology Center and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan; i-Center for Advanced Science and Technology (iCAST), National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
5
|
Chen W, Chen Y, Song B, Zhai L, Tao G, Wang B, Liu B, Wang H, Zhang CX, Gu HM, Yin D, Qin S, Zhang DW. Inducible global knockout of surfeit locus protein 4 in adult mice results in hypolipidemia, intestinal lipid accumulation, liver injury, and increased mortality. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159577. [PMID: 39549860 DOI: 10.1016/j.bbalip.2024.159577] [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: 08/26/2024] [Revised: 11/03/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
Surfeit locus protein 4 (SURF4) acts as a cargo receptor to mediate endoplasmic reticulum export of various cargos. We have shown that SURF4 is essential for secretion of hepatic very low-density lipoprotein and intestinal chylomicron. Knockdown of hepatic Surf4 also significantly reduces the development of atherosclerosis and liver fibrosis without causing overt liver damage. However, constitutive global Surf4 knockout results in embryonic lethality. To further understand the physiological role of SURF4, we generated tamoxifen-inducible global Surf4 knockout mice. We found that conditional knockout of Surf4 in adult mice (Surf4ig-ko) significantly reduced mouse body weight. Male and female Surf4ig-ko mice died approximately 30 and 50 days after tamoxifen administration, respectively. Triglyceride secretion and serum levels of total cholesterol, triglycerides, free fatty acids, apolipoprotein B-100, and apolipoprotein B-48 were significantly reduced in Surf4ig-ko mice compared with Surf4flox mice. Proteomics analysis of mouse serum samples revealed 308 proteins with significantly altered expression in Surf4ig-ko mice that have unique functions and are involved in various biological processes. In addition, Surf4ig-ko mice exhibited lipid accumulation in the intestine but not in the liver. However, in Surf4ig-ko mice, liver weight was significantly reduced, and serum transaminase activity was significantly increased, indicating liver damage. Therefore, SURF4 is essential for survival in adult mice, suggesting that the therapeutic use of SURF4 requires precise tissue/cell type-specific targeting.
Collapse
Affiliation(s)
- Wei Chen
- The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; School of Clinic and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuan Chen
- The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; School of Clinic and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Baoye Song
- College of Animal Science and Technology, Shandong Agricultural University, Taian, China
| | - Lei Zhai
- The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; School of Clinic and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Geru Tao
- The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; School of Clinic and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Bingxiang Wang
- The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; School of Clinic and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Boyan Liu
- The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; School of Clinic and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Wang
- The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; School of Clinic and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Cindy X Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hong-Mei Gu
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Deling Yin
- The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Shucun Qin
- The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China; School of Clinic and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
6
|
Zhang L, Wang X, Chen XW. The biogenesis and transport of triglyceride-rich lipoproteins. Trends Endocrinol Metab 2025; 36:262-277. [PMID: 39164120 DOI: 10.1016/j.tem.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Triglyceride-rich lipoproteins (TRLs) play essential roles in human health and disease by transporting bulk lipids into the circulation. This review summarizes the fundamental mechanisms and diverse factors governing lipoprotein production, secretion, and regulation. Emphasizing the broader implications for human health, we outline the intricate landscape of lipoprotein research and highlight the potential coordination between the biogenesis and transport of TRLs in physiology, particularly the unexpected coupling of metabolic enzymes and transport machineries. Challenges and opportunities in lipoprotein biology with respect to inherited diseases and viral infections are also discussed. Further characterization of the biogenesis and transport of TRLs will advance both basic research in lipid biology and translational medicine for metabolic diseases.
Collapse
Affiliation(s)
- Linqi Zhang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China; Peking University (PKU)-Tsinghua University (THU) Joint Center for Life Sciences, Peking University, Beijing 100871, PR China.
| |
Collapse
|
7
|
Hakim A, Connally NJ, Schnitzler GR, Cho MH, Jiang ZG, Sunyaev SR, Gupta RM. Missing Regulation Between Genetic Association and Transcriptional Abundance for Hypercholesterolemia Genes. Genes (Basel) 2025; 16:84. [PMID: 39858631 PMCID: PMC11764661 DOI: 10.3390/genes16010084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Low-density lipoprotein cholesterol (LDL-C) is a well-established risk factor for cardiovascular disease, and it plays a causal role in the development of atherosclerosis. Genome-wide association studies (GWASs) have successfully identified hundreds of genetic variants associated with LDL-C. Most of these risk loci fall in non-coding regions of the genome, and it is unclear how these non-coding variants affect circulating lipid levels. One hypothesis is that genetically mediated variation in transcript abundance, detected via the analysis of expressed quantitative trait loci (eQTLs), is key to the biologic function of causal variants. Here, we investigate the hypothesis that non-coding GWAS risk variants affect the homeostatic expression of a nearby putatively causal gene for serum LDL-C levels. Methods: We establish a set of twenty-one expert-curated and validated genes implicated in hypercholesterolemia via dose-dependent pharmacologic modulation in human adults, for which the relevant tissue type has been established. We show that the expression of these LDL-C genes is impacted by eQTLs in relevant tissues and that there are significant genomic-risk loci in LDL-GWAS near these causal genes. We evaluate, using statistical colocalization, whether a single variant or set of variants in each genetic locus is responsible for the GWAS and eQTL signals. Results: Genome-wide association study results for serum LDL-C levels demonstrate that the 402 identified genomic-risk loci for LDL-C are highly enriched for known causal genes for LDL-C (OR 527, 95% CI 126-5376, p < 2.2 × 10-16). However, we find limited evidence for colocalization between GWAS signals near validated hypercholesterolemia genes and eQTLs in relevant tissues (colocalization rate of 26% at a locus-level colocalization probability > 50%). Conclusions: Our results highlight the complexity of genetic regulatory effects for causal hypercholesterolemia genes; we suggest that context-responsive eQTLs may explain the effects of non-coding GWAS hits that do not overlap with standard eQTLs.
Collapse
Affiliation(s)
- Aaron Hakim
- Division of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.H.); (G.R.S.)
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA;
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; (N.J.C.); (S.R.S.)
| | - Noah J. Connally
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; (N.J.C.); (S.R.S.)
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02215, USA
| | - Gavin R. Schnitzler
- Division of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.H.); (G.R.S.)
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; (N.J.C.); (S.R.S.)
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA;
| | - Z. Gordon Jiang
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Shamil R. Sunyaev
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; (N.J.C.); (S.R.S.)
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02215, USA
| | - Rajat M. Gupta
- Division of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (A.H.); (G.R.S.)
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; (N.J.C.); (S.R.S.)
| |
Collapse
|
8
|
Ansari A, Yadav PK, Zhou L, Prakash B, Ijaz L, Christiano A, Ahmad S, Rimbert A, Hussain MM. Casz1 and Znf101/Zfp961 differentially regulate apolipoproteins A1 and B, alter plasma lipoproteins, and reduce atherosclerosis. JCI Insight 2025; 10:e182260. [PMID: 39782688 PMCID: PMC11721306 DOI: 10.1172/jci.insight.182260] [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: 04/19/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
High apolipoprotein B-containing (apoB-containing) low-density lipoproteins (LDLs) and low apoA1-containing high-density lipoproteins (HDLs) are associated with atherosclerotic cardiovascular diseases. In search of a molecular regulator that could simultaneously and reciprocally control both LDL and HDL levels, we screened a microRNA (miR) library using human hepatoma Huh-7 cells. We identified miR-541-3p that both significantly decreases apoB and increases apoA1 expression by inducing mRNA degradation of 2 different transcription factors, Znf101 and Casz1. We found that Znf101 enhances apoB expression, while Casz1 represses apoA1 expression. The hepatic knockdown of Casz1 in mice increased plasma apoA1, HDL, and cholesterol efflux capacity. The hepatic knockdown of Zfp961, an ortholog of Znf101, reduced lipogenesis and production of triglyceride-rich lipoproteins and atherosclerosis, without causing hepatic lipid accumulation. This study identifies hepatic Znf101/Zfp961 and Casz1 as potential therapeutic targets to alter plasma lipoproteins and reduce atherosclerosis without causing liver steatosis.
Collapse
Affiliation(s)
- Abulaish Ansari
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Pradeep Kumar Yadav
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Liye Zhou
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Binu Prakash
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Laraib Ijaz
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Amanda Christiano
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Sameer Ahmad
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l’institut du thorax, F-44000 Nantes, France
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
- VA New York Harbor Healthcare System, Brooklyn, New York, USA
| |
Collapse
|
9
|
Munkhsaikhan U, Ait-Aissa K, Sahyoun AM, Apu EH, Abidi AH, Kassan A, Kassan M. Lomitapide: navigating cardiovascular challenges with innovative therapies. Mol Biol Rep 2024; 51:1082. [PMID: 39432146 DOI: 10.1007/s11033-024-10003-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
Dyslipidemia is the most significant risk factor for cardiovascular diseases (CVDs) Secondary dyslipidemia: its treatments and association with atherosclerosis. Glob Health Med, Efficacy and safety of saroglitazar for the management of dyslipidemia: A systematic review and meta-analysis of interventional studies. The current treatment strategies for managing dyslipidemia focus on reducing low-density lipoprotein cholesterol (LDL-C) to minimize the risks of atherosclerosis and myocardial infarction (MI). Homozygous Familial Hypercholesterolemia (HoFH) is an inherited autosomal dominant disease caused by a mutation in the LDL receptor (LDLr), which can lead to extremely high levels of LDL-C The Beneficial Effect of Lomitapide on the Cardiovascular System in LDLr(-/-) Mice with Obesity, The microsomal triglyceride transfer protein inhibitor lomitapide improves vascular function in mice with obesity. Although statin therapy has been the primary treatment for dyslipidemia, HoFH patients do not respond well to statins, requiring alternative therapies. Microsomal triglyceride transfer protein (MTP) inhibition has emerged as a potential therapeutic target for treating HoFH. MTP is primarily responsible for transferring triglyceride and other lipids into apolipoprotein B (ApoB) during the assembly of very low-density lipoprotein (VLDL) particles in the liver. Lomitapide, an inhibitor of MTP, has been approved for treatingof HoFH adults. Unlike statins, lomitapide does not act on the LDLr to reduce cholesterol. Instead, lomitapide lowers the levels of ApoB-containing proteins, primarily VLDL, eventually decreasing LDL-C levels. Studies have shown that lomitapide can reduce LDL-C levels by more than 50% in patients with HoFH who have failed to respond adequately to other treatments. Lowering LDL-C levels is important for preventing atherosclerosis, reducing cardiovascular risk, improving endothelial function, and promoting overall cardiovascular health, especially for patients with HoFH Efficacy and safety of a microsomal triglyceride transfer protein inhibitor in patients with homozygous familial hypercholesterolaemia: a single-arm, open-label, phase 3 study. This review paper focuses on research findings regarding the therapeutic benefits of lomitapide, highlighting its effectiveness in lowering cholesterol levels and reducing the risk of CVDs The microsomal triglyceride transfer protein inhibitor lomitapide improves vascular function in mice with obesity.
Collapse
Affiliation(s)
- Undral Munkhsaikhan
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Karima Ait-Aissa
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Amal M Sahyoun
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Ehsanul Hoque Apu
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Ammaar H Abidi
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Adam Kassan
- School of Pharmacy, West Coast University, 590 N. Vermont Ave, Los Angeles, CA, 90004, USA.
| | - Modar Kassan
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA.
| |
Collapse
|
10
|
Ansari A, Yadav PK, Valmiki S, Laine A, Rimbert A, Islam S, Osman I, Najafi-Shoushtari SH, Hussain MM. MicroRNA-615-3p decreases apo B expression in human liver cells. J Lipid Res 2024; 65:100659. [PMID: 39332527 PMCID: PMC11513542 DOI: 10.1016/j.jlr.2024.100659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024] Open
Abstract
Plasma lipids are mainly carried in apolipoprotein B (apoB) containing lipoproteins. High levels of these lipoproteins are associated with several metabolic diseases and lowering their plasma levels is associated with reduced incidence of atherosclerotic cardiovascular disease. MicroRNAs (miRs) are small non-coding RNAs that reduce the protein expression of their target mRNAs and are potential therapeutic agents. Here, we identified a novel miR-615-3p that interacts with human 3'-UTR of apoB mRNA, induces post-transcriptional mRNA degradation, and reduces cellular and secreted apoB100 in human hepatoma Huh-7 cells. Reducing cellular miR-615-3p levels by CRISPR-sgRNA increased cellular and secreted apoB100 indicating endogenous miR regulates apoB expression. Overexpression of miR-615-3p along with or without palmitic acid treatment decreased cellular and media apoB and increased cellular triglyceride levels without inducing endoplasmic reticulum stress. These studies have identified miR-615-3p as a negative regulator of apoB expression in human liver-derived cells. It is likely that there are more miRs that regulate apoB-containing lipoprotein assembly and secretion. Discovery of additional miRs may uncover novel mechanisms that control lipoprotein assembly and secretion.
