1
|
Varghese A, Gusarov I, Gamallo-Lana B, Dolgonos D, Mankan Y, Shamovsky I, Phan M, Jones R, Gomez-Jenkins M, White E, Wang R, Jones DR, Papagiannakopoulos T, Pacold ME, Mar AC, Littman DR, Nudler E. Unravelling cysteine-deficiency-associated rapid weight loss. Nature 2025:10.1038/s41586-025-08996-y. [PMID: 40399674 DOI: 10.1038/s41586-025-08996-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/08/2025] [Indexed: 05/23/2025]
Abstract
Around 40% of the US population and 1 in 6 individuals worldwide have obesity, with the incidence surging globally1,2. Various dietary interventions, including carbohydrate, fat and, more recently, amino acid restriction, have been explored to combat this epidemic3-6. Here we investigated the impact of removing individual amino acids on the weight profiles of mice. We show that conditional cysteine restriction resulted in the most substantial weight loss when compared to essential amino acid restriction, amounting to 30% within 1 week, which was readily reversed. We found that cysteine deficiency activated the integrated stress response and oxidative stress response, which amplify each other, leading to the induction of GDF15 and FGF21, partly explaining the phenotype7-9. Notably, we observed lower levels of tissue coenzyme A (CoA), which has been considered to be extremely stable10, resulting in reduced mitochondrial functionality and metabolic rewiring. This results in energetically inefficient anaerobic glycolysis and defective tricarboxylic acid cycle, with sustained urinary excretion of pyruvate, orotate, citrate, α-ketoglutarate, nitrogen-rich compounds and amino acids. In summary, our investigation reveals that cysteine restriction, by depleting GSH and CoA, exerts a maximal impact on weight loss, metabolism and stress signalling compared with other amino acid restrictions. These findings suggest strategies for addressing a range of metabolic diseases and the growing obesity crisis.
Collapse
Affiliation(s)
- Alan Varghese
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ivan Gusarov
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Begoña Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Daria Dolgonos
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Yatin Mankan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ilya Shamovsky
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Mydia Phan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Rebecca Jones
- Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY, USA
| | - Maria Gomez-Jenkins
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, USA
| | - Eileen White
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Rui Wang
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Michael E Pacold
- Department of Radiation Oncology and Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Adam C Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Dan R Littman
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
2
|
Shi S, Liu Y, Li X, Zhao W, Feng H, He J, Guo J, Hung W, Wang F, Zhang L, Li Y, He J, Wang R. Oat β-Glucan and Lacticaseibacillus paracasei K56 synergistically ameliorate hypercholesterolemia in mice by modulating gut microbiota and bile acid metabolism. Int J Biol Macromol 2025; 315:144420. [PMID: 40398770 DOI: 10.1016/j.ijbiomac.2025.144420] [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: 02/17/2025] [Revised: 04/23/2025] [Accepted: 05/18/2025] [Indexed: 05/23/2025]
Abstract
Hypercholesterolemia is one of the major risk factors for cardiovascular diseases. While probiotics and prebiotics show cholesterol-lowering potential, current evidence suggests limited efficacy when used alone. This study investigated the synergistic effects of oat β-glucan (OBG) and Lacticaseibacillus paracasei K56 (L. paracasei K56) against diet-induced hypercholesterolemia in mice. Hypercholesterolemic mice were treated for 12 weeks with atorvastatin (5 mg/kg/day), L. paracasei K56 (5 × 109 CFU/kg), OBG (1 g/kg/day) or their combination. The combination treatment demonstrated superior efficacy, reducing total cholesterol (TC) by 41.19 ± 7.13 % (vs model group) and low-density lipoprotein cholesterol (LDL-C) by 37.47 ± 6.55 % (vs model group), with 24.37 ± 9.18 % (vs OBG group) and 15.10 ± 10.30 % (vs K56 group) greater TC reduction. Liver histopathology showed 3.7-fold reduction in steatosis scores versus model group. Mechanistically, compared with the model group, the combination upregulated hepatic CYP7A1/CYP27A1 expression by 5.75 ± 0.51-fold and 2.34 ± 0.21-fold respectively, increased fecal bile acids and enriched bile acid producing bacteria. In summary, L. paracasei K56 and OBG synergistically regulate gut microbiota and bile acid metabolism, promoting the conversion of cholesterol into bile acids, thereby alleviating hypercholesterolemia in mice.
Collapse
Affiliation(s)
- Shaoqi Shi
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China
| | - Yan Liu
- National Center of Technology Innovation for Dairy, Hohhot 100118, China
| | - Xiaoxia Li
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China
| | - Wen Zhao
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China
| | - Haotian Feng
- National Center of Technology Innovation for Dairy, Hohhot 100118, China
| | - Jingjing He
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China
| | - Jie Guo
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China
| | - Weilian Hung
- National Center of Technology Innovation for Dairy, Hohhot 100118, China
| | - Fuqing Wang
- Tibet Tianhong Science and Technology Co., Ltd, Xizang 850000, China
| | - Liwei Zhang
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China
| | - Yixuan Li
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot 100118, China.
| | - Ran Wang
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China.
| |
Collapse
|
3
|
Kannan S, Altae-Tran H, Zhu S, Xu P, Strebinger D, Oshiro R, Faure G, Moeller L, Pham J, Mears KS, Ni HM, Macrae RK, Zhang F. Evolution-guided protein design of IscB for persistent epigenome editing in vivo. Nat Biotechnol 2025:10.1038/s41587-025-02655-3. [PMID: 40335752 DOI: 10.1038/s41587-025-02655-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/26/2025] [Indexed: 05/09/2025]
Abstract
Naturally existing enzymes have been adapted for a variety of molecular technologies, with enhancements or modifications to the enzymes introduced to improve the desired function; however, it is difficult to engineer variants with enhanced activity while maintaining specificity. Here we engineer the compact Obligate Mobile Element Guided Activity (OMEGA) RNA-guided endonuclease IscB and its guiding RNA (ωRNA) by combining ortholog screening, structure-guided protein domain design and RNA engineering, and deep learning-based structure prediction to generate an improved variant, NovaIscB. We show that the compact NovaIscB achieves up to 40% indel activity (~100-fold improvement over wild-type OgeuIscB) on the human genome with improved specificity relative to existing IscBs. We further show that NovaIscB can be fused with a methyltransferase to create a programmable transcriptional repressor, OMEGAoff, that is compact enough to be packaged in a single adeno-associated virus vector for persistent in vivo gene repression. This study highlights the power of combining natural diversity with protein engineering to design enhanced enzymes for molecular biology applications.
Collapse
Affiliation(s)
- Soumya Kannan
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Han Altae-Tran
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Shiyou Zhu
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Peiyu Xu
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Daniel Strebinger
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Rachel Oshiro
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Guilhem Faure
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Lukas Moeller
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Julie Pham
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Kepler S Mears
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Heyuan M Ni
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rhiannon K Macrae
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Yang Tan Collective, Cambridge, MA, USA
| | - Feng Zhang
- Howard Hughes Medical Institute, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Yang Tan Collective, Cambridge, MA, USA.
| |
Collapse
|
4
|
Ma F, Longo M, Meroni M, Bhattacharya D, Paolini E, Mughal S, Hussain S, Anand SK, Gupta N, Zhu Y, Navarro-Corcuera A, Li K, Prakash S, Cogliati B, Wang S, Huang X, Wang X, Yurdagul A, Rom O, Wang L, Fried SK, Dongiovanni P, Friedman SL, Cai B. EHBP1 suppresses liver fibrosis in metabolic dysfunction-associated steatohepatitis. Cell Metab 2025; 37:1152-1170.e7. [PMID: 40015280 PMCID: PMC12058419 DOI: 10.1016/j.cmet.2025.01.020] [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: 06/22/2023] [Revised: 11/27/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025]
Abstract
Excess cholesterol accumulation contributes to fibrogenesis in metabolic dysfunction-associated steatohepatitis (MASH), but how hepatic cholesterol metabolism becomes dysregulated in MASH is not completely understood. We show that human fibrotic MASH livers have decreased EH-domain-binding protein 1 (EHBP1), a genome-wide association study (GWAS) locus associated with low-density lipoprotein (LDL) cholesterol, and that EHBP1 loss- and gain-of-function increase and decrease MASH fibrosis in mice, respectively. Mechanistic studies reveal that EHBP1 promotes sortilin-mediated PCSK9 secretion, leading to LDL receptor (LDLR) degradation, decreased LDL uptake, and reduced TAZ, a fibrogenic effector. At a cellular level, EHBP1 deficiency affects the intracellular localization of retromer, a protein complex required for sortilin stabilization. Our therapeutic approach to stabilizing retromer is effective in mitigating MASH fibrosis. Moreover, we show that the tumor necrosis factor alpha (TNF-α)/peroxisome proliferator-activated receptor alpha (PPARα) pathway suppresses EHBP1 in MASH. These data not only provide mechanistic insights into the role of EHBP1 in cholesterol metabolism and MASH fibrosis but also elucidate an interplay between inflammation and EHBP1-mediated cholesterol metabolism.