Collapse
Affiliation(s)
- Abulaish Ansari
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA; Department of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Pradeep Kumar Yadav
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA; Department of Botany, Faculty of Science, University of Allahabad, Prayagraj, India
| | - Swati Valmiki
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Antoine Laine
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Shahidul Islam
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Iman Osman
- Department of Medicine, New York University Langone Medical Center, New York, NY, USA
| | - S Hani Najafi-Shoushtari
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA; Department of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - M Mahmood Hussain
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA; Research Department, New York Harbor Healthcare System, Brooklyn, NY, USA.
| |
Collapse
|
11
|
Meurs A, Ndoj K, van den Berg M, Marinković G, Tantucci M, Veenendaal T, Kuivenhoven JA, Klumperman J, Zelcer N. A suite of genome-engineered hepatic cells provides novel insights into the spatiotemporal metabolism of apolipoprotein B and apolipoprotein B-containing lipoprotein secretion. Cardiovasc Res 2024; 120:1253-1264. [PMID: 38833612 PMCID: PMC11416059 DOI: 10.1093/cvr/cvae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/29/2024] [Accepted: 04/18/2024] [Indexed: 06/06/2024] Open
Abstract
AIMS Apolipoprotein B (APOB)-containing very LDL (VLDL) production, secretion, and clearance by hepatocytes is a central determinant of hepatic and circulating lipid levels. Impairment of any of the aforementioned processes is associated with the development of multiple diseases. Despite the discovery of genes and processes that govern hepatic VLDL metabolism, our understanding of the different mechanistic steps involved is far from complete. An impediment to these studies is the lack of tractable hepatocyte-based systems to interrogate and follow APOB in cells, which the current study addresses. METHODS AND RESULTS To facilitate the cellular study of VLDL metabolism, we generated human hepatic HepG2 and Huh-7 cell lines in which CRISPR/Cas9-based genome engineering was used to introduce the fluorescent protein mNeonGreen into the APOB gene locus. This results in the production of APOB100-mNeon that localizes predominantly to the endoplasmic reticulum (ER) and Golgi by immunofluorescence and electron microscopy imaging. The production and secretion of APOB100-mNeon can be quantitatively followed in medium over time and results in the production of lipoproteins that are taken up via the LDL receptor pathway. Importantly, the production and secretion of APOB-mNeon is sensitive to established pharmacological and physiological treatments and to genetic modifiers known to influence VLDL production in humans. As a showcase, we used HepG2-APOBmNeon cells to interrogate ER-associated degradation of APOB. The use of a dedicated sgRNA library targeting all established membrane-associated ER-resident E3 ubiquitin ligases led to the identification of SYNV1 as the E3 responsible for the degradation of poorly lipidated APOB in HepG2 cells. CONCLUSIONS In summary, the engineered cells reported here allow the study of hepatic VLDL assembly and secretion and facilitate spatiotemporal interrogation induced by pharmacologic and genetic perturbations.
Collapse
Affiliation(s)
- Amber Meurs
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism and Amsterdam Cardiovascular Sciences, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Klevis Ndoj
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism and Amsterdam Cardiovascular Sciences, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Marlene van den Berg
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism and Amsterdam Cardiovascular Sciences, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Goran Marinković
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism and Amsterdam Cardiovascular Sciences, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Matteo Tantucci
- Center for Molecular Medicine—Cell Biology, University Medical Center Utrecht, University of Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Tineke Veenendaal
- Center for Molecular Medicine—Cell Biology, University Medical Center Utrecht, University of Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Judith Klumperman
- Center for Molecular Medicine—Cell Biology, University Medical Center Utrecht, University of Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Noam Zelcer
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism and Amsterdam Cardiovascular Sciences, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| |
Collapse
|
12
|
Wu HT, Wu BX, Fang ZX, Wu Z, Hou YY, Deng Y, Cui YK, Liu J. Lomitapide repurposing for treatment of malignancies: A promising direction. Heliyon 2024; 10:e32998. [PMID: 38988566 PMCID: PMC11234027 DOI: 10.1016/j.heliyon.2024.e32998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
The development of novel drugs from basic science to clinical practice requires several years, much effort, and cost. Drug repurposing can promote the utilization of clinical drugs in cancer therapy. Recent studies have shown the potential effects of lomitapide on treating malignancies, which is currently used for the treatment of familial hypercholesterolemia. We systematically review possible functions and mechanisms of lomitapide as an anti-tumor compound, regarding the aspects of apoptosis, autophagy, and metabolism of tumor cells, to support repurposing lomitapide for the clinical treatment of tumors.
Collapse
Affiliation(s)
- Hua-Tao Wu
- Department of General Surgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Bing-Xuan Wu
- Department of General Surgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Ze-Xuan Fang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
- Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Zheng Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
- Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Yan-Yu Hou
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
- Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Yu Deng
- Department of General Surgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yu-Kun Cui
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Jing Liu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
- Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou, 515041, China
| |
Collapse
|
13
|
Caña-Bozada VH, Ovando-Vázquez C, Flores-Méndez LC, Martínez-Brown JM, Morales-Serna FN. Identifying potential drug targets in the kinomes of two monogenean species. Helminthologia 2024; 61:142-150. [PMID: 39040804 PMCID: PMC11260314 DOI: 10.2478/helm-2024-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 05/24/2024] [Indexed: 07/24/2024] Open
Abstract
Protein kinases are enzymes involved in essential biological processes such as signal transduction, transcription, metabolism, and the cell cycle. Human kinases are targets for several drugs approved by the US Food and Drug Administration. Therefore, the identification and classification of kinases in other organisms, including pathogenic parasites, is an interesting subject of study. Monogeneans are platyhelminths, mainly ectoparasites, capable of causing health problems in farmed fish. Although some genomes and transcriptomes are available for monogenean species, their full repertoire of kinases is unknown. The aim of this study was to identify and classify the putative kinases in the transcriptomes of two monogeneans, Rhabdosynochus viridisi and Scutogyrus longicornis, and then to predict potential monogenean drug targets (MDTs) and selective inhibitor drugs using computational approaches. Monogenean kinases having orthologs in the lethal phenotype of C. elegans but not in fish or humans were considered MDTs. A total of 160 and 193 kinases were identified in R. viridisi and S. longicornis, respectively. Of these, 22 kinases, belonging mainly to the major groups CAMK, AGC, and TK, were classified as MDTs, five of which were evaluated further. Molecular docking analysis indicated that dihydroergotamine, ergotamine, and lomitapide have the highest affinity for the kinases BRSK and MEKK1. These well-known drugs could be evaluated in future studies for potential repurposing as anti-monogenean agents. The present study contributes valuable data for the development of new antiparasitic candidates for finfish aquaculture.
Collapse
Affiliation(s)
- V. H. Caña-Bozada
- Centro de Investigación en Alimentación y Desarrollo, A.C., Mazatlán, Sinaloa82112, Mexico
| | - C. Ovando-Vázquez
- Centro Nacional de Supercómputo, Instituto Potosino de investigación Científica y Tecnológica, San Luis Potosí78216, Mexico
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Ciudad de México, Mexico
| | - L. C. Flores-Méndez
- Centro de Investigación en Alimentación y Desarrollo, A.C., Mazatlán, Sinaloa82112, Mexico
- Present address:Universidad Autónoma de Occidente, Unidad Regional Mazatlán, Mazatlán, 82100, Sinaloa, Mexico
| | - J. M. Martínez-Brown
- Centro de Investigación en Alimentación y Desarrollo, A.C., Mazatlán, Sinaloa82112, Mexico
| | - F. N. Morales-Serna
- Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Mazatlán82040, Sinaloa, Mexico
| |
Collapse
|
14
|
Sharma A, Sharma C, Sharma L, Wal P, Mishra P, Sachdeva N, Yadav S, Vargas De-La Cruz C, Arora S, Subramaniyan V, Rawat R, Behl T, Nandave M. Targeting the vivid facets of apolipoproteins as a cardiovascular risk factor in rheumatoid arthritis. Can J Physiol Pharmacol 2024; 102:305-317. [PMID: 38334084 DOI: 10.1139/cjpp-2023-0259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Mostly, cardiovascular diseases are blamed for casualties in rheumatoid arthritis (RA) patients. Customarily, dyslipidemia is probably the most prevalent underlying cause of untimely demise in people suffering from RA as it hastens the expansion of atherosclerosis. The engagement of inflammatory cytokines like tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6), etc., is crucial in the progression and proliferation of both RA and abnormal lipid parameters. Thus, lipid abnormalities should be monitored frequently in patients with both primary and advanced RA stages. An advanced lipid profile examination, i.e., direct role of apolipoproteins associated with various lipid molecules is a more dependable approach for better understanding of the disease and selecting suitable therapeutic targets. Therefore, studying their apolipoproteins is more relevant than assessing RA patients' altered lipid profile levels. Among the various apolipoprotein classes, Apo A1 and Apo B are primarily being focused. In addition, it also addresses how calculating Apo B:Apo A1 ratio can aid in analyzing the disease's risk. The marketed therapies available to control lipid abnormalities are associated with many other risk factors. Hence, directly targeting Apo A1 and Apo B would provide a better and safer option.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Chakshu Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology, Pharmacy, Kanpur, Uttar Pradesh, India
| | - Preeti Mishra
- Raja Balwant Singh Engineering Technical Campus, Bichpuri, Agra, India
| | - Nitin Sachdeva
- Department of Anesthesia, Mediclinic Aljowhara Hospital, Al Ain, United Arab Emirates
| | - Shivam Yadav
- School of Pharmacy, Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
| | - Celia Vargas De-La Cruz
- Department of Pharmacology, Bromatology and Toxicology, Faculty of Pharmacy and Biochemistry, Universidad Nacional Mayor de San Marcos, Lima 15001, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima 15001, Peru
| | - Sandeep Arora
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
- Centre for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 600077, India
| | - Ravi Rawat
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, Uttarakhand, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Delhi, India
| |
Collapse
|
15
|
Kobayashi J, Minamizuka T, Tada H, Yokote K. Familial hypercholesterolemia with special focus on Japan. Clin Chim Acta 2024; 556:117847. [PMID: 38417778 DOI: 10.1016/j.cca.2024.117847] [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: 01/23/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Familial hypercholesterolemia (FH) is an inherited disorder characterized by increased low-density lipoprotein LDL) cholesterol and atherosclerotic cardiovascular disease. Although initial genetic analysis linked FH to LDL receptor mutations, subsequent work demonstrated that a gain-of-function mutation in the proprotein convertase subtilisin/kexin type 9 (PCSK9), which causes LDL-R degradation, was shown to be the cause of FH. In this review, we describe the history of research on FH, its clinical phenotyping and genotyping and advances in treatment with special focus on Japan.