Collapse
Affiliation(s)
- Fanglin Ma
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miriam Longo
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Marica Meroni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Shama Mughal
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Syed Hussain
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Neha Gupta
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yiwei Zhu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amaia Navarro-Corcuera
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kenneth Li
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Satya Prakash
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bruno Cogliati
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shuang Wang
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xin Huang
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Liheng Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
5
|
Alidoost M, Huang JY, Dermentzaki G, Blazier AS, Gaglia G, Hammond TR, Frau F, McCorry MC, Ofengeim D, Wilson JL. Uncovering New Therapeutic Targets for Amyotrophic Lateral Sclerosis and Neurological Diseases Using Real-World Data. Clin Pharmacol Ther 2025. [PMID: 40310263 DOI: 10.1002/cpt.3682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025]
Abstract
Although attractive for relevance to real-world scenarios, real-world data (RWD) is typically used for drug repurposing and not therapeutic target discovery. Repurposing studies have identified few effective options in neurological diseases such as the rare disease, amyotrophic lateral sclerosis (ALS), which has no disease-modifying treatments available. We previously reclassified drugs by their simulated effects on proteins downstream of drug targets and observed class-level effects in the EHR, implicating the downstream protein as the source of the effect. Here, we developed a novel ALS-focused network medicine model using data from patient samples, the public domain, and consortia. With this model, we simulated drug effects on ALS and measured class effects on overall survival in retrospective EHR studies. We observed an increased but non-significant risk of death for patients taking drugs with complement system proteins downstream of their targets and experimentally validated drug effects on complement activation. We repeated this for six protein classes, three of which, including multiple chemokine receptors, were associated with a significantly increased risk for death, suggesting that targeting proteins such as CXCR5, CXCR3, chemokine signaling generally, or neuropeptide Y (NPY) could be advantageous therapeutic targets for these patients. We expanded our analysis to the neuroinflammatory condition, myasthenia gravis, and neurodegenerative disease, Parkinson's, and recovered similar effect sizes. We demonstrated the utility of network medicine for testing novel therapeutic effects using RWD and believe this approach may accelerate target discovery in neurological diseases, addressing the critical need for new therapeutic options.
Collapse
Affiliation(s)
- Mohammadali Alidoost
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California, USA
| | - Jeremy Y Huang
- Precision Medicine & Computational Biology, Sanofi Research US, Cambridge, Massachusetts, USA
| | - Georgia Dermentzaki
- Rare & Neurologic Diseases, Sanofi Research US, Cambridge, Massachusetts, USA
| | - Anna S Blazier
- Rare & Neurologic Diseases, Sanofi Research US, Cambridge, Massachusetts, USA
| | - Giorgio Gaglia
- Precision Medicine & Computational Biology, Sanofi Research US, Cambridge, Massachusetts, USA
| | - Timothy R Hammond
- Rare & Neurologic Diseases, Sanofi Research US, Cambridge, Massachusetts, USA
| | - Francesca Frau
- Evidence Generation & Decision Sciences, Sanofi Development, Frankfurt, Germany
| | - Mary Clare McCorry
- Scientific Relations & Initiatives, Sanofi Research US, Cambridge, Massachusetts, USA
| | - Dimitry Ofengeim
- Rare & Neurologic Diseases, Sanofi Research US, Cambridge, Massachusetts, USA
| | - Jennifer L Wilson
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
6
|
Gim DH, Choi IY, Ki YJ, Kim HK, Kim SS, Park KH, Song H, Choi DH. Short-term effects of PCSK-9 inhibitors on percutaneous coronary intervention in patients with acute coronary syndrome. Korean J Intern Med 2025; 40:438-448. [PMID: 40360221 PMCID: PMC12081118 DOI: 10.3904/kjim.2024.363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/20/2024] [Accepted: 12/28/2024] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/AIMS Proprotein-converting enzyme subtilisin-kexin type 9 (PCSK9) inhibitors act more promptly and efficiently than statins and reduce the risk of cardiovascular events in patients with acute coronary syndrome (ACS). This study aimed to assess the short-term effects of perioperative administration of a single-dose PCSK9 inhibitor in patients with ACS. METHODS This study included 789 consecutive patients undergoing percutaneous coronary intervention (PCI) for ACS. The primary clinical endpoint was the occurrence of major adverse cardiovascular events (MACEs) within one month, including cardiac death, non-fatal myocardial infarction, unanticipated revascularization, stroke, stent thrombosis, and rehospitalization for ischemic causes or heart failure. RESULTS PCSK9 inhibitors were administered to 201 of 789 patients. MACEs occurred in eight patients (4.0%) in the treated group and 60 patients (10.2%) in the non-treated group for one month (hazard ratio 0.38, 95% confidence interval 0.18 to 0.80, p = 0.010). The benefit of PCSK9 inhibitors in terms of MACEs was greater in the subgroup of patients treated more than 1 hour before PCI than in the subgroup treated less than 1 hour before PCI or treated after PCI and in the non-treated group. CONCLUSION In patients undergoing PCI for ACS, the risk of MACEs was lower in those treated with perioperative single-dose PCSK9 inhibitors than in those in the untreated group. This benefit was especially noticeable in the subgroups treated > 1 hour before PCI than in those treated less than 1 hour before PCI or after PCI, regardless of the clinical presentation of ACS.
Collapse
Affiliation(s)
- Dong Hyun Gim
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju,
Korea
| | - In Young Choi
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju,
Korea
| | - Young-Jae Ki
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju,
Korea
| | - Hyun Kuk Kim
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju,
Korea
| | - Sung Soo Kim
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju,
Korea
| | - Keun-Ho Park
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju,
Korea
| | - Heesang Song
- Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju,
Korea
| | - Dong-Hyun Choi
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju,
Korea
| |
Collapse
|
7
|
Siddiq A A, Dileep SA, Sj AR, Singam SSR, Martin A. Saffron and its active constituents ameliorate hypercholesterolemia by inhibiting PCSK9 and modulating Sortilin, LDLR, and SREBP-2 signaling in high fat diet induced hypercholesterolemic C57BL/6 mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119697. [PMID: 40157403 DOI: 10.1016/j.jep.2025.119697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 03/17/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saffron (Crocus sativus L.) has long been used in Ayurveda, Iranian, and Chinese traditional medicine as a natural remedy for hypercholesterolemia, obesity, and liver disorders though its therapeutic mechanism remains unclear. AIM OF THE STUDY This study explores the mechanism by which saffron extract (SE), crocin (CN), and crocetin (CR) mitigate high fat diet (HFD) induced hypercholesterolemia and hepatic inflammation in C57BL/6 mice, focusing on their inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9). MATERIALS AND METHODS C57BL/6 mice (N = 10/group) were fed either a, normal diet, HFD, or HFD supplemented with SE, CN, CR, or atorvastatin for 12 weeks. Plasma lipids and inflammatory markers were measured. Histopathological changes were assessed via H&E and Sudan black staining. Gene expression was analyzed using qRT-PCR, and ligand-protein interactions were studied using molecular docking, simulation, and thermophoresis. RESULTS HFD-fed mice exhibited dyslipidemia, liver damage, and inflammation, which SE, CN, and CR significantly improved. Treatments reduced cholesterol, triglycerides, and reactive oxygen species, reversed fatty liver degeneration, and downregulated PCSK9 and sortilin expression while upregulating LDLR. They suppressed transcription factors SREBP-1C and SREBP-2 and reduced inflammatory markers, including TNF-α, while increasing IL-10 expression. CR reduced plasma PCSK9 secretion by 39.9 % (p < 0.05). Docking and simulation studies confirmed the strong binding potential of CR and CN to PCSK9. CONCLUSION Saffron and its active components (CN and CR) are novel natural PCSK9 inhibitors that effectively ameliorate hypercholesterolemia by modulating sortilin, LDLR and SREBP-2 pathway, potentially opening the way for developing new therapeutic approaches for managing cholesterol related disorders.
Collapse
Affiliation(s)
- Aisha Siddiq A
- Department of Biochemistry, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, India.
| | - Shaik Abdul Dileep
- Food Safety and Analytical Quality Control Laboratory, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India.
| | - Aditya Rao Sj
- Plant Cell Biotechnology Department, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India.
| | - Siva Sankara Reddy Singam
- Food Safety and Analytical Quality Control Laboratory, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India.
| | - Asha Martin
- Department of Biochemistry, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, India.
| |
Collapse
|
8
|
Lorincz-Comi N, Cheng F. Bayesian estimation of shared polygenicity identifies drug targets and repurposable medicines for human complex diseases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.17.25324106. [PMID: 40166559 PMCID: PMC11957083 DOI: 10.1101/2025.03.17.25324106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Complex diseases may share portions of their polygenic architectures which can be leveraged to identify drug targets with low off-target potential or repurposable candidates. However, the literature lacks methods which can make these inferences at scale using publicly available data. Methods We introduce a Bayesian model to estimate the polygenic structure of a trait using only gene-based association test statistics from GWAS summary data and returns gene-level posterior risk probabilities (PRPs). PRPs were used to infer shared polygenicity between 496 trait pairs and we introduce measures that can prioritize drug targets with low off-target effects or drug repurposing potential. Results Across 32 traits, we estimated that 69.5 to 97.5% of disease-associated genes are shared between multiple traits, and the estimated number of druggable genes that were only associated with a single disease ranged from 1 (multiple sclerosis) to 59 (schizophrenia). Estimating the shared genetic architecture of ALS with all other traits identified the KIT gene as a potentially harmful drug target because of its deleterious association with triglycerides, but also identified TBK1 and SCN11B as putatively safer because of their non-association with any of the other 31 traits. We additionally found 21 genes which are candidate repourposable targets for Alzheimer's disease (AD) (e.g., PLEKHA1, PPIB) and 5 for ALS (e.g., GAK, DGKQ). Conclusions The sets of candidate drug targets which have limited off-target potential are generally smaller compared to the sets of pleiotropic and putatively repurposable drug targets, but both represent promising directions for future experimental studies.
Collapse
Affiliation(s)
- Noah Lorincz-Comi
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| |
Collapse
|
9
|
Jiao P, Yang N, Jia Q, Fan B, Feng K, Yu J, Zhao S. A dual-reporter LDLR system integrating fluorescence and luminescence for understanding LDLR regulation and facilitating drug discovery. Front Mol Biosci 2025; 12:1552085. [PMID: 40182619 PMCID: PMC11966430 DOI: 10.3389/fmolb.2025.1552085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction The low-density lipoprotein receptor (LDLR) is integral to cholesterol metabolism and cardiovascular health. Enhancing LDLR expression is a promising strategy for treating hyperlipidemia and reducing the risk of atherosclerosis. However, current LDLR reporter systems have limitations in detecting both transcriptional and translational regulation. To address this, we developed a novel dual-reporter LDLR system incorporating Enhanced Green Fluorescent Protein (EGFP) and Gaussia luciferase (Gluc) to enable precise monitoring of LDLR expression and function. Methods A CRISPR/Cas9-mediated knock-in strategy was used to integrate EGFP and Gluc upstream of the stop codon located in exon 18 of the LDLR gene in HEK293 cells. The dual-reporter system allows real-time visualization of LDLR expression via EGFP fluorescence and quantitative assessment through secreted Gluc activity. The system was validated using western blotting, immunofluorescence, and functional assays, including DiI-LDL uptake and drug response analyses with statins and PCSK9 inhibitors. Results The established LDLR-EGFP-Gluc knock-in cell line faithfully recapitulates endogenous LDLR expression and function. EGFP fluorescence accurately reflects LDLR expression dynamics, while Gluc activity provides a highly sensitive and quantitative readout. Functional assays confirmed that LDLR expression responds appropriately to statins and PCSK9 inhibitors. Additionally, screening for transcriptional regulators identified FOXP3 and CREB as novel modulators of LDLR expression, with CREB-mediated regulation involving the sterol regulatory element-binding protein 2 (SREBP2) pathway. Discussion This dual-reporter system enables complementary monitoring of LDLR dynamics, providing enhanced sensitivity, accuracy, and versatility for studying LDLR regulation and function, as well as facilitating drug discovery targeting hyperlipidemia and cardiovascular diseases.
Collapse
Affiliation(s)
- Peng Jiao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Na Yang
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Qianfeng Jia
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Baozhen Fan
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Ke Feng
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Jian Yu
- Department of Basic Medical Education, Yantai Nursing School, Yantai, Shandong, China
| | - Shengtian Zhao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, Shandong, China
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
10
|
Nejabat M, Hadizadeh F, Almahmeed W, Sahebkar A. Effects of PCSK9 inhibitors on cancer, diabetes, and cardiovascular diseases. Drug Discov Today 2025; 30:104316. [PMID: 39986645 DOI: 10.1016/j.drudis.2025.104316] [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: 02/16/2024] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) have potential applications in cancer therapy and as cholesterol-lowering treatments. The impact of PCSK9 suppression on both tumor growth and metastasis, as well as the management of diabetes, has been demonstrated. PCSK9i can also enhance outcomes and reduce cardiovascular (CV) events in individuals with a history of such events. In this review, we provide insights into the pharmacology, safety, and impact of PCSK9i. We highlight cutting-edge investigations, the development of innovative PCSK9i-based products, and a more comprehensive understanding of the potential effects of these drugs on cancer, diabetes, and CV and cerebrovascular diseases.
Collapse
Affiliation(s)
- Mojgan Nejabat
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Jiang Y, Wang Y, Ma S, Qian L, Jing Y, Chen X, Yang J. Efficacy and safety of PCSK9 inhibitors, potent statins, and their combinations for reducing low-density lipoprotein cholesterol in hyperlipidemia patients: a systematic network meta-analysis. Front Cardiovasc Med 2025; 11:1415668. [PMID: 39975967 PMCID: PMC11836037 DOI: 10.3389/fcvm.2024.1415668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/31/2024] [Indexed: 02/21/2025] Open
Abstract
Background The objective of this study is to assess the relative efficacy of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, such as alirocumab, evolocumab, and inclisiran, in conjunction with potent statins like atorvastatin and rosuvastatin, in patients presenting with hyperlipidemia or heightened cardiovascular risk attributable to elevated low-density lipoprotein cholesterol (LDL-C). Methods A systematic search was conducted across databases including PubMed, Embase, and the Cochrane Library to explore lipid-lowering therapies in hyperlipidemia from their inception to 7 November 2023. A network meta-analysis (NMA) was conducted via Stata 17 software, with two authors independently conducting the search, screening, and data abstraction. Results A total of 68 clinical studies involving 21,288 patients with hyperlipidemia were incorporated into the NMA. PSCK9 inhibitors and potent statins significantly reduced LDL-C levels from baseline vs. placebo regardless of background therapy. Regarding the efficacy of lipid reduction, four principal medications were evaluated: evolocumab and atorvastatin [mean standard deviation (MD) -3.41, 95% CI -4.81 to -2.00] and evolocumab with rosuvastatin (MD -3.44, 95% CI -5.10 to -1.78) vs. placebo; alirocumab combined with rosuvastatin (MD -2.91, 95% CI -3.95 to -1.88) and alirocumab with atorvastatin (MD -2.90, 95% CI -3.97 to -1.84) vs. placebo. Meanwhile, compared with placebo, evolocumab (MD -1.89, 95% CI -2.27 to -1.50), alirocumab (MD -1.83, 95% CI -2.09 to -1.57), rosuvastatin (MD -1.93, 95% CI -2.30 to -1.56), inclisiran (MD -1.68, 95% CI -2.10 to -1.27), and atorvastatin (MD -1.68, 95% CI -2.04 to -1.31) could also play a role in the treatment of LDL-C reduction. Moreover, the incidence of adverse events (AEs) was similar to that observed in the control group, which included both placebo and potent statin groups, with no significant differences identified in our study (P > 0.05). Conclusions The combination of PCSK9 inhibitors with robust statins like rosuvastatin and atorvastatin markedly decreases LDL-C levels in patients with hyperlipidemia when compared to placebo or monotherapy. Notably, the pairing of evolocumab and atorvastatin exhibited exceptional efficacy in this investigation. In the interim, the combination of PCSK9 inhibitors and potent statins demonstrates a notable safety profile when contrasted with the control group.
Collapse
Affiliation(s)
- Yuhua Jiang
- Institute of Basic Theory of Traditional Chinese Medicine, China Academic of Chinese Medical Sciences, Beijing, China
| | - Yingying Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Sijia Ma
- Institute of Basic Theory of Traditional Chinese Medicine, China Academic of Chinese Medical Sciences, Beijing, China
| | - Linlin Qian
- Institute of Basic Theory of Traditional Chinese Medicine, China Academic of Chinese Medical Sciences, Beijing, China
| | - Yeteng Jing
- Institute of Basic Theory of Traditional Chinese Medicine, China Academic of Chinese Medical Sciences, Beijing, China
| | - Xi Chen
- Institute of Basic Theory of Traditional Chinese Medicine, China Academic of Chinese Medical Sciences, Beijing, China
| | - Jinsheng Yang
- Institute of Basic Theory of Traditional Chinese Medicine, China Academic of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Chen B, Bao R, Pan J, Zhu Z, Chen Q, Wang D, Wu Y, Yu H, Zhang Y, Wang T. Taurine alleviates dysfunction of cholesterol metabolism under hyperuricemia by inhibiting A2AR-SREBP-2/CREB/HMGCR axis. J Lipid Res 2025; 66:100746. [PMID: 39848583 PMCID: PMC11875148 DOI: 10.1016/j.jlr.2025.100746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/24/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
Dysfunctional cholesterol metabolism is highly prevalent in patients with hyperuricemia. Both uric acid and cholesterol are independent risk factors for atherosclerosis, contributing to an increased incidence of cardiovascular disease in hyperuricemia. Investigating the pathological mechanisms underlying cholesterol metabolism dysfunction in hyperuricemia is essential. This study identified adenosine and inosine, two major purine metabolites, as key regulators of cholesterol biosynthesis. These metabolites upregulate 3-hydroxy-3-methylglutaryl-CoA. Further mechanistic studies revealed that adenosine/inosine up-regulated the expression of 3-hydroxy-3-methylglutaryl-CoA by activating adenosine A2A receptor via the Srebp-2/Creb axis in hyperuricemia. Additionally, we found that taurine deficiency contributes to cholesterol metabolism dysfunction in hyperuricemia. Taurine administration in hyperuricemia mice significantly reduced cholesterol elevation by inhibiting adenosine A2A receptor. This study provides a promising strategy for treating comorbid hypercholesterolemia and hyperuricemia.
Collapse
Affiliation(s)
- Beibei Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruixia Bao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jujie Pan
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zicheng Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qian Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dan Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuzheng Wu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Yi Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Tao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
13
|
Liu Q, Yang R, Wang D, Liu Q. Role of low-density cholesterol and Interleukin-17 interaction in breast cancer pathogenesis and treatment. Cell Biol Int 2025; 49:139-153. [PMID: 39318044 DOI: 10.1002/cbin.12250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024]
Abstract
Breast cancer (BC) has become the most prevalent cancer worldwide, and further research is being conducted to deepen our understanding of its pathogenesis and treatment. Lipid metabolism disorder is a significant alteration in cancer cells, and the investigation into the role of Interleukin-17 (IL-17) in malignant tumors has emerged as a research focus in recent years. Thus, exploring changes in lipid metabolism and inflammatory factors in BC cells is crucial in identifying potential therapeutic targets. This article summarizes the progress made in the research on the main low-density cholesterol (LDL) transporter and IL-17 in lipid metabolism, and their potential involvement in the development of BC. The article aims to establish a theoretical foundation for the development of BC-related therapies.