Collapse
Affiliation(s)
- Junji Kobayashi
- Department of Endocrinology, Metabolism, Hematology and Geriatrics, Chiba University; Department of Clinical Laboratory Science, Graduate School of Medical Sciences, Kanazawa University.
| | - Takuya Minamizuka
- Department of Endocrinology, Metabolism, Hematology and Geriatrics, Chiba University
| | - Hayato Tada
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University
| | - Koutaro Yokote
- Department of Endocrinology, Metabolism, Hematology and Geriatrics, Chiba University
| |
Collapse
|
16
|
Zhang Z, Yang R, Zhu J, Yang X, Luo H, Wang H, Luo X. Failure of lipid control by PCSK9 inhibitors in compound heterozygous familial hypercholesterolemia complicated with premature myocardial infarction: A case report. Clin Case Rep 2024; 12:e8498. [PMID: 38487640 PMCID: PMC10939999 DOI: 10.1002/ccr3.8498] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/26/2023] [Accepted: 01/16/2024] [Indexed: 03/17/2024] Open
Abstract
Key Clinical Message A certain level of low-density lipoprotein receptor activity is crucial for the efficacy of PCSK9i. Therapeutic strategies for familial hypercholesterolemia patients should consider drug efficacy, and genetic testing will be helpful. Abstract Familial hypercholesterolemia (FH) is a serious autosomal dominant disorder. Managing blood lipids in FH patients poses greater challenges for clinicians. Drug therapy may not always yield satisfactory results, particularly in individuals with low-density lipoprotein receptor (LDLR) negative mutations. Herein, we report a young female harboring an LDLR frameshift mutation. This patient developed xanthomas at 7 months old and underwent several years of treatment involving four classes of lipid-lowering drugs, including PCSK9i. However, the response to drug therapy was limited in this patient and eventually culminated in premature myocardial infarction. The efficacy of PCSK9i depends on the activity of LDLR. The inefficacy of PCSK9i may arise from the extensive mutations which leading to loss of LDLR activity. Therapy plans for these patients should take into account the efficacy of drug therapy. Early genetic testing is crucial for clinicians to make informed decisions regarding therapy options.
Collapse
Affiliation(s)
- Ziyue Zhang
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
- 96608 Hospital of PLAHan ZhongShanxiP. R. China
| | - Rongpei Yang
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - Jun Zhu
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - XiaoLi Yang
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - Hao Luo
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - Hongyong Wang
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - Xiaoli Luo
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| |
Collapse
|
17
|
Anaganti N, Valmiki S, Recacha R, Islam S, Farber S, Ruddock L, Hussain MM. Bulky hydrophobic side chains in the β1-sandwich of microsomal triglyceride transfer protein are critical for the transfer of both triglycerides and phospholipids. J Biol Chem 2024; 300:105726. [PMID: 38325741 PMCID: PMC10907164 DOI: 10.1016/j.jbc.2024.105726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 02/09/2024] Open
Abstract
Hyperlipidemia predisposes individuals to cardiometabolic diseases, the most common cause of global mortality. Microsomal triglyceride transfer protein (MTP) transfers multiple lipids and is essential for the assembly of apolipoprotein B-containing lipoproteins. MTP inhibition lowers plasma lipids but causes lipid retention in the liver and intestine. Previous studies suggested two lipid transfer domains in MTP and that specific inhibition of triglyceride (TG) and not phospholipid (PL) transfer can lower plasma lipids without significant tissue lipid accumulation. However, how MTP transfers different lipids and the domains involved in these activities are unknown. Here, we tested a hypothesis that two different β-sandwich domains in MTP transfer TG and PL. Mutagenesis of charged amino acids in β2-sandwich had no effect on PL transfer activity indicating that they are not critical. In contrast, amino acids with bulky hydrophobic side chains in β1-sandwich were critical for both TG and PL transfer activities. Substitutions of these residues with smaller hydrophobic side chains or positive charges reduced, whereas negatively charged side chains severely attenuated MTP lipid transfer activities. These studies point to a common lipid transfer domain for TG and PL in MTP that is enriched with bulky hydrophobic amino acids. Furthermore, we observed a strong correlation in different MTP mutants with respect to loss of both the lipid transfer activities, again implicating a common binding site for TG and PL in MTP. We propose that targeting of areas other than the identified common lipid transfer domain might reduce plasma lipids without causing cellular lipid retention.
Collapse
Affiliation(s)
- Narasimha Anaganti
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Swati Valmiki
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Rosario Recacha
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Shahidul Islam
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Steven Farber
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lloyd Ruddock
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - M Mahmood Hussain
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA.
| |
Collapse
|
18
|
van Zwol W, van de Sluis B, Ginsberg HN, Kuivenhoven JA. VLDL Biogenesis and Secretion: It Takes a Village. Circ Res 2024; 134:226-244. [PMID: 38236950 PMCID: PMC11284300 DOI: 10.1161/circresaha.123.323284] [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: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 01/23/2024]
Abstract
The production and secretion of VLDLs (very-low-density lipoproteins) by hepatocytes has a direct impact on liver fat content, as well as the concentrations of cholesterol and triglycerides in the circulation and thus affects both liver and cardiovascular health, respectively. Importantly, insulin resistance, excess caloric intake, and lack of physical activity are associated with overproduction of VLDL, hepatic steatosis, and increased plasma levels of atherogenic lipoproteins. Cholesterol and triglycerides in remnant particles generated by VLDL lipolysis are risk factors for atherosclerotic cardiovascular disease and have garnered increasing attention over the last few decades. Presently, however, increased risk of atherosclerosis is not the only concern when considering today's cardiometabolic patients, as they often also experience hepatic steatosis, a prevalent disorder that can progress to steatohepatitis and cirrhosis. This duality of metabolic risk highlights the importance of understanding the molecular regulation of the biogenesis of VLDL, the lipoprotein that transports triglycerides and cholesterol out of the liver. Fortunately, there has been a resurgence of interest in the intracellular assembly, trafficking, degradation, and secretion of VLDL by hepatocytes, which has led to many exciting new molecular insights that are the topic of this review. Increasing our understanding of the biology of this pathway will aid to the identification of novel therapeutic targets to improve both the cardiovascular and the hepatic health of cardiometabolic patients. This review focuses, for the first time, on this duality.
Collapse
Affiliation(s)
- Willemien van Zwol
- Department of Paediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bart van de Sluis
- Department of Paediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henry. N. Ginsberg
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Jan Albert Kuivenhoven
- Department of Paediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
19
|
Ai JY, Zhao PC, Zhang W, Rao GW. Research Progress in the Clinical Treatment of Familial Hypercholesterolemia. Curr Med Chem 2024; 31:1082-1106. [PMID: 36733200 DOI: 10.2174/0929867330666230202111849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/30/2022] [Accepted: 12/15/2022] [Indexed: 02/04/2023]
Abstract
Familial hypercholesterolemia (FH) is an autosomal dominant inheritable disease with severe disorders of lipid metabolism. It is mainly marked by increasing levels of plasma total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C), xanthoma, corneal arch, and early-onset coronary heart disease (CHD). The prevalence of FH is high, and it is dangerous and clinically underdiagnosed. The clinical treatment for FH includes both pharmacological and non-pharmacological treatment, of which non-pharmacological treatment mainly includes therapeutic lifestyle change and dietary therapy, LDL apheresis, liver transplantation and gene therapy. In recent years, many novel drugs have been developed to treat FH more effectively. In addition, the continuous maturity of non-pharmacological treatment techniques has also brought more hope for the treatment of FH. This paper analyzes the pathogenic mechanism and the progress in clinical treatment of FH. Furthermore, it also summarizes the mechanism and structure-activity relationship of FH therapeutic drugs that have been marketed. In a word, this article provides a reference value for the research and development of FH therapeutic drugs.
Collapse
Affiliation(s)
- Jing-Yan Ai
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, P.R. China
| | - Peng-Cheng Zhao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, P.R. China
| | - Wen Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, P.R. China
| | - Guo-Wu Rao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, P.R. China
| |
Collapse
|
20
|
Danilov A, Frishman WH, Aronow WS. Antihyperlipidemic Treatment Options in Statin Resistance and Intolerance. Cardiol Rev 2024; 32:51-56. [PMID: 36305712 DOI: 10.1097/crd.0000000000000498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cardiovascular disease is the global leading cause of death and hypercholesterolemia is implicated as one of its top contributors. Moreover, there is growing recognition that lower low-density lipoprotein cholesterol levels offer greater protection against cardiovascular disease. Statins are the first-line lipid-lowering agents for both primary and secondary prevention of cardiovascular disease in patients with hypercholesterolemia. However, statin resistance and intolerance lead to undertreatment in patients who would likely derive the most benefit from antihyperlipidemic drugs. Several non-statin therapies are increasingly prescribed to such patients, most commonly ezetimibe and the PCSK9 monoclonal antibodies, but numerous other options have been developed in recent years and investigations into new therapies are ongoing. The use of these non-statin therapies requires the clinician to take a highly personalized approach to cholesterol reduction in complex patients. In this review, we describe current non-statin options for statin-resistant and statin-intolerant patients in addition to areas of active research.
Collapse
Affiliation(s)
| | - William H Frishman
- Department of Medicine, New York Medical College, and Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Wilbert S Aronow
- Department of Medicine, New York Medical College, and Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
21
|
Sachan V, Le Dévéhat M, Roubtsova A, Essalmani R, Laurendeau JF, Garçon D, Susan-Resiga D, Duval S, Mikaeeli S, Hamelin J, Evagelidis A, Chong M, Paré G, Chernetsova E, Gao ZH, Robillard I, Ruiz M, Trinh VQH, Estall JL, Faraj M, Austin RC, Sauvageau M, Prat A, Kiss RS, Seidah NG. PCSK7: A novel regulator of apolipoprotein B and a potential target against non-alcoholic fatty liver disease. Metabolism 2024; 150:155736. [PMID: 37967646 DOI: 10.1016/j.metabol.2023.155736] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Epidemiological evidence links the proprotein convertase subtilisin/kexin 7 (PCSK7) to triglyceride (TG) metabolism. We associated the known PCSK7 gain-of-function non-coding SNP rs236918 with higher levels of plasma apolipoprotein B (apoB) and the loss-of-function coding variant p.Pro777Leu (SNP rs201598301) with lower apoB and TG. Herein, we aimed to unravel the in vivo role of liver PCSK7. METHODS We biochemically defined the functional role of PCSK7 in lipid metabolism using hepatic cell lines and Pcsk7-/- mice. Our findings were validated following subcutaneous administration of hepatocyte-targeted N-acetylgalactosamine (GalNAc)-antisense oligonucleotides (ASOs) against Pcsk7. RESULTS Independent of its proteolytic activity, membrane-bound PCSK7 binds apoB100 in the endoplasmic reticulum and enhances its secretion. Mechanistically, the loss of PCSK7/Pcsk7 leads to apoB100 degradation, triggering an unfolded protein response, autophagy, and β-oxidation, eventually reducing lipid accumulation in hepatocytes. Non-alcoholic fatty liver disease (NAFLD) was induced by a 12-week high fat/fructose/cholesterol diet in wild type (WT) and Pcsk7-/- mice that were then allowed to recover on a 4-week control diet. Pcsk7-/- mice recovered more effectively than WT mice from all NAFLD-related liver phenotypes. Finally, subcutaneous administration of GalNAc-ASOs targeting hepatic Pcsk7 to WT mice validated the above results. CONCLUSIONS Our data reveal hepatic PCSK7 as one of the major regulators of apoB, and its absence reduces apoB secretion from hepatocytes favoring its ubiquitination and degradation by the proteasome. This results in a cascade of events, eventually reducing hepatic lipid accumulation, thus supporting the notion of silencing PCSK7 mRNA in hepatocytes for targeting NAFLD.