Collapse
Affiliation(s)
- Qingqing Liu
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
| | - Rongyuan Yang
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
| | - Dawei Wang
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
- The 1st Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangdong, 510405, China
| | - Qing Liu
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
| |
Collapse
|
14
|
Ajoolabady A, Pratico D, Mazidi M, Davies IG, Lip GYH, Seidah N, Libby P, Kroemer G, Ren J. PCSK9 in metabolism and diseases. Metabolism 2025; 163:156064. [PMID: 39547595 DOI: 10.1016/j.metabol.2024.156064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/02/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
PCSK9 is a serine protease that regulates plasma levels of low-density lipoprotein (LDL) and cholesterol by mediating the endolysosomal degradation of LDL receptor (LDLR) in the liver. When PCSK9 functions unchecked, it leads to increased degradation of LDLR, resulting in elevated circulatory levels of LDL and cholesterol. This dysregulation contributes to lipid and cholesterol metabolism abnormalities, foam cell formation, and the development of various diseases, including cardiovascular disease (CVD), viral infections, cancer, and sepsis. Emerging clinical and experimental evidence highlights an imperative role for PCSK9 in metabolic anomalies such as hypercholesterolemia and hyperlipidemia, as well as inflammation, and disturbances in mitochondrial homeostasis. Moreover, metabolic hormones - including insulin, glucagon, adipokines, natriuretic peptides, and sex steroids - regulate the expression and circulatory levels of PCSK9, thus influencing cardiovascular and metabolic functions. In this comprehensive review, we aim to elucidate the regulatory role of PCSK9 in lipid and cholesterol metabolism, pathophysiology of diseases such as CVD, infections, cancer, and sepsis, as well as its pharmaceutical and non-pharmaceutical targeting for therapeutic management of these conditions.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Mohsen Mazidi
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK; King's College London, Department of Twin Research & Genetic Epidemiology, South Wing St Thomas', London, UK; Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ian G Davies
- School of Sport and Exercise Sciences, Faculty of Science, Liverpool John Moores University, Copperas Hill, Liverpool L3 5AJ, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Nabil Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM, affiliated to the University of Montreal), Montreal, QC H2W 1R7, Canada.
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
15
|
Paryani M, Gupta N, Jain SK, Butani S. Lowering LDL cholesterol by PCSK9 inhibition: a new era of gene silencing, RNA, and alternative therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03826-4. [PMID: 39883121 DOI: 10.1007/s00210-025-03826-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) discovery has added a new paradigm to our understanding of cholesterol homeostasis and lipid metabolism. Since its discovery, PCSK9 inhibitors have become a widely investigated therapeutic class for lipid management in cardiovascular diseases and hypercholesterolemia. Scientists have explored different approaches for PCSK9 inhibition, such as monoclonal antibodies (mAbs), gene silencing and gene editing techniques, vaccines, mimetic peptides, and small molecules. European Medicines Agency (EMA) and United States Food and Drug Administration (US FDA) have approved only three PCSK9 inhibitors, including two monoclonal antibodies and one small interfering ribonucleic acid (siRNA). Despite the efficacy of approved large molecules, high costs and the need for regular injection have limited their adherence to the patient. This review aims to provide an understanding of PCSK9's function in Low-Density Lipoprotein Cholesterol (LDL-C) management, its current treatment, recent advancements, and potential future development of small molecules in the class of PCSK9 inhibitors.
Collapse
Affiliation(s)
- Mitali Paryani
- Institute of Pharmacy, Nirma University, Gujarat, 382481, India
| | - Nikita Gupta
- Institute of Pharmacy, Nirma University, Gujarat, 382481, India
| | | | - Shital Butani
- Institute of Pharmacy, Nirma University, Gujarat, 382481, India.
| |
Collapse
|
16
|
Barale C, Tempesta G, Melchionda E, Morotti A, Frascaroli C, Danzero AC, Femminò S, Penna C, Russo I. PCSK9 Expression in Vascular Smooth Muscle Cells: Role of Insulin Resistance and High Glucose. Int J Mol Sci 2025; 26:1003. [PMID: 39940773 PMCID: PMC11817378 DOI: 10.3390/ijms26031003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/06/2024] [Accepted: 01/04/2025] [Indexed: 02/16/2025] Open
Abstract
Beyond the regulation of cholesterol metabolism, a number of extrahepatic functions of proprotein convertase subtilisin/kexin type 9 (PCSK9) have been increasingly identified. The main purpose of this study was to verify whether PCSK9 expression in vascular smooth muscle cells (VSMC) is influenced by insulin resistance and high glucose (HG). In cultured rat aortic VSMC from lean insulin-sensitive Zucker rats (LZRs) and obese insulin-resistant Zucker rats (OZRs), a classical animal model of insulin resistance, we evaluated PCSK9 expression with or without the monoclonal antibodies against PCSK9 Alirocumab and Evolocumab or the synthetic PCSK9-binding peptide PEP 2-8. Effects and molecular mechanisms underlying altered PCSK9 expression were evaluated by proliferation and migration assay, reactive oxygen species (ROS) production, and involvement of PKC, NADPH-oxidase, MAPK/ERK-1/2 pathway activation. As a result, we found that, in comparison with LZR, VSMC from OZR showed basal PCSK9 overexpression mitigated by Alirocumab, Evolocumab, PEP 2-8, and the inhibitors of PKC, NADPH-oxidase, and MAPK. The finding of PCSK9 upregulation in VSMC from OZR paralleled with increased ROS production, proliferation, and migration. HG increased PCSK9 expression in VSMC from LZR, but not in OZR, via oxidative stress and with effects reduced by PCSK9 inhibitors. These findings suggest that a dysregulation of PCSK9 in VSMC could be involved in vascular damage in metabolic disorders, such as obesity and diabetes.
Collapse
Affiliation(s)
- Cristina Barale
- Department of Clinical and Biological Sciences, Turin University, 10043 Orbassano, Italy; (C.B.); (G.T.); (E.M.); (A.M.); (A.C.D.); (S.F.); (C.P.)
| | - Giulia Tempesta
- Department of Clinical and Biological Sciences, Turin University, 10043 Orbassano, Italy; (C.B.); (G.T.); (E.M.); (A.M.); (A.C.D.); (S.F.); (C.P.)
| | - Elena Melchionda
- Department of Clinical and Biological Sciences, Turin University, 10043 Orbassano, Italy; (C.B.); (G.T.); (E.M.); (A.M.); (A.C.D.); (S.F.); (C.P.)
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, Turin University, 10043 Orbassano, Italy; (C.B.); (G.T.); (E.M.); (A.M.); (A.C.D.); (S.F.); (C.P.)
| | | | - Alice Costanza Danzero
- Department of Clinical and Biological Sciences, Turin University, 10043 Orbassano, Italy; (C.B.); (G.T.); (E.M.); (A.M.); (A.C.D.); (S.F.); (C.P.)
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, Turin University, 10043 Orbassano, Italy; (C.B.); (G.T.); (E.M.); (A.M.); (A.C.D.); (S.F.); (C.P.)
| | - Claudia Penna
- Department of Clinical and Biological Sciences, Turin University, 10043 Orbassano, Italy; (C.B.); (G.T.); (E.M.); (A.M.); (A.C.D.); (S.F.); (C.P.)
| | - Isabella Russo
- Department of Clinical and Biological Sciences, Turin University, 10043 Orbassano, Italy; (C.B.); (G.T.); (E.M.); (A.M.); (A.C.D.); (S.F.); (C.P.)
| |
Collapse
|
17
|
Mei W, Faraj Tabrizi S, Godina C, Lovisa AF, Isaksson K, Jernström H, Tavazoie SF. A commonly inherited human PCSK9 germline variant drives breast cancer metastasis via LRP1 receptor. Cell 2025; 188:371-389.e28. [PMID: 39657676 PMCID: PMC11770377 DOI: 10.1016/j.cell.2024.11.009] [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: 02/12/2024] [Revised: 08/12/2024] [Accepted: 11/10/2024] [Indexed: 12/12/2024]
Abstract
Identifying patients at risk for metastatic relapse is a critical medical need. We identified a common missense germline variant in proprotein convertase subtilisin/kexin type 9 (PCSK9) (rs562556, V474I) that is associated with reduced survival in multiple breast cancer patient cohorts. Genetic modeling of this gain-of-function single-nucleotide variant in mice revealed that it causally promotes breast cancer metastasis. Conversely, host PCSK9 deletion reduced metastatic colonization in multiple breast cancer models. Host PCSK9 promoted metastatic initiation events in lung and enhanced metastatic proliferative competence by targeting tumoral low-density lipoprotein receptor related protein 1 (LRP1) receptors, which repressed metastasis-promoting genes XAF1 and USP18. Antibody-mediated therapeutic inhibition of PCSK9 suppressed breast cancer metastasis in multiple models. In a large Swedish early-stage breast cancer cohort, rs562556 homozygotes had a 22% risk of distant metastatic relapse at 15 years, whereas non-homozygotes had a 2% risk. Our findings reveal that a commonly inherited genetic alteration governs breast cancer metastasis and predicts survival-uncovering a hereditary basis underlying breast cancer metastasis.
Collapse
Affiliation(s)
- Wenbin Mei
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | | | - Christopher Godina
- Division of Oncology, Department of Clinical Sciences in Lund, Lund University Cancer Center/Kamprad, Lund, Sweden
| | - Anthea F Lovisa
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Karolin Isaksson
- Division of Surgery, Department of Clinical Sciences in Lund, Lund University and Department of Surgery Kristianstad Hospital, Lund, Sweden
| | - Helena Jernström
- Division of Oncology, Department of Clinical Sciences in Lund, Lund University Cancer Center/Kamprad, Lund, Sweden
| | - Sohail F Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
18
|
Sadowski M, Thompson M, Mefford J, Haldar T, Oni-Orisan A, Border R, Pazokitoroudi A, Cai N, Ayroles JF, Sankararaman S, Dahl AW, Zaitlen N. Characterizing the genetic architecture of drug response using gene-context interaction methods. CELL GENOMICS 2024; 4:100722. [PMID: 39637863 DOI: 10.1016/j.xgen.2024.100722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/24/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
Identifying factors that affect treatment response is a central objective of clinical research, yet the role of common genetic variation remains largely unknown. Here, we develop a framework to study the genetic architecture of response to commonly prescribed drugs in large biobanks. We quantify treatment response heritability for statins, metformin, warfarin, and methotrexate in the UK Biobank. We find that genetic variation modifies the primary effect of statins on LDL cholesterol (9% heritable) as well as their side effects on hemoglobin A1c and blood glucose (10% and 11% heritable, respectively). We identify dozens of genes that modify drug response, which we replicate in a retrospective pharmacogenomic study. Finally, we find that polygenic score (PGS) accuracy varies up to 2-fold depending on treatment status, showing that standard PGSs are likely to underperform in clinical contexts.
Collapse
Affiliation(s)
- Michal Sadowski
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Mike Thompson
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Joel Mefford
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tanushree Haldar
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA; Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Akinyemi Oni-Orisan
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA; Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Richard Border
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computational Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ali Pazokitoroudi
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Na Cai
- Helmholtz Pioneer Campus, Helmholtz Munich, 85764 Neuherberg, Germany; Computational Health Centre, Helmholtz Munich, 85764 Neuherberg, Germany; School of Medicine and Health, Technical University of Munich, 80333 Munich, Germany
| | - Julien F Ayroles
- Department of Ecology and Evolution, Princeton University, Princeton, NJ 08544, USA; Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Sriram Sankararaman
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computational Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Andy W Dahl
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Noah Zaitlen
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computational Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
19
|
Medoro A, Scapagnini G, Brogi S, Jafar TH, Trung TT, Saso L, Davinelli S. Carotenoid Interactions with PCSK9: Exploring Novel Cholesterol-Lowering Strategies. Pharmaceuticals (Basel) 2024; 17:1597. [PMID: 39770439 PMCID: PMC11676125 DOI: 10.3390/ph17121597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: This study investigated the potential of green algae-derived carotenoids as natural inhibitors of the proprotein convertase subtilisin/kexin type 9 (PCSK9), a key regulator of cholesterol metabolism. PCSK9 promotes the degradation of low-density lipoprotein receptors (LDLR), thereby increasing blood cholesterol levels and elevating the risk of cardiovascular diseases. Methods/Results: We screened the pharmacophore fit score of 27 carotenoids with PCSK9 and identified 14 that were analyzed for binding affinity and molecular interactions. Astaxanthin, siphonaxanthin, and prasinoxanthin were identified as the top candidates, demonstrating strong binding affinity (-10.5, -10.3, and -9.4 Kcal/mol, respectively) and stable interactions with several known key residues within the active site of PCSK9, including Pro-331, Arg-357, Cys-358, Val-359, Asp-360, Ile-416, Leu-436, Thr-437, Pro-438, Leu-440, Arg-458, Val-460, Trp-461, Arg-476, Cys-477, Ala-478, Ala-649, Val-650, and Asp-651. Density functional theory analysis confirmed the stability of astaxanthin and its favorable electronic properties, suggesting its potential as an effective inhibitor. Molecular dynamics simulations of the PCSK9-astaxanthin complex revealed sustained structural stability and key interactions critical for maintaining the functional integrity of the protein. Conclusions: These findings provide evidence that specific carotenoids, particularly astaxanthin, may offer a cost-effective alternative to existing PCSK9 inhibitors, providing a potential approach for managing cholesterol levels and reducing cardiovascular risk. Pre-clinical and clinical validations are required to confirm the therapeutic potential of these compounds.
Collapse
Affiliation(s)
- Alessandro Medoro
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.M.); (G.S.); (T.H.J.)
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.M.); (G.S.); (T.H.J.)
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy;
| | - Tassadaq Hussain Jafar
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.M.); (G.S.); (T.H.J.)
| | - Truong Tan Trung
- Laboratory of Computation and Nanoscience, Dong Nai Technology University, Bien Hoa City 810000, Vietnam;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, 00185 Rome, Italy;
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.M.); (G.S.); (T.H.J.)
| |
Collapse
|
20
|
Jeswani BM, Sharma S, Rathore SS, Nazir A, Bhatheja R, Kapoor K. PCSK9 Inhibitors: The Evolving Future. Health Sci Rep 2024; 7:e70174. [PMID: 39479289 PMCID: PMC11522611 DOI: 10.1002/hsr2.70174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/14/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction PCSK9 inhibitors are a novel class of medications that lower LDL cholesterol (LDL-C) by increasing LDL receptor activity, promoting clearance of LDL-C from the bloodstream. Over the years, PCSK9 inhibitors have been explored as adjunct therapies to statins or as monotherapy in high-risk cardiovascular patients. Aim This review aims to provide an updated perspective on PCSK9 inhibitors, assessing their clinical efficacy, safety, and significance, especially in light of recent clinical trials. Methods The review examines the role of PCSK9 in cholesterol regulation and summarizes the results of major cardiovascular trials, including FOURIER, SPIRE-1, SPIRE-2, and ODYSSEY Outcomes. It also discusses emerging treatments like small interfering RNA (siRNA) therapies and evaluates PCSK9 inhibitor effects on LDL-C and lipoprotein(a) levels. Results Clinical trials have shown PCSK9 inhibitors reduce LDL-C by up to 60%. In the FOURIER trial, evolocumab reduced LDL-C by 59% and major cardiovascular events by 15%-20%. The SPIRE-2 trial, despite early termination, showed a 21% risk reduction in the primary composite endpoint with bococizumab. The ODYSSEY Outcomes trial reported a 57% LDL-C reduction with alirocumab, alongside a 15% reduction in adverse events. Emerging treatments like Inclisiran offer long-term LDL-C control with fewer doses. PCSK9 inhibitors are generally well-tolerated, with the most common side effect being injection site reactions. Conclusion PCSK9 inhibitors significantly lower LDL-C and reduce cardiovascular events, offering promising therapies for high-risk patients, including those with familial hypercholesterolemia (FH) and those who cannot tolerate statins. Future research will focus on optimizing these inhibitors, integrating complementary therapies, and exploring gene-editing technologies to improve patient outcomes.
Collapse
Affiliation(s)
- Bijay Mukesh Jeswani
- Department of MedicineGCS Medical College, Hospital & Research CentreAhmedabadIndia
| | | | | | - Abubakar Nazir
- Department of MedicineKing Edward Medical UniversityLahorePakistan
- Department of MedicineOli Health Magazine Organization, Research, and EducationKigaliRwanda
| | | | - Kapil Kapoor
- Cardiology, AdventHealth OrlandoOrlandoFloridaUSA
| |
Collapse
|
21
|
Ahmad P, Alvi SS, Waiz M, Khan MS, Ahmad S, Khan MS. Naturally occurring organosulfur compounds effectively inhibits PCSK-9 activity and restrict PCSK-9-LDL-receptor interaction via in-silico and in-vitro approach. Nat Prod Res 2024; 38:3924-3933. [PMID: 37842787 DOI: 10.1080/14786419.2023.2269465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 09/20/2023] [Accepted: 10/01/2023] [Indexed: 10/17/2023]
Abstract
The present study intended to divulge the potential role of garlic-derived organosulfur compounds (OSCs) in targeting PCSK-9 and averting its interaction with the EGF-A portion of LDL-R via in-vitro and in-silico analysis. Our in-silico screening data showed that 3-(Propylsulfinyl)-L-alanine (PSA), S-Ethyl-L-cysteine (SEC), alliin, and S-Allyl-L-cysteine (SAC) exhibited higher binding energy (-7.05, -7.00, -6.65, and -6.31 Kcal/mol, respectively) against PCSK-9, among other selected OSCs. Further, the protein-protein interaction study of PCSK-9-OSCs-complex with EGF-A demonstrated a similar binding pattern with E-total values ranging from -430.01 to -405.6 Kcal/mol. These results were further validated via in-vitro analysis which showed that SEC, SAC, and diallyl trisulphide (DAT) exhibited the lowest IC50 values of 4.70, 5.26, and 5.29 µg/mL, respectively. In conclusion, the presented data illustrated that SEC, SAC, and DAT were the best inhibitors of PCSK-9 activity and may have the potential to improve the LDL-R function and lower the circulatory LDL-C level.