Collapse
Affiliation(s)
- Vatsal Sachan
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Maïlys Le Dévéhat
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Anna Roubtsova
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Rachid Essalmani
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Jean-Francois Laurendeau
- RNA and Noncoding Mechanisms of Disease, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Damien Garçon
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Delia Susan-Resiga
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Stéphanie Duval
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Sahar Mikaeeli
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Josée Hamelin
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Alexandra Evagelidis
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Michael Chong
- Department of Biochemistry & Biomedical Sciences, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Department of Biochemistry & Biomedical Sciences, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | | | - Zu-Hua Gao
- Department of Pathology, McGill University Health Centre, Montréal, QC, Canada
| | - Isabelle Robillard
- Montreal Heart Institute, Metabolomics Platform, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| | - Matthieu Ruiz
- Montreal Heart Institute, Metabolomics Platform, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| | - Vincent Quoc-Huy Trinh
- Departement of Pathology and Cellular Biology, Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, QC, Canada
| | - Jennifer L Estall
- Molecular Mechanisms of Diabetes, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - May Faraj
- Nutrition Department, Université de Montréal, Research Unit on Nutrition, Lipoproteins and Cardiometabolic Diseases, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Center for Kidney Research, Hamilton, ON, Canada
| | - Martin Sauvageau
- RNA and Noncoding Mechanisms of Disease, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Annik Prat
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada
| | - Robert S Kiss
- McGill University Health Centre Research Institute, Montréal, QC, Canada
| | - Nabil G Seidah
- Biochemical Neuroendocrinology, Institut de Recherches Cliniques de Montréal (IRCM), affiliated to the Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
22
|
Gogate A, Belcourt J, Shah M, Wang AZ, Frankel A, Kolmel H, Chalon M, Stephen P, Kolli A, Tawfik SM, Jin J, Bahal R, Rasmussen TP, Manautou JE, Zhong XB. Targeting the Liver with Nucleic Acid Therapeutics for the Treatment of Systemic Diseases of Liver Origin. Pharmacol Rev 2023; 76:49-89. [PMID: 37696583 PMCID: PMC10753797 DOI: 10.1124/pharmrev.123.000815] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
Systemic diseases of liver origin (SDLO) are complex diseases in multiple organ systems, such as cardiovascular, musculoskeletal, endocrine, renal, respiratory, and sensory organ systems, caused by irregular liver metabolism and production of functional factors. Examples of such diseases discussed in this article include primary hyperoxaluria, familial hypercholesterolemia, acute hepatic porphyria, hereditary transthyretin amyloidosis, hemophilia, atherosclerotic cardiovascular diseases, α-1 antitrypsin deficiency-associated liver disease, and complement-mediated diseases. Nucleic acid therapeutics use nucleic acids and related compounds as therapeutic agents to alter gene expression for therapeutic purposes. The two most promising, fastest-growing classes of nucleic acid therapeutics are antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs). For each listed SDLO disease, this article discusses epidemiology, symptoms, genetic causes, current treatment options, and advantages and disadvantages of nucleic acid therapeutics by either ASO or siRNA drugs approved or under development. Furthermore, challenges and future perspectives on adverse drug reactions and toxicity of ASO and siRNA drugs for the treatment of SDLO diseases are also discussed. In summary, this review article will highlight the clinical advantages of nucleic acid therapeutics in targeting the liver for the treatment of SDLO diseases. SIGNIFICANCE STATEMENT: Systemic diseases of liver origin (SDLO) contain rare and common complex diseases caused by irregular functions of the liver. Nucleic acid therapeutics have shown promising clinical advantages to treat SDLO. This article aims to provide the most updated information on targeting the liver with antisense oligonucleotides and small interfering RNA drugs. The generated knowledge may stimulate further investigations in this growing field of new therapeutic entities for the treatment of SDLO, which currently have no or limited options for treatment.
Collapse
Affiliation(s)
- Anagha Gogate
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Jordyn Belcourt
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Milan Shah
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Alicia Zongxun Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Alexis Frankel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Holly Kolmel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Matthew Chalon
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Prajith Stephen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Aarush Kolli
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Sherouk M Tawfik
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Jing Jin
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Raman Bahal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Theodore P Rasmussen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - José E Manautou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Xiao-Bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
23
|
Kim HI, Park J. Natural Antioxidants in Obesity and Related Diseases. Antioxidants (Basel) 2023; 12:1966. [PMID: 38001819 PMCID: PMC10669253 DOI: 10.3390/antiox12111966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Obesity is a chronic complex disease defined by excessive adiposity that impairs health [...].
Collapse
Affiliation(s)
- Hyo In Kim
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
24
|
Hendrix S, Kingma J, Ottenhoff R, Valiloo M, Svecla M, Zijlstra LF, Sachdev V, Kovac K, Levels JHM, Jongejan A, de Boer JF, Kuipers F, Rimbert A, Norata GD, Loregger A, Zelcer N. Hepatic SREBP signaling requires SPRING to govern systemic lipid metabolism in mice and humans. Nat Commun 2023; 14:5181. [PMID: 37626055 PMCID: PMC10457316 DOI: 10.1038/s41467-023-40943-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The sterol regulatory element binding proteins (SREBPs) are transcription factors that govern cholesterol and fatty acid metabolism. We recently identified SPRING as a post-transcriptional regulator of SREBP activation. Constitutive or inducible global ablation of Spring in mice is not tolerated, and we therefore develop liver-specific Spring knockout mice (LKO). Transcriptomics and proteomics analysis reveal attenuated SREBP signaling in livers and hepatocytes of LKO mice. Total plasma cholesterol is reduced in male and female LKO mice in both the low-density lipoprotein and high-density lipoprotein fractions, while triglycerides are unaffected. Loss of Spring decreases hepatic cholesterol and triglyceride content due to diminished biosynthesis, which coincides with reduced very-low-density lipoprotein secretion. Accordingly, LKO mice are protected from fructose diet-induced hepatosteatosis. In humans, we find common genetic SPRING variants that associate with circulating high-density lipoprotein cholesterol and ApoA1 levels. This study positions SPRING as a core component of hepatic SREBP signaling and systemic lipid metabolism in mice and humans.
Collapse
Affiliation(s)
- Sebastian Hendrix
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Jenina Kingma
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Roelof Ottenhoff
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Masoud Valiloo
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Monika Svecla
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Lobke F Zijlstra
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Vinay Sachdev
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Kristina Kovac
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Johannes H M Levels
- Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Aldo Jongejan
- Department of Epidemiology and Data Science, Bioinformatics Laboratory, of Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Jan F de Boer
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Antoine Rimbert
- l'institut du thorax, Nantes Université, CNRS, INSERM, F-44000, Nantes, France
| | - Giuseppe D Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Anke Loregger
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
- Myllia Biotechnology GmbH, Am Kanal 27, 1110, Vienna, Austria
| | - Noam Zelcer
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Munkhsaikhan U, Kwon YI, Sahyoun AM, Galán M, Gonzalez AA, Ait-Aissa K, Abidi AH, Kassan A, Kassan M. The Beneficial Effect of Lomitapide on the Cardiovascular System in LDLr -/- Mice with Obesity. Antioxidants (Basel) 2023; 12:1287. [PMID: 37372017 PMCID: PMC10295391 DOI: 10.3390/antiox12061287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/26/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
OBJECTIVES Homozygous familial hypercholesteremia (HoFH) is a rare, life-threatening metabolic disease, mainly caused by a mutation in the LDL receptor. If untreated, HoFH causes premature death from acute coronary syndrome. Lomitapide is approved by the FDA as a therapy to lower lipid levels in adult patients with HoFH. Nevertheless, the beneficial effect of lomitapide in HoFH models remains to be defined. In this study, we investigated the effect of lomitapide on cardiovascular function using LDL receptor-knockout mice (LDLr-/-). METHODS Six-week-old LDLr-/- mice were fed a standard diet (SD) or a high-fat diet (HFD) for 12 weeks. Lomitapide (1 mg/Kg/Day) was given by oral gavage for the last 2 weeks in the HFD group. Body weight and composition, lipid profile, blood glucose, and atherosclerotic plaques were measured. Vascular reactivity and markers for endothelial function were determined in conductance arteries (thoracic aorta) and resistance arteries (mesenteric resistance arteries (MRA)). Cytokine levels were measured by using the Mesoscale discovery V-Plex assays. RESULTS Body weight (47.5 ± 1.5 vs. 40.3 ± 1.8 g), % of fat mass (41.6 ± 1.9% vs. 31.8 ± 1.7%), blood glucose (215.5 ± 21.9 vs. 142.3 ± 7.7 mg/dL), and lipid levels (cholesterol: 600.9 ± 23.6 vs. 451.7 ± 33.4 mg/dL; LDL/VLDL: 250.6 ± 28.9 vs. 161.1 ± 12.24 mg/dL; TG: 299.5 ± 24.1 vs. 194.1 ± 28.1 mg/dL) were significantly decreased, and the % of lean mass (56.5 ± 1.8% vs. 65.2 ± 2.1%) was significantly increased in the HFD group after lomitapide treatment. The atherosclerotic plaque area also decreased in the thoracic aorta (7.9 ± 0.5% vs. 5.7 ± 0.1%). After treatment with lomitapide, the endothelium function of the thoracic aorta (47.7 ± 6.3% vs. 80.7 ± 3.1%) and mesenteric resistance artery (66.4 ± 4.3% vs. 79.5 ± 4.6%) was improved in the group of LDLr-/- mice on HFD. This was correlated with diminished vascular endoplasmic (ER) reticulum stress, oxidative stress, and inflammation. CONCLUSIONS Treatment with lomitapide improves cardiovascular function and lipid profile and reduces body weight and inflammatory markers in LDLr-/- mice on HFD.
Collapse
Affiliation(s)
- Undral Munkhsaikhan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Bioscience Research and General Dentistry, College of Dentistry, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Young In Kwon
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Amal M. Sahyoun
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Food Science and Agriculture Chemistry, McGill University, Montreal, QC H9X 3V9, Canada
| | - María Galán
- Faculty of Health Sciences, University Rey Juan Carlos, 28922 Alcorcón, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain
| | - Alexis A. Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso 300, Chile
| | - Karima Ait-Aissa
- College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| | - Ammaar H. Abidi
- Department of Bioscience Research and General Dentistry, College of Dentistry, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
- College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| | - Adam Kassan
- Department of Pharmaceutical Sciences, School of Pharmacy, West Coast University, Los Angeles, CA 91606, USA
| | - Modar Kassan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| |
Collapse
|
26
|
Srivastava RAK. A Review of Progress on Targeting LDL Receptor-Dependent and -Independent Pathways for the Treatment of Hypercholesterolemia, a Major Risk Factor of ASCVD. Cells 2023; 12:1648. [PMID: 37371118 DOI: 10.3390/cells12121648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Since the discovery of the LDL receptor in 1973 by Brown and Goldstein as a causative protein in hypercholesterolemia, tremendous amounts of effort have gone into finding ways to manage high LDL cholesterol in familial hypercholesterolemic (HoFH and HeFH) individuals with loss-of-function mutations in the LDL receptor (LDLR) gene. Statins proved to be the first blockbuster drug, helping both HoFH and HeFH individuals by inhibiting the cholesterol synthesis pathway rate-limiting enzyme HMG-CoA reductase and inducing the LDL receptor. However, statins could not achieve the therapeutic goal of LDL. Other therapies targeting LDLR include PCSK9, which lowers LDLR by promoting LDLR degradation. Inducible degrader of LDLR (IDOL) also controls the LDLR protein, but an IDOL-based therapy is yet to be developed. Among the LDLR-independent pathways, such as angiopoietin-like 3 (ANGPTL3), apolipoprotein (apo) B, apoC-III and CETP, only ANGPTL3 offers the advantage of treating both HoFH and HeFH patients and showing relatively better preclinical and clinical efficacy in animal models and hypercholesterolemic individuals, respectively. While loss-of-LDLR-function mutations have been known for decades, gain-of-LDLR-function mutations have recently been identified in some individuals. The new information on gain of LDLR function, together with CRISPR-Cas9 genome/base editing technology to target LDLR and ANGPTL3, offers promise to HoFH and HeFH individuals who are at a higher risk of developing atherosclerotic cardiovascular disease (ASCVD).