Collapse
Affiliation(s)
- Parvej Ahmad
- IIRC-5, Clinical Biochemistry and Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Sahir S Alvi
- Department of Immunology and Microbiology, South TX Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Mohd Waiz
- IIRC-5, Clinical Biochemistry and Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Kingdom of Saudi Arabia
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - M Salman Khan
- IIRC-5, Clinical Biochemistry and Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
22
|
Rosman Z, Maor Y, Zohar I, Balmor GR, Pravda MS, Goldstein AL, Tocut M, Soroksky A. Proprotein Convertase Subtilisin Kexin 9 Inhibitor in Severe Sepsis and Septic Shock Patients in a Phase II Prospective Cohort Study-Preliminary Results. Infect Dis Rep 2024; 16:1036-1044. [PMID: 39584843 PMCID: PMC11586949 DOI: 10.3390/idr16060083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/12/2024] [Accepted: 10/22/2024] [Indexed: 11/26/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction syndrome caused by a dysregulated host response to infection that has a high mortality rate. Proprotein convertase subtilisin kexin 9 (PCSK9) is a serine protease secreted by the liver. Its binding to the low-density lipoprotein (LDL) receptor enhances its degradation, causing an increase in LDL levels in the blood. Objectives: Administering a PCSK9 inhibitor leading to an increase in lipid uptake by the liver may positively affect septic patients due to the increased removal of endotoxins. Methods: This preliminary study aimed to examine the safety of PCSK9 inhibitor use in septic and septic shock patients. We treated five septic patients in the intensive care unit with 300 mg of alirocumab following serious adverse events for 28 days. Results: Four of our patients did not experience any adverse events, and all of them survived. One patient died after discharge from the intensive care unit, and this death was presumably not related to the study drug. The patients rapidly recovered from the inflammatory stage of sepsis. Conclusions: Alirocumab appears safe in severe sepsis and septic shock patients. The outcome data are promising. Only a basic safety profile can be assessed based on this pilot study. Further study with a PCSK-9 inhibitor in septic or septic shock patients is required to further determine its benefit in ICU patients.
Collapse
Affiliation(s)
- Ziv Rosman
- Intensive Care Department, E. Wolfson Medical Center, Holon 5822012, Israel; (G.R.B.); (M.S.P.); (A.S.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.M.); (I.Z.); (M.T.)
| | - Yasmin Maor
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.M.); (I.Z.); (M.T.)
- Infectious Disease Unit, E. Wolfson Medical Center, Holon 5822012, Israel
| | - Iris Zohar
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.M.); (I.Z.); (M.T.)
- Infectious Disease Unit, E. Wolfson Medical Center, Holon 5822012, Israel
| | - Gingy Ronen Balmor
- Intensive Care Department, E. Wolfson Medical Center, Holon 5822012, Israel; (G.R.B.); (M.S.P.); (A.S.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.M.); (I.Z.); (M.T.)
| | - Miri Schamroth Pravda
- Intensive Care Department, E. Wolfson Medical Center, Holon 5822012, Israel; (G.R.B.); (M.S.P.); (A.S.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.M.); (I.Z.); (M.T.)
| | - Adam Lee Goldstein
- Surgical Department A Trauma Division, E. Wolfson Medical Center, Holon 5822012, Israel;
| | - Milena Tocut
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.M.); (I.Z.); (M.T.)
- Internal Medicine C Department, E. Wolfson Medical Center, Holon 5822012, Israel
| | - Arie Soroksky
- Intensive Care Department, E. Wolfson Medical Center, Holon 5822012, Israel; (G.R.B.); (M.S.P.); (A.S.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (Y.M.); (I.Z.); (M.T.)
| |
Collapse
|
23
|
Borràs C, Canyelles M, Girona J, Ibarretxe D, Santos D, Revilla G, Llorente-Cortes V, Rotllan N, Kovanen PT, Jauhiainen M, Lee-Rueckert M, Masana L, Arrieta F, Martínez-Botas J, Gómez-Coronado D, Ribalta J, Tondo M, Blanco-Vaca F, Escolà-Gil JC. PCSK9 Antibodies Treatment Specifically Enhances the Macrophage-specific Reverse Cholesterol Transport Pathway in Heterozygous Familial Hypercholesterolemia. JACC Basic Transl Sci 2024; 9:1195-1210. [PMID: 39534644 PMCID: PMC11551875 DOI: 10.1016/j.jacbts.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 11/16/2024]
Abstract
We investigated the potential of proprotein convertase subtilisin/kexin type 9 (PCSK9) antibodies to restore macrophage cholesterol efflux in subjects with heterozygous familial hypercholesterolemia (FH) and to enhance the macrophage-specific reverse cholesterol transport pathway in mice. Analyses of macrophage-derived cholesterol distribution of plasma from FH patients revealed that low-density lipoprotein (LDL) particles contained less, and high-density lipoprotein particles contained more radiolabeled cholesterol after treatment with either PCSK9 inhibitor. PCSK9 antibodies facilitated the transfer of macrophage-derived cholesterol and LDL-derived cholesterol to feces exclusively in heterozygous LDL receptor-deficient mice expressing human APOB100. PCSK9 inhibitors act as positive regulators of the macrophage-specific reverse cholesterol transport pathway in individuals with heterozygous FH.
Collapse
Affiliation(s)
- Carla Borràs
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Marina Canyelles
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Josefa Girona
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Rovira i Virgili University, IISPV, Reus, Spain
| | - Daiana Ibarretxe
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Rovira i Virgili University, IISPV, Reus, Spain
| | - David Santos
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Giovanna Revilla
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vicenta Llorente-Cortes
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Noemí Rotllan
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | | | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research and Finnish Institute for Health and Welfare, Department of Public Health and Welfare, Biomedicum, Helsinki, Finland
| | | | - Luis Masana
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Rovira i Virgili University, IISPV, Reus, Spain
| | - Francisco Arrieta
- Servicio de Endocrinología y Nutrición, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Javier Martínez-Botas
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Diego Gómez-Coronado
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Josep Ribalta
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Rovira i Virgili University, IISPV, Reus, Spain
| | - Mireia Tondo
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Francisco Blanco-Vaca
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Joan Carles Escolà-Gil
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| |
Collapse
|
24
|
Ogura M. Do You Prescribe PCSK9 Inhibitors Properly? Circ J 2024; 88:1637-1638. [PMID: 38556298 DOI: 10.1253/circj.cj-24-0186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Affiliation(s)
- Masatsune Ogura
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University
| |
Collapse
|
25
|
Luo Y, Hou Y, Zhao W, Yang B. Recent progress in gene therapy for familial hypercholesterolemia treatment. iScience 2024; 27:110641. [PMID: 39262805 PMCID: PMC11387600 DOI: 10.1016/j.isci.2024.110641] [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: 09/13/2024] Open
Abstract
Familial hypercholesterolemia (FH) is a genetic disorder that affects 1 in 300 people, leading to high cholesterol levels and significantly increased cardiovascular risk. The limitations of existing FH treatments underscore the need for innovative therapeutics, and gene therapy offers a promising alternative to address FH more effectively. In this review, we survey approved gene therapy drugs first and then delve into the landscape of gene addition, gene inactivation, and gene editing therapies for hypercholesterolemia, highlighting both approved interventions and those in various stages of development. We also discussed recent advancements in gene editing tools that are essential for their application in gene therapy. Safety considerations inherent to gene therapy are also discussed, emphasizing the importance of mitigating potential risks associated with such treatments. Overall, this review highlights the progress and prospects of gene therapies for FH treatments, underscoring their potential to revolutionize the management of this prevalent and challenging condition.
Collapse
Affiliation(s)
- Yaxin Luo
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yaofeng Hou
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wenwen Zhao
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Bei Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
- Shanghai Frontiers Science Center for Biomacromolecules and Precision Medicine, ShanghaiTech University, Shanghai 200031, China
| |
Collapse
|
26
|
Zhang Y, Wei J, Wang H, Wang Y. Characterization of NiCas12b for In Vivo Genome Editing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400469. [PMID: 39076074 PMCID: PMC11423069 DOI: 10.1002/advs.202400469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/08/2024] [Indexed: 07/31/2024]
Abstract
The RNA-guided clustered regularly interspaced short palindromic repeats (CRISPR)/Cas12b system represents the third family of CRISPR-Cas systems that are harnessed for genome editing. However, only a few nucleases have demonstrated activity in human cells, and their in vivo therapeutic potential remains uncertain. In this study, a green fluorescent protein (GFP)-activation assay is conducted to screen a panel of 15 Cas12b orthologs, and four of them exhibited editing activity in mammalian cells. Particularly noteworthy is the NiCas12b derived from Nitrospira sp., which recognizes a "TTN" protospacer adjacent motif (PAM) and facilitates efficient genome editing in various cell lines. Importantly, NiCas12b also exhibits a high degree of specificity, rendering it suitable for therapeutic applications. As proof of concept, the adeno-associated virus (AAV) is employed to introduce NiCas12b to target the cholesterol regulatory gene proprotein convertase subtilisin/ kexin type 9 (Pcsk9) in the mouse liver. After 4 weeks of injections, an impressive is observed over 16.0% insertion/deletion (indel) efficiency, resulting in a significant reduction in serum cholesterol levels. NiCas12b provides a novel option for both basic research and clinical applications.