Collapse
Affiliation(s)
- Rai Ajit K Srivastava
- Integrated Pharma Solutions LLC, Boston, MA 02101-02117, USA
- College of Professional Studies, Northeastern University, Boston, MA 02101-02117, USA
| |
Collapse
|
27
|
Bassani Borges J, Fernandes Oliveira V, Dagli-Hernandez C, Monteiro Ferreira G, Kristini Almendros Afonso Barbosa T, da Silva Rodrigues Marçal E, Los B, Barbosa Malaquias V, Hernandes Bortolin R, Caroline Costa Freitas R, Akira Mori A, Medeiros Bastos G, Marques Gonçalves R, Branco Araújo D, Zatz H, Bertolami A, Arpad Faludi A, Chiara Bertolami M, Guerra de Moraes Rego Souza A, Ítalo Dias França J, Strelow Thurow H, Dominguez Crespo Hirata T, Takashi Imoto Nakaya H, Elim Jannes C, da Costa Pereira A, Nogueira Silbiger V, Ducati Luchessi A, Nayara Góes Araújo J, Arruda Nakazone M, Silva Carmo T, Rossi Silva Souza D, Moriel P, Yu Ting Wang J, Satya Naslavsky M, Gorjão R, Cristina Pithon-Curi T, Curi R, Moreno Fajardo C, Lin Wang HT, Regina Garófalo A, Cerda A, Ferraz Sampaio M, Dominguez Crespo Hirata R, Hiroyuki Hirata M. Identification of pathogenic variants in the Brazilian cohort with Familial Hypercholesterolemia using exon-targeted gene sequencing. Gene 2023; 875:147501. [PMID: 37217153 DOI: 10.1016/j.gene.2023.147501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 05/24/2023]
Abstract
Familial hypercholesterolemia (FH) is a monogenic disease characterized by high plasma low-density lipoprotein cholesterol (LDL-c) levels and increased risk of premature atherosclerotic cardiovascular disease. Mutations in FH-related genes account for 40% of FH cases worldwide. In this study, we aimed to assess the pathogenic variants in FH-related genes in the Brazilian FH cohort FHBGEP using exon-targeted gene sequencing (ETGS) strategy. FH patients (n=210) were enrolled at five clinical sites and peripheral blood samples were obtained for laboratory testing and genomic DNA extraction. ETGS was performed using MiSeq platform (Illumina). To identify deleterious variants in LDLR, APOB, PCSK9, and LDLRAP1, the long-reads were subjected to Burrows-Wheeler Aligner (BWA) for alignment and mapping, followed by variant calling using Genome Analysis Toolkit (GATK) and ANNOVAR for variant annotation. The variants were further filtered using in-house custom scripts and classified according to the American College Medical Genetics and Genomics (ACMG) guidelines. A total of 174 variants were identified including 85 missense, 3 stop-gain, 9 splice-site, 6 InDel, and 71 in regulatory regions (3'UTR and 5'UTR). Fifty-two patients (24.7%) had 30 known pathogenic or likely pathogenic variants in FH-related genes according to the American College Medical and Genetics and Genomics guidelines. Fifty-three known variants were classified as benign, or likely benign and 87 known variants have shown uncertain significance. Four novel variants were discovered and classified as such due to their absence in existing databases. In conclusion, ETGS and in silico prediction studies are useful tools for screening deleterious variants and identification of novel variants in FH-related genes, they also contribute to the molecular diagnosis in the FHBGEP cohort.
Collapse
Affiliation(s)
- Jéssica Bassani Borges
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil; Laboratory of Molecular Research in Cardiology, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil; Department of Teaching and Research, Real and Benemerita Associação Portuguesa de Beneficiencia, Sao Paulo 01323-001, Brazil
| | - Victor Fernandes Oliveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil
| | - Carolina Dagli-Hernandez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil
| | - Glaucio Monteiro Ferreira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil; Laboratory of Molecular Research in Cardiology, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | | | - Elisangela da Silva Rodrigues Marçal
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil; Laboratory of Molecular Research in Cardiology, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | - Bruna Los
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil
| | - Vanessa Barbosa Malaquias
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil
| | - Raul Hernandes Bortolin
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil; Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, United States
| | - Renata Caroline Costa Freitas
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil; Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, United States
| | - Augusto Akira Mori
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil
| | - Gisele Medeiros Bastos
- Laboratory of Molecular Research in Cardiology, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil; Department of Teaching and Research, Real and Benemerita Associação Portuguesa de Beneficiencia, Sao Paulo 01323-001, Brazil
| | | | - Daniel Branco Araújo
- Medical Clinic Division, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | - Henry Zatz
- Medical Clinic Division, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | - Adriana Bertolami
- Medical Clinic Division, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | - André Arpad Faludi
- Medical Clinic Division, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | | | | | - João Ítalo Dias França
- Laboratory of Epidemiology and Statistics, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | - Helena Strelow Thurow
- Department of Teaching and Research, Real and Benemerita Associação Portuguesa de Beneficiencia, Sao Paulo 01323-001, Brazil
| | - Thiago Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil
| | - Helder Takashi Imoto Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil
| | - Cinthia Elim Jannes
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of Sao Paulo, Sao Paulo 05403-900, Brazil
| | - Alexandre da Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of Sao Paulo, Sao Paulo 05403-900, Brazil
| | - Vivian Nogueira Silbiger
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal 59078-900 Brazil; Northeast Biotechnology Network (RENORBIO), Graduate Program in Biotechnology, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil
| | - André Ducati Luchessi
- Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal 59078-900 Brazil; Northeast Biotechnology Network (RENORBIO), Graduate Program in Biotechnology, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil
| | - Jéssica Nayara Góes Araújo
- Northeast Biotechnology Network (RENORBIO), Graduate Program in Biotechnology, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil
| | - Marcelo Arruda Nakazone
- Department of Cardiology and Cardiovascular Surgery, Faculdade de Medicina de São José do Rio Preto, Sao Jose do Rio Preto 15090-000, Brazil
| | - Tayanne Silva Carmo
- Department of Cardiology and Cardiovascular Surgery, Faculdade de Medicina de São José do Rio Preto, Sao Jose do Rio Preto 15090-000, Brazil
| | - Dorotéia Rossi Silva Souza
- Department of Biochemistry and Molecular Biology, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto 15090-000, Brazil
| | - Patricia Moriel
- Department of Clinical Pathology, Faculty of Pharmaceutical Sciences, State University of Campinas-UNICAMP, Campinas 13083-871, Brazil
| | - Jaqueline Yu Ting Wang
- Human Genome and Stem-Cell Research Center, Biosciences Institute, University of Sao Paulo, Sao Paulo 05508-090, Brazil
| | - Michel Satya Naslavsky
- Human Genome and Stem-Cell Research Center, Biosciences Institute, University of Sao Paulo, Sao Paulo 05508-090, Brazil
| | - Renata Gorjão
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Sao Paulo 01311-925, Brazil
| | - Tania Cristina Pithon-Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Sao Paulo 01311-925, Brazil
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, Sao Paulo 01311-925, Brazil
| | - Cristina Moreno Fajardo
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil
| | - Hui-Tzu Lin Wang
- Laboratory of Molecular Research in Cardiology, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | - Adriana Regina Garófalo
- Laboratory of Molecular Research in Cardiology, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | - Alvaro Cerda
- Department of Basic Sciences, Center of Excellence in Translational Medicine, BIOREN, Universidad de La Frontera, Temuco 4810296, Chile
| | - Marcelo Ferraz Sampaio
- Department of Cardiology, Real and Benemerita Associação Portuguesa de Beneficiencia, Sao Paulo 01323-001, Brazil
| | - Rosario Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000 SP, Brazil.
| |
Collapse
|
28
|
Liu JT, Doueiry C, Jiang YL, Blaszkiewicz J, Lamprecht MP, Heslop JA, Peterson YK, Carten JD, Traktman P, Yuan Y, Khetani SR, Twal WO, Duncan SA. A human iPSC-derived hepatocyte screen identifies compounds that inhibit production of Apolipoprotein B. Commun Biol 2023; 6:452. [PMID: 37095219 PMCID: PMC10125972 DOI: 10.1038/s42003-023-04739-9] [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: 03/22/2022] [Accepted: 03/21/2023] [Indexed: 04/26/2023] Open
Abstract
Familial hypercholesterolemia (FH) patients suffer from excessively high levels of Low Density Lipoprotein Cholesterol (LDL-C), which can cause severe cardiovascular disease. Statins, bile acid sequestrants, PCSK9 inhibitors, and cholesterol absorption inhibitors are all inefficient at treating FH patients with homozygous LDLR gene mutations (hoFH). Drugs approved for hoFH treatment control lipoprotein production by regulating steady-state Apolipoprotein B (apoB) levels. Unfortunately, these drugs have side effects including accumulation of liver triglycerides, hepatic steatosis, and elevated liver enzyme levels. To identify safer compounds, we used an iPSC-derived hepatocyte platform to screen a structurally representative set of 10,000 small molecules from a proprietary library of 130,000 compounds. The screen revealed molecules that could reduce the secretion of apoB from cultured hepatocytes and from humanized livers in mice. These small molecules are highly effective, do not cause abnormal lipid accumulation, and share a chemical structure that is distinct from any known cholesterol lowering drug.
Collapse
Affiliation(s)
- Jui-Tung Liu
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Caren Doueiry
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Yu-Lin Jiang
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Josef Blaszkiewicz
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Mary Paige Lamprecht
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - James A Heslop
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Yuri K Peterson
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Juliana Debrito Carten
- Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Paula Traktman
- Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Yang Yuan
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Salman R Khetani
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | | | - Stephen A Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Grùthan Biosciences LLC, Hollywood, SC, 29449, USA.
| |
Collapse
|
29
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
30
|
Khalifeh M, Santos RD, Oskuee RK, Badiee A, Aghaee-Bakhtiari SH, Sahebkar A. A novel regulatory facet for hypertriglyceridemia: The role of microRNAs in the regulation of triglyceride-rich lipoprotein biosynthesis. Prog Lipid Res 2023; 89:101197. [PMID: 36400247 DOI: 10.1016/j.plipres.2022.101197] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is one of the major leading global causes of death. Genetic and epidemiological studies strongly support the causal association between triacylglycerol-rich lipoproteins (TAGRL) and atherogenesis, even in statin-treated patients. Recent genetic evidence has clarified that variants in several key genes implicated in TAGRL metabolism are strongly linked to the increased ASCVD risk. There are several triacylglycerol-lowering agents; however, new therapeutic options are in development, among which are miRNA-based therapeutic approaches. MicroRNAs (miRNAs) are small non-coding RNAs (18-25 nucleotides) that negatively modulate gene expression through translational repression or degradation of target mRNAs, thereby reducing the levels of functional genes. MiRNAs play a crucial role in the development of hypertriglyceridemia as several miRNAs are dysregulated in both synthesis and clearance of TAGRL particles. MiRNA-based therapies in ASCVD have not yet been applied in human trials but are attractive. This review provides a concise overview of current interventions for hypertriglyceridemia and the development of novel miRNA and siRNA-based drugs. We summarize the miRNAs involved in the regulation of key genes in the TAGRLs synthesis pathway, which has gained attention as a novel target for therapeutic applications in CVD.
Collapse
Affiliation(s)
- Masoumeh Khalifeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raul D Santos
- Lipid Clinic Heart Institute (Incor), University of São Paulo, Medical School Hospital, São Paulo, Brazil
| | - Reza Kazemi Oskuee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
31
|
Rajan S, Hofer P, Christiano A, Stevenson M, Ragolia L, Villa-Cuesta E, Fried SK, Lau R, Braithwaite C, Zechner R, Schwartz GJ, Hussain MM. Microsomal triglyceride transfer protein regulates intracellular lipolysis in adipocytes independent of its lipid transfer activity. Metabolism 2022; 137:155331. [PMID: 36228741 DOI: 10.1016/j.metabol.2022.155331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/21/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The triglyceride (TG) transfer activity of microsomal triglyceride transfer protein (MTP) is essential for lipoprotein assembly in the liver and intestine; however, its function in adipose tissue, which does not assemble lipoproteins, is unknown. Here we have elucidated the function of MTP in adipocytes. APPROACH AND RESULTS We demonstrated that MTP is present on lipid droplets in human adipocytes. Adipose-specific MTP deficient (A-Mttp-/-) male and female mice fed an obesogenic diet gained less weight than Mttpf/f mice, had less fat mass, smaller adipocytes and were insulin sensitive. A-Mttp-/- mice showed higher energy expenditure than Mttpf/f mice. During a cold challenge, A-Mttp-/- mice maintained higher body temperature by mobilizing more fatty acids. Biochemical studies indicated that MTP deficiency de-repressed adipose triglyceride lipase (ATGL) activity and increased TG lipolysis. Both wild type MTP and mutant MTP deficient in TG transfer activity interacted with and inhibited ATGL activity. Thus, the TG transfer activity of MTP is not required for ATGL inhibition. C-terminally truncated ATGL that retains its lipase activity interacted less efficiently than full-length ATGL. CONCLUSION Our findings demonstrate that adipose-specific MTP deficiency increases ATGL-mediated TG lipolysis and enhances energy expenditure, thereby resisting diet-induced obesity. We speculate that the regulatory function of MTP involving protein-protein interactions might have evolved before the acquisition of TG transfer activity in vertebrates. Adipose-specific inhibition of MTP-ATGL interactions may ameliorate obesity while avoiding the adverse effects associated with inhibition of the lipid transfer activity of MTP.
Collapse
Affiliation(s)
- Sujith Rajan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, United States of America
| | - Peter Hofer
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Amanda Christiano
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, United States of America
| | - Matthew Stevenson
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, United States of America
| | - Louis Ragolia
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, United States of America
| | - Eugenia Villa-Cuesta
- Department of Biology, College of Arts and Science, Adelphi University, Garden City, NY 11530, United States of America
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Raymond Lau
- Department of Surgery, New York University Long Island School of Medicine, Mineola, NY 11501, United States of America
| | - Collin Braithwaite
- Department of Surgery, New York University Long Island School of Medicine, Mineola, NY 11501, United States of America
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz, Austria; BioHealth Field of Excellence, University of Graz, Graz, Austria
| | - Gary J Schwartz
- Department of Medicine and Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States of America.
| | - M Mahmood Hussain
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, United States of America; Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY, United States of America.
| |
Collapse
|
32
|
Farzam K. Lipid disorders in children: diagnosis and treatment. Future Cardiol 2022; 18:915-920. [PMID: 36321773 DOI: 10.2217/fca-2022-0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cardiovascular disease is a leading cause of death in western countries and pediatric lipid disorders create a lifelong continuous risk that starts from childhood. Increased knowledge and awareness on these disorders could prove to be life saving for many patients.
Collapse
Affiliation(s)
- Khashayar Farzam
- Curex Medical Centre, 380 King St N Unit 5, Waterloo, Ontario, N2J 2Z3, Canada
| |
Collapse
|
33
|
Shabbir R, Hayat Malik MN, Zaib M, Alamgeer, Jahan S, Khan MT. Amino Acid Conjugates of 2-Mercaptobenzimidazole Ameliorates High-Fat Diet-Induced Hyperlipidemia in Rats via Attenuation of HMGCR, APOB, and PCSK9. ACS OMEGA 2022; 7:40502-40511. [PMID: 36385864 PMCID: PMC9647896 DOI: 10.1021/acsomega.2c05735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/19/2022] [Indexed: 07/28/2023]
Abstract
PURPOSE This study was designed to explore the antihyperlipidemic effects of amino acid derivatives of 2-mercaptobenzimidazole (4J and 4K) in high-fat diet (HFD)-fed rats. METHODS Male Sprague-Dawley rats were divided into nine groups which received either standard diet or HFD for 28 days. Blood samples were taken on 27th day from HFD-fed rats to ensure hyperlipidemia. HFD-induced hyperlipidemic rats later received daily dosing of either vehicle or simvastatin (SIM; 20 mg/kg) or 4J/4K compounds (10, 20, and 30 mg/kg) for 12 consecutive days. On 40th day, animals were sacrificed, and blood samples were collected for the determination of serum lipid profile and liver function parameters. Liver samples were harvested for histopathological, antioxidant, and qPCR analyses. Molecular docking of tested compounds with HMGCR was also performed to assess the binding affinities. RESULTS 4J and 4K dose dependently decreased serum total cholesterol, triglycerides, low-density lipoprotein, very low-density lipoproteins, alanine transaminase (ALT), and aspartate aminotransferase (AST) levels while significantly alleviated high-density lipoproteins. However, SIM failed to reduce AST and ALT levels. Moreover, tested compounds displayed antioxidant effects by inducing superoxide dismutase and glutathione levels. Histopathology data also displayed protective effects of 4J and 4K against HFD-induced fatty changes and hepatic damage. In addition, 4J and 4K downregulated transcript levels of HMGCR, APOB, PCSK9, and VCAM1, and molecular docking analysis also supported the experimental data. CONCLUSION It is conceivable from this study that 4J and 4K exert their antihyperlipidemic effects by modulating multiple targets regulating lipid levels.
Collapse
Affiliation(s)
- Ramla Shabbir
- Department
of Pharmacology, Faculty of Pharmacy, The
University of Lahore, Lahore 54590, Pakistan
| | | | - Maryam Zaib
- Department
of Pharmacology, Faculty of Pharmacy, The
University of Lahore, Lahore 54590, Pakistan
| | - Alamgeer
- University
College of Pharmacy, University of the Punjab, Lahore 54590, Pakistan
| | - Shah Jahan
- Department
of Immunology, University of Health Sciences, Lahore 54600, Pakistan
| | - Muhammad Tariq Khan
- Department
of Pharmacy, Capital University of Science
and Technology, Islamabad 44000, Pakistan
| |
Collapse
|
34
|
Zhang L, Cai J, Xiao J, Ye Z. Identification of core genes and pathways between geriatric multimorbidity and renal insufficiency: potential therapeutic agents discovered using bioinformatics analysis. BMC Med Genomics 2022; 15:212. [PMID: 36209090 PMCID: PMC9548100 DOI: 10.1186/s12920-022-01370-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/21/2022] [Indexed: 12/03/2022] Open
Abstract
Background Geriatric people are prone to suffer from multiple chronic diseases, which can directly or indirectly affect renal function. Through bioinformatics analysis, this study aimed to identify key genes and pathways associated with renal insufficiency in patients with geriatric multimorbidity and explore potential drugs against renal insufficiency. Methods The text mining tool Pubmed2Ensembl was used to detect genes associated with the keywords including "Geriatric", "Multimorbidity" and "Renal insufficiency". The GeneCodis program was used to specify Gene Ontology (GO) biological process terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Protein–protein interaction (PPI) networks were constructed using STRING and visualized in Cytoscape. Module analysis was performed using CytoHubba and Molecular Complex Detection (MCODE) plugins. GO and KEGG analysis of gene modules was performed using the Database for Annotation, Visualization and Integrated Discover (DAVID) platform database. Genes clustered in salient modules were selected as core genes. Then, the functions and pathways of core genes were visualized using ClueGO and CluePedia. Finally, the drug-gene interaction database was used to explore drug-gene interactions of the core genes to identify drug candidates for renal insufficiency in patients with geriatric multimorbidity. Results Through text mining, 351 genes associated with "Geriatric", "Multimorbidity" and "Renal insufficiency" were identified. A PPI network consisting of 216 nodes and 1087 edges was constructed and CytoHubba was used to sequence the genes. Five gene modules were obtained by MCODE analysis. The 26 genes clustered in module1 were selected as core candidate genes primarily associated with renal insufficiency in patients with geriatric multimorbidity. The HIF-1, PI3K-Akt, MAPK, Rap1, and FoxO signaling pathways were enriched. We found that 21 of the 26 selected genes could be targeted by 34 existing drugs. Conclusion This study indicated that CST3, SERPINA1, FN1, PF4, IGF1, KNG1, IL6, VEGFA, ALB, TIMP1, TGFB1, HGF, SERPINE1, APOA1, APOB, FGF23, EGF, APOE, VWF, TF, CP, GAS6, APP, IGFBP3, P4HB, and SPP1 were key genes potentially involved with renal insufficiency in patients with geriatric multimorbidity. In addition, 34 drugs were identified as potential agents for the treatment and management of renal insufficiency.
Collapse
Affiliation(s)
- Lingyun Zhang
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, People's Republic of China
| | - Jiasheng Cai
- Department of Cardiology, Huadong Hospital Affiliated to Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, People's Republic of China
| | - Jing Xiao
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, People's Republic of China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, People's Republic of China
| | - Zhibin Ye
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, People's Republic of China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
35
|
Gu J, Wu Q, Zhang Q, You Q, Wang L. A decade of approved first-in-class small molecule orphan drugs: Achievements, challenges and perspectives. Eur J Med Chem 2022; 243:114742. [PMID: 36155354 DOI: 10.1016/j.ejmech.2022.114742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 12/01/2022]
Abstract
In the past decade (2011-2020), there was a growing interest in the discovery and development of orphan drugs for the treatment of rare diseases. However, rare diseases only account for a population of 0.65‰-1‰ which usually occur with previously unknown biological mechanisms and lack of specific therapeutics, thus to increase the demands for the first-in-class (FIC) drugs with new biological targets or mechanisms. Considering the achievements in the past 10 years, a total of 410 drugs were approved by U.S. Food and Drug Administration (FDA), which contained 151 FIC drugs and 184 orphan drugs, contributing to make up significant numbers of the approvals. Notably, more than 50% of FIC drugs are developed as orphan drugs and some of them have already been milestones in drug development. In this review, we aim to discuss the FIC small molecules for the development of orphan drugs case by case and highlight the R&D strategy with novel targets and scientific breakthroughs.
Collapse
Affiliation(s)
- Jinying Gu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qiuyu Wu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
36
|
Stankov S, Vitali C, Rader DJ. Gain-of-Function Variants in Lipid Genes Enhance Biological Insight and Point Toward Therapeutic Opportunities. Circulation 2022; 146:740-742. [PMID: 36067277 PMCID: PMC10122829 DOI: 10.1161/circulationaha.122.061233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Sylvia Stankov
- Division of Translational Medicine and Therapeutics, Department of Medicine (S.S., C.V., D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Cecilia Vitali
- Division of Translational Medicine and Therapeutics, Department of Medicine (S.S., C.V., D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel J Rader
- Division of Translational Medicine and Therapeutics, Department of Medicine (S.S., C.V., D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Department of Genetics (D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
37
|
Novel Gene-Correction-Based Therapeutic Modalities for Monogenic Liver Disorders. Bioengineering (Basel) 2022; 9:bioengineering9080392. [PMID: 36004917 PMCID: PMC9404740 DOI: 10.3390/bioengineering9080392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
The majority of monogenic liver diseases are autosomal recessive disorders, with few being sex-related or co-dominant. Although orthotopic liver transplantation (LT) is currently the sole therapeutic option for end-stage patients, such an invasive surgical approach is severely restricted by the lack of donors and post-transplant complications, mainly associated with life-long immunosuppressive regimens. Therefore, the last decade has witnessed efforts for innovative cellular or gene-based therapeutic strategies. Gene therapy is a promising approach for treatment of many hereditary disorders, such as monogenic inborn errors. The liver is an organ characterized by unique features, making it an attractive target for in vivo and ex vivo gene transfer. The current genetic approaches for hereditary liver diseases are mediated by viral or non-viral vectors, with promising results generated by gene-editing tools, such as CRISPR-Cas9 technology. Despite massive progress in experimental gene-correction technologies, limitations in validated approaches for monogenic liver disorders have encouraged researchers to refine promising gene therapy protocols. Herein, we highlighted the most common monogenetic liver disorders, followed by proposed genetic engineering approaches, offered as promising therapeutic modalities.
Collapse
|
38
|
Shen Y, Gu HM, Zhai L, Wang B, Qin S, Zhang DW. The role of hepatic Surf4 in lipoprotein metabolism and the development of atherosclerosis in apoE -/- mice. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159196. [PMID: 35803528 DOI: 10.1016/j.bbalip.2022.159196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 11/26/2022]
Abstract
Elevated plasma levels of low-density lipoprotein-C (LDL-C) increase the risk of atherosclerotic cardiovascular disease. Circulating LDL is derived from very low-density lipoprotein (VLDL) metabolism and cleared by LDL receptor (LDLR). We have previously demonstrated that cargo receptor Surfeit 4 (Surf4) mediates VLDL secretion. Inhibition of hepatic Surf4 impairs VLDL secretion, significantly reduces plasma LDL-C levels, and markedly mitigates the development of atherosclerosis in LDLR knockout (Ldlr-/-) mice. Here, we investigated the role of Surf4 in lipoprotein metabolism and the development of atherosclerosis in another commonly used mouse model of atherosclerosis, apolipoprotein E knockout (apoE-/-) mice. Adeno-associated viral shRNA was used to silence Surf4 expression mainly in the liver of apoE-/- mice. In apoE-/- mice fed a regular chow diet, knockdown of Surf4 expression significantly reduced triglyceride secretion and plasma levels of non-HDL cholesterol and triglycerides without causing hepatic lipid accumulation or liver damage. When Surf4 was knocked down in apoE-/- mice fed the Western-type diet, we observed a significant reduction in plasma levels of non-HDL cholesterol, but not triglycerides. Knockdown of Surf4 did not increase hepatic cholesterol and triglyceride levels or cause liver damage, but significantly diminished atherosclerosis lesions. Therefore, our findings indicate the potential of hepatic Surf4 inhibition as a novel therapeutic strategy to reduce the risk of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Yishi Shen
- Group on the Molecular and Cell Biology of Lipids and Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hong-Mei Gu
- Group on the Molecular and Cell Biology of Lipids and Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lei Zhai
- Institute of Atherosclerosis in Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China
| | - Binxiang Wang
- Institute of Atherosclerosis in Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China
| | - Shucun Qin
- Institute of Atherosclerosis in Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China.