Collapse
Affiliation(s)
- Yunqian Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jingjing Wei
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai, 200438, China
| | - Hongyan Wang
- Obstetrics & Gynecology Hospital, State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438, China
- Children's Hospital, Fudan University, Shanghai, 201102, China
| | - Yongming Wang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai, 200438, China
- Shanghai Engineering Research Center of Industrial Microorganisms, Shanghai, 200438, China
| |
Collapse
|
27
|
Pihtili Taş N, Aydogan Baykara R, Kamanli A, Gürbüz A, Cure E, Cumhur Cüre M, Erdem M, Tasar Yildirim T. Proprotein convertase subtilisin/kexin type 9 and apelin in fibromyalgia syndrome. Arch Rheumatol 2024; 39:375-383. [PMID: 39507838 PMCID: PMC11537681 DOI: 10.46497/archrheumatol.2024.10462] [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: 09/04/2023] [Accepted: 10/16/2023] [Indexed: 11/08/2024] Open
Abstract
OBJECTIVES This study aimed to investigate the potential roles of proprotein convertase subtilisin/ kexin type 9 (PCSK9) and apelin in the etiology of fibromyalgia syndrome (FS). PATIENTS AND METHODS The retrospective study was conducted between May 2022 and February 2023. Fifty-eight female FS patients (mean age: 45.2±9.9 years; range, 25 to 66 years) and 30 age- and body mass index-matched control subjects (mean age: 43.1±9.9 years; range, 26 to 67 years) were included in the study. Apelin and PCSK9 levels of all individuals were measured using appropriate methods. RESULTS The levels of PCSK9 (173.2±62.2 vs. 75.1±44.1, p<0.001) and apelin (354.6±195.5 vs. 229.0±83.2, p<0.001) were significantly higher in patients with FS compared to the control group. A positive correlation was found between PCSK9 and apelin levels and various measures, including the Fibromyalgia Impact Questionnaire (FIQ), Symptom Severity Scale (SSS), Pittsburgh Sleep Quality Index (PSQI), and Beck Depression Inventory (BDI). Additionally, there was a positive correlation between apelin levels and FIQ, SSS, PSQI, Beck Anxiety Inventory, and BDI scores. The optimal cutoff value for PCSK9 in predicting FS was 110.0 ng/mL, with a sensitivity of 84.5% and specificity of 83.9% (area under the curve [AUC]=0.920, 95% confidence interval [CI]: 0.852-0.987, p<0.001). For apelin, the optimal cutoff value for predicting FS was 258.8 ng/L, with a sensitivity of 63.8% and specificity of 64.5% (AUC=0.732, 95% CI: 0.623-0.840, p<0.001). CONCLUSION Our findings suggest that PCSK9 may play a role in FS etiology and potentially contribute to oxidative stress. Increased apelin levels may be a compensatory response to high oxidative stress, possibly leading to hyperalgesia. Both PCSK9 and apelin can be predictive markers for FS.
Collapse
Affiliation(s)
- Nevsun Pihtili Taş
- Department of Physical Medicine and Rehabilitation, Health Sciences University, Elazığ Fethi Sekin City Health Application and Research Center, Elazığ, Türkiye
| | - Rabia Aydogan Baykara
- Department of Physical Medicine and Rehabilitation, Malatya Turgut Özal University, Trainnig and Research Hospital, Malatya, Türkiye
| | - Ayhan Kamanli
- Department of Physical Medicine and Rehabilitation, Sakarya University Faculty of Medicine, Sakarya, Türkiye
| | - Ali Gürbüz
- Department of Physical Medicine and Rehabilitation, Elazığ Fethi Sekin City Hospital, Elazığ, Türkiye
| | - Erkan Cure
- Department of Internal Medicine, Bağcılar Medilife Hospital, İstanbul, Türkiye
| | | | - Mehmet Erdem
- Department of Biochemistry, Malatya Turgut Özal University, Malatya, Türkiye
| | | |
Collapse
|
28
|
Modica R, La Salvia A, Liccardi A, Cozzolino A, Di Sarno A, Russo F, Colao A, Faggiano A. Dyslipidemia, lipid-lowering agents and neuroendocrine neoplasms: new horizons. Endocrine 2024; 85:520-531. [PMID: 38509261 PMCID: PMC11291585 DOI: 10.1007/s12020-024-03767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/29/2024] [Indexed: 03/22/2024]
Abstract
PURPOSE Neuroendocrine neoplasms (NENs) are a heterogeneous group of malignancies originating from cells with a neuroendocrine phenotype. The complex relationship between lipid metabolism and cancer is gaining interest and a potential anti-cancer effect of lipid lowering agents is being considered. This review aims to discuss the current understanding and treatment of dyslipidaemia in NENs, focusing on the role of lipid lowering agents, including new therapeutic approaches, and future perspectives as possible tool in cancer prevention and tumor-growth control. METHODS We performed an electronic-based search using PubMed updated until December 2023, summarizing the available evidence both in basic and clinical research about lipid lowering agents in NENs. RESULTS Dyslipidemia is an important aspect to be considered in NENs management, although randomized studies specifically addressing this topic are lacking, unlike other cancer types. Available data mainly regard statins, and in vitro studies have demonstrated direct antitumor effects, including antiproliferative effects in some cancers, supporting possible pleiotropic effects also in NENs, but data remain conflicting. Ezetimibe, omega 3-fatty acids, fibrates and inhibitors of proprotein convertase subtilisin/kexin type 9 (PCSK9) may enhance the regulation of lipid homeostasis, as demonstrated in other cancers. CONCLUSIONS Targeting dyslipidemia in NENs should be part of the multidisciplinary management and an integrated approach may be the best option for both metabolic and tumor control. Whether lipid lowering agents may directly contribute to tumor control remains to be confirmed with specific studies, focusing on association with other metabolic risk, disease stage and primary site.
Collapse
Affiliation(s)
- Roberta Modica
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131, Naples, Italy.
| | - Anna La Salvia
- National Center for Drug Research and Evaluation, National Institute of Health (ISS), 00161, Rome, Italy
| | - Alessia Liccardi
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131, Naples, Italy
| | - Alessia Cozzolino
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Antonella Di Sarno
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131, Naples, Italy
| | - Flaminia Russo
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189, Rome, Italy
| | - Annamaria Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131, Naples, Italy
- UNESCO Chair, Education for Health and Sustainable Development, Federico II University, 80131, Naples, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189, Rome, Italy
| |
Collapse
|
29
|
Katsuki S, Jha PK, Aikawa E, Aikawa M. The role of proprotein convertase subtilisin/kexin 9 (PCSK9) in macrophage activation: a focus on its LDL receptor-independent mechanisms. Front Cardiovasc Med 2024; 11:1431398. [PMID: 39149582 PMCID: PMC11324467 DOI: 10.3389/fcvm.2024.1431398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
Recent clinical trials demonstrated that proprotein convertase subtilisin/kexin 9 (PCSK9) inhibitors reduce cardiovascular events without affecting systemic inflammation in the patients with coronary artery disease, as determined by high sensitivity C-reactive protein (CRP) levels. However, its pro-inflammatory effects in cardiovascular disease in humans and experimental animals beyond the traditional cholesterol receptor-dependent lipid metabolism have also called attention of the scientific community. PCSK9 may target receptors associated with inflammation other than the low-density lipoprotein receptor (LDLR) and members of the LDLR family. Accumulating evidence suggests that PCSK9 promotes macrophage activation not only via lipid-dependent mechanisms, but also lipid-independent and LDLR-dependent or -independent mechanisms. In addition to dyslipidemia, PCSK9 may thus be a potential therapeutic target for various pro-inflammatory diseases.
Collapse
Affiliation(s)
- Shunsuke Katsuki
- Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Prabhash Kumar Jha
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine (MA), Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
30
|
Varghese A, Gusarov I, Gamallo-Lana B, Dolgonos D, Mankan Y, Shamovsky I, Phan M, Jones R, Gomez-Jenkins M, White E, Wang R, Jones D, Papagiannakopoulos T, Pacold ME, Mar AC, Littman DR, Nudler E. Unraveling cysteine deficiency-associated rapid weight loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605703. [PMID: 39131293 PMCID: PMC11312522 DOI: 10.1101/2024.07.30.605703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Forty percent of the US population and 1 in 6 individuals worldwide are obese, and the incidence of this disease is surging globally1,2. Various dietary interventions, including carbohydrate and fat restriction, and more recently amino acid restriction, have been explored to combat this epidemic3-6. We sought to investigate the impact of removing individual amino acids on the weight profiles of mice. Compared to essential amino acid restriction, induction of conditional cysteine restriction resulted in the most dramatic weight loss, amounting to 20% within 3 days and 30% within one week, which was readily reversed. This weight loss occurred despite the presence of substantial cysteine reserves stored in glutathione (GSH) across various tissues7. Further analysis demonstrated that the weight reduction primarily stemmed from an increase in the utilization of fat mass, while locomotion, circadian rhythm and histological appearance of multiple other tissues remained largely unaffected. Cysteine deficiency activated the integrated stress response (ISR) and NRF2-mediated oxidative stress response (OSR), which amplify each other, leading to the induction of GDF15 and FGF21, hormones associated with increased lipolysis, energy homeostasis and food aversion8-10. We additionally observed rapid tissue coenzyme A (CoA) depletion, resulting in energetically inefficient anaerobic glycolysis and TCA cycle, with sustained urinary excretion of pyruvate, orotate, citrate, α-ketoglutarate, nitrogen rich compounds and amino acids. In summary, our investigation highlights that cysteine restriction, by depleting GSH and CoA, exerts a maximal impact on weight loss, metabolism, and stress signaling compared to other amino acid restrictions. These findings may pave the way for innovative strategies for addressing a range of metabolic diseases and the growing obesity crisis.