| | - Da-Wei Zhang
- Group on the Molecular and Cell Biology of Lipids and Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
39
|
Pan X, Hussain MM. Bmal1 regulates production of larger lipoproteins by modulating cAMP-responsive element-binding protein H and apolipoprotein AIV. Hepatology 2022; 76:78-93. [PMID: 34626126 PMCID: PMC8993942 DOI: 10.1002/hep.32196] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND AIMS High plasma lipid/lipoprotein levels are risk factors for various metabolic diseases. We previously showed that circadian rhythms regulate plasma lipids and deregulation of these rhythms causes hyperlipidemia and atherosclerosis in mice. Here, we show that global and liver-specific brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1 (Bmal1)-deficient mice maintained on a chow or Western diet developed hyperlipidemia, denoted by the presence of higher amounts of triglyceride-rich and apolipoprotein AIV (ApoAIV)-rich larger chylomicron and VLDL due to overproduction. APPROACH AND RESULTS Bmal1 deficiency decreased small heterodimer partner (Shp) and increased microsomal triglyceride transfer protein (MTP), a key protein that facilitates primordial lipoprotein assembly and secretion. Moreover, we show that Bmal1 regulates cAMP-responsive element-binding protein H (Crebh) to modulate ApoAIV expression and the assembly of larger lipoproteins. This is supported by the observation that Crebh-deficient and ApoAIV-deficient mice, along with Bmal1-deficient mice with knockdown of Crebh, had smaller lipoproteins. Further, overexpression of Bmal1 in Crebh-deficient mice had no effect on ApoAIV expression and lipoprotein size. CONCLUSIONS These studies indicate that regulation of ApoAIV and assembly of larger lipoproteins by Bmal1 requires Crebh. Mechanistic studies showed that Bmal1 regulates Crebh expression by two mechanisms. First, Bmal1 interacts with the Crebh promoter to control circadian regulation. Second, Bmal1 increases Rev-erbα expression, and nuclear receptor subfamily 1 group D member 1 (Nr1D1, Rev-erbα) interacts with the Crebh promoter to repress expression. In short, Bmal1 modulates both the synthesis of primordial lipoproteins and their subsequent expansion into larger lipoproteins by regulating two different proteins, MTP and ApoAIV, through two different transcription factors, Shp and Crebh. It is likely that disruptions in circadian mechanisms contribute to hyperlipidemia and that avoiding disruptions in circadian rhythms may limit/prevent hyperlipidemia and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - M Mahmood Hussain
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
- VA New York Harbor Healthcare System, Brooklyn, NY, USA
| |
Collapse
|
40
|
Chen R, Lin S, Chen X. The promising novel therapies for familial hypercholesterolemia. J Clin Lab Anal 2022; 36:e24552. [PMID: 35712827 PMCID: PMC9279988 DOI: 10.1002/jcla.24552] [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: 04/20/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Background The incidence of premature atherosclerotic cardiovascular disease in familial hypercholesterolemia (FH) is high. In recent years, novel therapeutic modalities have shown significant lipid‐lowering ability. In this paper, we summarize the recent developments in novel therapies for FH via the treatment of different targets and discuss the characteristics of each targeted therapy. Based on the process of protein synthesis, we attempt to summarize the direct‐effect targets including protein, RNA, and DNA. Methods For this systematic review, relevant studies are assessed by searching in several databases including PubMed, Web of Science, Scopus, and Google Scholar. The publications of original researches are considered for screening. Results Most drugs are protein‐targeted such as molecule‐based and monoclonal antibodies, including statins, ezetimibe, alirocumab, evolocumab, and evinacumab. Both antisense oligonucleotide (ASO) and small interfering RNA (siRNA) approaches, such as mipomersen, vupanorsen, inclisiran, and ARO‐ANG3, are designed to reduce the number of mRNA transcripts and then degrade proteins. DNA‐targeted therapies such as adeno‐associated virus or CRISPR–Cas9 modification could be used to deliver or edit genes to address a genetic deficiency and improve the related phenotype. Conclusion While the therapies based on different targets including protein, RNA, and DNA are on different stages of development, the mechanisms of these novel therapies may provide new ideas for precision medicine.
Collapse
Affiliation(s)
- Ruoyu Chen
- School of Medicine of Ningbo University, Ningbo, China
| | - Shaoyi Lin
- The Affiliated Ningbo First Hospital, School of Medicine of Ningbo University, Ningbo, China
| | - Xiaomin Chen
- The Affiliated Ningbo First Hospital, School of Medicine of Ningbo University, Ningbo, China.,Ningbo First Hospital Affiliated to School of Medicine of Zhejiang University, Ningbo, China
| |
Collapse
|
41
|
Duell PB, Welty FK, Miller M, Chait A, Hammond G, Ahmad Z, Cohen DE, Horton JD, Pressman GS, Toth PP. Nonalcoholic Fatty Liver Disease and Cardiovascular Risk: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2022; 42:e168-e185. [PMID: 35418240 DOI: 10.1161/atv.0000000000000153] [Citation(s) in RCA: 307] [Impact Index Per Article: 102.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an increasingly common condition that is believed to affect >25% of adults worldwide. Unless specific testing is done to identify NAFLD, the condition is typically silent until advanced and potentially irreversible liver impairment occurs. For this reason, the majority of patients with NAFLD are unaware of having this serious condition. Hepatic complications from NAFLD include nonalcoholic steatohepatitis, hepatic cirrhosis, and hepatocellular carcinoma. In addition to these serious complications, NAFLD is a risk factor for atherosclerotic cardiovascular disease, which is the principal cause of death in patients with NAFLD. Accordingly, the purpose of this scientific statement is to review the underlying risk factors and pathophysiology of NAFLD, the associations with atherosclerotic cardiovascular disease, diagnostic and screening strategies, and potential interventions.
Collapse
|
42
|
Yadav PK, Haruehanroengra P, Irani S, Wang T, Ansari A, Sheng J, Hussain MM. Novel efficacious microRNA-30c analogs reduce apolipoprotein B secretion in human hepatoma and primary hepatocyte cells. J Biol Chem 2022; 298:101813. [PMID: 35278429 PMCID: PMC8980335 DOI: 10.1016/j.jbc.2022.101813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 12/18/2022] Open
Abstract
High plasma lipid levels have been demonstrated to increase cardiovascular disease risk. Despite advances in treatments to decrease plasma lipids, additional therapeutics are still needed because many people are intolerant or nonresponsive to these therapies. We previously showed that increasing cellular levels of microRNA-30c (miR-30c) using viral vectors or liposomes reduces plasma lipids and atherosclerosis. In this study, we aimed to synthesize potent miR-30c analogs that can be delivered to hepatoma cells without the aid of viral vectors and lipid emulsions. We hypothesized that modification of the passenger strand of miR-30c would increase the stability of miR-30c and augment its delivery to liver cells. Here, we report the successful synthesis of a series of miR-30c analogs by using different chemically modified nucleosides. In these analogs, we left the active sense strand untouched so that its biological activity remained unaltered, and we modified the passenger strand of miR-30c to enhance the stability and uptake of miR-30c by hepatoma cells through phosphorothiorate linkages and the addition of GalNAc. We show that these analogs significantly reduced apolipoprotein B secretion in Huh-7 human hepatoma cells and human primary hepatocytes without affecting apolipoprotein A1 secretion and cellular lipid levels. Our results provide a proof of concept that the passenger strand of miR-30c can be modified to increase its stability and delivery to cells while retaining the potency of the sense strand. We anticipate these miR-30c analogs will be useful in the development of more efficacious analogs for the treatment of hyperlipidemias and cardiovascular diseases.
Collapse
Affiliation(s)
- Pradeep Kumar Yadav
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY 11501, USA
| | | | - Sarah Irani
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY USA
| | - Ting Wang
- Department of Chemistry, The RNA Institute, University at Albany, SUNY, Albany, NY 12222, USA
| | - Abulaish Ansari
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY 11501, USA
| | - Jia Sheng
- Department of Chemistry, The RNA Institute, University at Albany, SUNY, Albany, NY 12222, USA.
| | - M Mahmood Hussain
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY 11501, USA; Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY USA; VA New York Harbor Healthcare System, Brooklyn, NY 11209, USA.
| |
Collapse
|
43
|
Wei N, Hu Y, Liu G, Li S, Yuan G, Shou X, Zhang X, Shi J, Zhai H. A Bibliometric Analysis of Familial Hypercholesterolemia From 2011 to 2021. Curr Probl Cardiol 2022; 48:101151. [PMID: 35202707 DOI: 10.1016/j.cpcardiol.2022.101151] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/16/2022] [Indexed: 01/08/2023]
Abstract
Familial Hypercholesterolemia (FH), an autosomal dominant genetic disease, is increasingly emerging as a global threat. To learn more about the development of FH, 1 617 papers about FH and related research were retrieved in the Web of Science Core Collection from 2011 to 2021. Then, these publications were scientometrically analyzed based on CiteSpace and VOSviewer in terms of spatiotemporal distribution, author distribution, subject categories, topic distribution, and references. The results showed that research on FH is at a stable stage. More FH research has been conducted in developed countries, implying the necessity for strengthening international cooperation and exchanges. We have obtained scholars, institutions, relevant journals, and representative literatures that play an important role in FH. The research direction of FH is on the mechanisms of FH and its complications, diagnosis, statin therapy, and new lipid-lowering drug therapy. Care is the research frontier in FH, and it is in an explosive period.
Collapse
Affiliation(s)
- Namin Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanhui Hu
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guoxiu Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Siyu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guozhen Yuan
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xintian Shou
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuesong Zhang
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingjing Shi
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huaqiang Zhai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
44
|
Paragh G, Németh Á, Harangi M, Banach M, Fülöp P. Causes, clinical findings and therapeutic options in chylomicronemia syndrome, a special form of hypertriglyceridemia. Lipids Health Dis 2022; 21:21. [PMID: 35144640 PMCID: PMC8832680 DOI: 10.1186/s12944-022-01631-z] [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: 11/25/2021] [Accepted: 01/30/2022] [Indexed: 02/07/2023] Open
Abstract
The prevalence of hypertriglyceridemia has been increasing worldwide. Attention is drawn to the fact that the frequency of a special hypertriglyceridemia entity, named chylomicronemia syndrome, is variable among its different forms. The monogenic form, termed familial chylomicronemia syndrome, is rare, occuring in 1 in every 1 million persons. On the other hand, the prevalence of the polygenic form of chylomicronemia syndrome is around 1:600. On the basis of the genetical alterations, other factors, such as obesity, alcohol consumption, uncontrolled diabetes mellitus and certain drugs may significantly contribute to the development of the multifactorial form. In this review, we aimed to highlight the recent findings about the clinical and laboratory features, differential diagnosis, as well as the epidemiology of the monogenic and polygenic forms of chylomicronemias. Regarding the therapy, differentiation between the two types of the chylomicronemia syndrome is essential, as well. Thus, proper treatment options of chylomicronemia and hypertriglyceridemia will be also summarized, emphasizing the newest therapeutic approaches, as novel agents may offer solution for the effective treatment of these conditions.
Collapse
Affiliation(s)
- György Paragh
- Division of Metabolic Diseases, Department of Internal Medicine, University of Debrecen Faculty of Medicine, Nagyerdei krt. 98, Debrecen, H-4032, Hungary.
| | - Ákos Németh
- Division of Metabolic Diseases, Department of Internal Medicine, University of Debrecen Faculty of Medicine, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Mariann Harangi
- Division of Metabolic Diseases, Department of Internal Medicine, University of Debrecen Faculty of Medicine, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Lodz, Poland.,Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Péter Fülöp
- Division of Metabolic Diseases, Department of Internal Medicine, University of Debrecen Faculty of Medicine, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| |
Collapse
|
45
|
Pan X. Cholesterol Metabolism in Chronic Kidney Disease: Physiology, Pathologic Mechanisms, and Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:119-143. [PMID: 35503178 PMCID: PMC11106795 DOI: 10.1007/978-981-19-0394-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High plasma levels of lipids and/or lipoproteins are risk factors for atherosclerosis, nonalcoholic fatty liver disease (NAFLD), obesity, and diabetes. These four conditions have also been identified as risk factors leading to the development of chronic kidney disease (CKD). Although many pathways that generate high plasma levels of these factors have been identified, most clinical and physiologic dysfunction results from aberrant assembly and secretion of lipoproteins. The results of several published studies suggest that elevated levels of low-density lipoprotein (LDL)-cholesterol are a risk factor for atherosclerosis, myocardial infarction, coronary artery calcification associated with type 2 diabetes, and NAFLD. Cholesterol metabolism has also been identified as an important pathway contributing to the development of CKD; clinical treatments designed to alter various steps of the cholesterol synthesis and metabolism pathway are currently under study. Cholesterol synthesis and catabolism contribute to a multistep process with pathways that are regulated at the cellular level in renal tissue. Cholesterol metabolism may also be regulated by the balance between the influx and efflux of cholesterol molecules that are capable of crossing the membrane of renal proximal tubular epithelial cells and podocytes. Cellular accumulation of cholesterol can result in lipotoxicity and ultimately kidney dysfunction and failure. Thus, further research focused on cholesterol metabolism pathways will be necessary to improve our understanding of the impact of cholesterol restriction, which is currently a primary intervention recommended for patients with dyslipidemia.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
46
|
Sun P, Zhao L, Zhang N, Zhou J, Zhang L, Wu W, Ji B, Zhou F. Bioactivity of Dietary Polyphenols: The Role in LDL-C Lowering. Foods 2021; 10:2666. [PMID: 34828946 PMCID: PMC8617782 DOI: 10.3390/foods10112666] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases are the leading causes of the death around the world. An elevation of the low-density lipoprotein cholesterol (LDL-C) level is one of the most important risk factors for cardiovascular diseases. To achieve optimal plasma LDL-C levels, clinal therapies were investigated which targeted different metabolism pathways. However, some therapies also caused various adverse effects. Thus, there is a need for new treatment options and/or combination therapies to inhibit the LDL-C level. Dietary polyphenols have received much attention in the prevention of cardiovascular diseases due to their potential LDL-C lowering effects. However, the effectiveness and potential mechanisms of polyphenols in lowering LDL-C is not comprehensively summarized. This review focused on dietary polyphenols that could reduce LDL-C and their mechanisms of action. This review also discussed the limitations and suggestions regarding previous studies.
Collapse
Affiliation(s)
- Peng Sun
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (P.S.); (N.Z.); (J.Z.); (L.Z.); (B.J.)
| | - Liang Zhao
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China;
| | - Nanhai Zhang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (P.S.); (N.Z.); (J.Z.); (L.Z.); (B.J.)
| | - Jingxuan Zhou
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (P.S.); (N.Z.); (J.Z.); (L.Z.); (B.J.)
| | - Liebing Zhang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (P.S.); (N.Z.); (J.Z.); (L.Z.); (B.J.)
| | - Wei Wu
- College of Engineering, China Agricultural University, Beijing 100083, China;
| | - Baoping Ji
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (P.S.); (N.Z.); (J.Z.); (L.Z.); (B.J.)
| | - Feng Zhou
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (P.S.); (N.Z.); (J.Z.); (L.Z.); (B.J.)
| |
Collapse
|
47
|
Vladimirova-Kitova L, Kitov S, Ganev M, Chochkova-Bukova L. Case Report: Difficulties in the Treatment of a 12-Year-Old Patient With Homozygous Familial Hypercholesterolemia, Compound Heterozygous Form - 5 Years Follow-Up. Front Cardiovasc Med 2021; 8:743341. [PMID: 34692794 PMCID: PMC8531482 DOI: 10.3389/fcvm.2021.743341] [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: 07/18/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
The literature review we conducted reveals the limited use of proprotein convertase subtilisin/kexin type 9-inhibitors (PCSK9i) in children with familial hypercholesterolemia (FH). In 2015, a 10-year-old boy presented with round, xanthochromic lesions on his right knee and elbow. The values of total and LDL-cholesterol (LDL-C)-18 and 15 mmol/l, respectively-along with normal triglycerides and HDL-cholesterol (HDL-C) confirmed the lesions were xanthomas. The data suggested a homozygous form of FH. The level of lipoprotein (a) was high: 270 mg/dl. Initial treatment, based on European recommendations, included Atorvastatin 20 mg and Ezetimibe 10 mg and led to a decrease in LDL-C by 46% for 5 months; however, the patient developed severe statin intolerance. Atorvastatin was replaced with Rosuvastatin 10 mg, but the symptoms persisted. Success was achieved by switching to an intermittent regimen: Rosuvastatin 10 mg three times a week with a daily intake of Ezetimibe 10 mg. However, the results were far from the desired LDL target. LDL-apheresis was advisable, but unfortunately, it is not performed in Bulgaria. In May 2017, a genetic analysis [two pathological mutations within the LDLR gene: c.1519A>G; p.(Lys507Glu) and c.2403_2406del; p.(Leu802Alafs*126)] confirmed the initial diagnosis: the patient had homozygous FH with compound heterozygosity indeed. Having turned 12 in September 2017, the patient was eligible for treatment with a PCSK9i: Evolocumab 140 mg. The mean reduction of LDL-C with the triple combination reached a reduction of 52.17% for the whole 2-year period. The LDL target was reached in January 2020. The triple therapy significantly reduced Apolipoprotein B by 29.16%. No statistically significant difference was found in Lp (a) levels (p > 0.05) Our clinical case demonstrates that the triple lipid-lowering combination in a patient with compound heterozygous FH is a good therapeutic option for reaching the LDL-target.
Collapse
Affiliation(s)
- Lyudmila Vladimirova-Kitova
- First Department of Internal Diseases, Section of Cardiology, Medical University of Plovdiv, Plovdiv, Bulgaria
- Clinic of Cardiology, St. George University Hospital, Plovdiv, Bulgaria
| | - Spas Kitov
- Clinic of Cardiology, St. George University Hospital, Plovdiv, Bulgaria
| | - Mihail Ganev
- Department of Medical Genetics, Medical University of Sofia, Sofia, Bulgaria
| | - Lubov Chochkova-Bukova
- Department of Paediatrics and Medical Genetics, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
48
|
Pandey M, Ojha D, Bansal S, Rode AB, Chawla G. From bench side to clinic: Potential and challenges of RNA vaccines and therapeutics in infectious diseases. Mol Aspects Med 2021; 81:101003. [PMID: 34332771 DOI: 10.1016/j.mam.2021.101003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/27/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022]
Abstract
The functional and structural versatility of Ribonucleic acids (RNAs) makes them ideal candidates for overcoming the limitations imposed by small molecule-based drugs. Hence, RNA-based biopharmaceuticals such as messenger RNA (mRNA) vaccines, antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNA mimics, anti-miRNA oligonucleotides (AMOs), aptamers, riboswitches, and CRISPR-Cas9 are emerging as vital tools for the treatment and prophylaxis of many infectious diseases. Some of the major challenges to overcome in the area of RNA-based therapeutics have been the instability of single-stranded RNAs, delivery to the diseased cell, and immunogenicity. However, recent advancements in the delivery systems of in vitro transcribed mRNA and chemical modifications for protection against nucleases and reducing the toxicity of RNA have facilitated the entry of several exogenous RNAs into clinical trials. In this review, we provide an overview of RNA-based vaccines and therapeutics, their production, delivery, current advancements, and future translational potential in treating infectious diseases.
Collapse
Affiliation(s)
- Manish Pandey
- RNA Biology Laboratory, Regional Centre for Biotechnology, Faridabad, 121001, India
| | - Divya Ojha
- Laboratory of Synthetic Biology, Regional Centre for Biotechnology, Faridabad, 121001, India
| | - Sakshi Bansal
- RNA Biology Laboratory, Regional Centre for Biotechnology, Faridabad, 121001, India
| | - Ambadas B Rode
- Laboratory of Synthetic Biology, Regional Centre for Biotechnology, Faridabad, 121001, India.
| | - Geetanjali Chawla
- RNA Biology Laboratory, Regional Centre for Biotechnology, Faridabad, 121001, India.
| |
Collapse
|
49
|
Izar MCDO, Giraldez VZR, Bertolami A, Santos Filho RDD, Lottenberg AM, Assad MHV, Saraiva JFK, Chacra APM, Martinez TLR, Bahia LR, Fonseca FAH, Faludi AA, Sposito AC, Chagas ACP, Jannes CE, Amaral CK, Araújo DBD, Cintra DE, Coutinho EDR, Cesena F, Xavier HT, Mota ICP, Giuliano IDCB, Faria Neto JR, Kato JT, Bertolami MC, Miname MH, Castelo MHCG, Lavrador MSF, Machado RM, Souza PGD, Alves RJ, Machado VA, Salgado Filho W. Update of the Brazilian Guideline for Familial Hypercholesterolemia - 2021. Arq Bras Cardiol 2021; 117:782-844. [PMID: 34709306 PMCID: PMC8528358 DOI: 10.36660/abc.20210788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
| | - Viviane Zorzanelli Rocha Giraldez
- Instituto do Coração (InCor) da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
- Grupo Fleury, São Paulo, SP - Brasil
| | | | | | - Ana Maria Lottenberg
- Hospital Israelita Albert Einstein (HIAE) - Faculdade Israelita de Ciências da Saúde Albert Einstein (FICSAE), São Paulo, SP - Brasil
- Faculdade de Medicina da Universidade de São Paulo, Laboratório de Lípides (LIM10), São Paulo, São Paulo, SP - Brasil
| | | | | | - Ana Paula M Chacra
- Instituto do Coração (InCor) da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
| | | | | | | | | | - Andrei C Sposito
- Universidade Estadual de Campinas (UNICAMP), Campinas, SP - Brasil
| | | | - Cinthia Elim Jannes
- Instituto do Coração (InCor) da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
| | | | | | | | | | - Fernando Cesena
- Hospital Israelita Albert Einstein (HIAE), São Paulo, SP - Brasil
| | | | | | | | | | | | | | - Marcio Hiroshi Miname
- Instituto do Coração (InCor) da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
| | - Maria Helane Costa Gurgel Castelo
- Universidade Federal do Ceará (UFC), Fortaleza, CE - Brasil
- Hospital do Coração de Messejana, Fortaleza, CE - Brasil
- Professora da Faculdade Unichristus, Fortaleza, CE - Brasil
| | - Maria Sílvia Ferrari Lavrador
- Hospital Israelita Albert Einstein (HIAE) - Faculdade Israelita de Ciências da Saúde Albert Einstein (FICSAE), São Paulo, SP - Brasil
| | - Roberta Marcondes Machado
- Faculdade de Medicina da Universidade de São Paulo, Laboratório de Lípides (LIM10), São Paulo, São Paulo, SP - Brasil
| | - Patrícia Guedes de Souza
- Hospital Universitário Professor Edgard Santos da Universidade Federal da Bahia (UFBA), Salvador, BA - Brasil
| | | | | | - Wilson Salgado Filho
- Instituto do Coração (InCor) da Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP - Brasil
| |
Collapse
|
50
|
Ke W, Afonin KA. Exosomes as natural delivery carriers for programmable therapeutic nucleic acid nanoparticles (NANPs). Adv Drug Deliv Rev 2021; 176:113835. [PMID: 34144087 PMCID: PMC8440450 DOI: 10.1016/j.addr.2021.113835] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 02/06/2023]
Abstract
With recent advances in nanotechnology and therapeutic nucleic acids (TNAs), various nucleic acid nanoparticles (NANPs) have demonstrated great promise in diagnostics and therapeutics. However, the full realization of NANPs' potential necessitates the development of a safe, efficient, biocompatible, stable, tissue-specific, and non-immunogenic delivery system. Exosomes, the smallest extracellular vesicles and an endogenous source of nanocarriers, offer these advantages while avoiding complications associated with manufactured agents. The lipid membranes of exosomes surround a hydrophilic core, allowing for the simultaneous incorporation of hydrophobic and hydrophilic drugs, nucleic acids, and proteins. Additional capabilities for post-isolation exosome surface modifications with imaging agents, targeting ligands, and covalent linkages also pave the way for their diverse biomedical applications. This review focuses on exosomes: their biogenesis, intracellular trafficking, transportation capacities, and applications with emphasis on the delivery of TNAs and programmable NANPs. We also highlight some of the current challenges and discuss opportunities related to the development of therapeutic exosome-based formulations and their clinical translation.
Collapse
Affiliation(s)
- Weina Ke
- Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Kirill A Afonin
- Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223, USA.
| |
Collapse
|