Collapse
Affiliation(s)
- Alan Varghese
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ivan Gusarov
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Begoña Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU School of Medicine, New York, NY 10016, USA
| | - Daria Dolgonos
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Yatin Mankan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ilya Shamovsky
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Mydia Phan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Rebecca Jones
- Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Maria Gomez-Jenkins
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ 08901, USA and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Eileen White
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ 08901, USA and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Rui Wang
- Department of Biology, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Drew Jones
- Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Michael E Pacold
- Department of Radiation Oncology and Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, NY 10016, USA
| | - Adam C Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU School of Medicine, New York, NY 10016, USA
| | - Dan R Littman
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| |
Collapse
|
31
|
Mousa SO, Afifi MF, Hassuna NA, Yassa MF, Moness HM. Assessment of serum proprotein convertase subtilisin/kexin type 9 in pediatric sepsis syndrome. Sci Rep 2024; 14:15634. [PMID: 38972879 PMCID: PMC11228027 DOI: 10.1038/s41598-024-65609-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 06/21/2024] [Indexed: 07/09/2024] Open
Abstract
Sepsis is a life-threatening condition that arises when the body's response to infection causes injury to its tissues and organs. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is an enzyme released in response to the drop in cholesterol level occurring in sepsis. Our study aimed to evaluate the prognostic role of serum Proprotein convertase subtilisin/kexin type 9 (PCSK9) level in children with sepsis and severe sepsis. Sixty children were included in this study. They were divided into two groups: 30 children in the sepsis group and 30 in the severe sepsis group. Another 30 apparently healthy children were included as a control group. Blood samples were withdrawn from all included children for complete blood count (CBC), renal function tests (RFT), liver function tests (LFT), LDL-cholesterol (LDL-C), blood culture, and serum PCSK9. In this study, PCSK9 and LDL-C were higher in the two sepsis groups than in the control group (p < 0.05). They were also higher in the severe sepsis group than the sepsis group and in the non-survivors than in the survivors (p < 0.05). PCSK9 was positively correlated with length of hospital stay in surviving children (r = 0.67, p = 0.001) and had predicted significant hematological dysfunction (adjusted B = - 96.95, p = 0.03). In conclusion, the PCSK9 assay can be used as a biomarker for bad prognosis in children suffering from clinical sepsis.
Collapse
Affiliation(s)
- Suzan Omar Mousa
- Pediatric Department, Minia University Children Hospital, Faculty of Medicine, Minia University, El-Minya, 61111, Egypt.
| | - Mohamed Farouk Afifi
- Pediatric Department, Minia University Children Hospital, Faculty of Medicine, Minia University, El-Minya, 61111, Egypt
| | - Noha Anwar Hassuna
- Medical Microbiology and Immunology Department, Faculty of Medicine, Minia University, El-Minya, Egypt
| | - Michael Fekry Yassa
- Pediatric Department, Minia University Children Hospital, Faculty of Medicine, Minia University, El-Minya, 61111, Egypt
| | - Hend Mohamed Moness
- Clinical Pathology Department, Faculty of Medicine, Minia University Hospitals, Minia University, El-Minya, Egypt
| |
Collapse
|
32
|
Kajani S, Laker RC, Ratkova E, Will S, Rhodes CJ. Hepatic glucagon action: beyond glucose mobilization. Physiol Rev 2024; 104:1021-1060. [PMID: 38300523 DOI: 10.1152/physrev.00028.2023] [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: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Glucagon's ability to promote hepatic glucose production has been known for over a century, with initial observations touting this hormone as a diabetogenic agent. However, glucagon receptor agonism [when balanced with an incretin, including glucagon-like peptide 1 (GLP-1) to dampen glucose excursions] is now being developed as a promising therapeutic target in the treatment of metabolic diseases, like metabolic dysfunction-associated steatotic disease/metabolic dysfunction-associated steatohepatitis (MASLD/MASH), and may also have benefit for obesity and chronic kidney disease. Conventionally regarded as the opposing tag-team partner of the anabolic mediator insulin, glucagon is gradually emerging as more than just a "catabolic hormone." Glucagon action on glucose homeostasis within the liver has been well characterized. However, growing evidence, in part thanks to new and sensitive "omics" technologies, has implicated glucagon as more than just a "glucose liberator." Elucidation of glucagon's capacity to increase fatty acid oxidation while attenuating endogenous lipid synthesis speaks to the dichotomous nature of the hormone. Furthermore, glucagon action is not limited to just glucose homeostasis and lipid metabolism, as traditionally reported. Glucagon plays key regulatory roles in hepatic amino acid and ketone body metabolism, as well as mitochondrial turnover and function, indicating broader glucagon signaling consequences for metabolic homeostasis mediated by the liver. Here we examine the broadening role of glucagon signaling within the hepatocyte and question the current dogma, to appreciate glucagon as more than just that "catabolic hormone."
Collapse
Affiliation(s)
- Sarina Kajani
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Rhianna C Laker
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Ekaterina Ratkova
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sarah Will
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Christopher J Rhodes
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| |
Collapse
|
33
|
Wakabayashi T, Takahashi M, Okazaki H, Okazaki S, Yokote K, Tada H, Ogura M, Ishigaki Y, Yamashita S, Harada-Shiba M, on behalf of the Committee on Primary Dyslipidemia under the Research Program on Rare and Intractable Disease of the Ministry of Health, Labour and Welfare of Japan. Current Diagnosis and Management of Familial Hypobetalipoproteinemia 1. J Atheroscler Thromb 2024; 31:1005-1023. [PMID: 38710625 PMCID: PMC11224688 DOI: 10.5551/jat.rv22018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
Familial hypobetalipoproteinemia (FHBL) 1 is a rare genetic disorder with an autosomal codominant mode of inheritance and is caused by defects in the apolipoprotein (apo) B (APOB) gene that disable lipoprotein formation. ApoB proteins are required for the formation of very low-density lipoproteins (VLDLs), chylomicrons, and their metabolites. VLDLs transport cholesterol and triglycerides from the liver to the peripheral tissues, whereas chylomicrons transport absorbed lipids and fat-soluble vitamins from the intestine. Homozygous or compound heterozygotes of FHBL1 (HoFHBL1) are extremely rare, and defects in APOB impair VLDL and chylomicron secretion, which result in marked hypolipidemia with malabsorption of fat and fat-soluble vitamins, leading to various complications such as growth disorders, acanthocytosis, retinitis pigmentosa, and neuropathy. Heterozygotes of FHBL1 are relatively common and are generally asymptomatic, except for moderate hypolipidemia and possible hepatic steatosis. If left untreated, HoFHBL1 can cause severe complications and disabilities that are pathologically and phenotypically similar to abetalipoproteinemia (ABL) (an autosomal recessive disorder) caused by mutations in the microsomal triglyceride transfer protein (MTTP) gene. Although HoFHBL1 and ABL cannot be distinguished from the clinical manifestations and laboratory findings of the proband, moderate hypolipidemia in first-degree relatives may help diagnose HoFHBL1. There is currently no specific treatment for HoFHBL1. Palliative therapy including high-dose fat-soluble vitamin supplementation may prevent or delay complications. Registry research on HoFHBL1 is currently ongoing to better understand the disease burden and unmet needs of this life-threatening disease with few therapeutic options.
Collapse
Affiliation(s)
- Tetsuji Wakabayashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Manabu Takahashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Hiroaki Okazaki
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Sachiko Okazaki
- Division for Health Service Promotion, The University of Tokyo, Tokyo, Japan
| | | | - Hayato Tada
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
| | - Masatsune Ogura
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, Tokyo, Japan
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
| | - Shizuya Yamashita
- Department of Cardiology, Rinku General Medical Center, Osaka, Japan
| | - Mariko Harada-Shiba
- Cardiovascular Center, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - on behalf of the Committee on Primary Dyslipidemia under the Research Program on Rare and Intractable Disease of the Ministry of Health, Labour and Welfare of Japan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
- Division for Health Service Promotion, The University of Tokyo, Tokyo, Japan
- Chiba University, Chiba, Japan
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, Tokyo, Japan
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
- Department of Cardiology, Rinku General Medical Center, Osaka, Japan
- Cardiovascular Center, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
34
|
Mahjoubin-Tehran M, Rezaei S, Santos RD, Jamialahmadi T, Almahmeed W, Sahebkar A. Targeting PCSK9 as a key player in lipid metabolism: exploiting the therapeutic and biosensing potential of aptamers. Lipids Health Dis 2024; 23:156. [PMID: 38796450 PMCID: PMC11128129 DOI: 10.1186/s12944-024-02151-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
The degradation of low-density lipoprotein receptor (LDLR) is induced by proprotein convertase subtilisin/kexin type 9 (PCSK9), resulting in elevated plasma concentrations of LDL cholesterol. Therefore, inhibiting the interactions between PCSK9 and LDLR is a desirable therapeutic goal for managing hypercholesterolemia. Aptamers, which are RNA or single-stranded DNA sequences, can recognize their targets based on their secondary structure. Aptamers exhibit high selectivity and affinity for binding to target molecules. The systematic evolution of ligands by exponential enrichment (SELEX), a combination of biological approaches, is used to screen most aptamers in vitro. Due to their unique advantages, aptamers have garnered significant interest since their discovery and have found extensive applications in various fields. Aptamers have been increasingly utilized in the development of biosensors for sensitive detection of pathogens, analytes, toxins, drug residues, and malignant cells. Furthermore, similar to monoclonal antibodies, aptamers can serve as therapeutic tools. Unlike certain protein therapeutics, aptamers do not elicit antibody responses, and their modified sugars at the 2'-positions generally prevent toll-like receptor-mediated innate immune responses. The focus of this review is on aptamer-based targeting of PCSK9 and the application of aptamers both as biosensors and therapeutic agents.
Collapse
Affiliation(s)
- Maryam Mahjoubin-Tehran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Rezaei
- 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
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
35
|
Sinha K, Basu I, Shah Z, Shah S, Chakrabarty S. Leveraging Bidirectional Nature of Allostery To Inhibit Protein-Protein Interactions (PPIs): A Case Study of PCSK9-LDLR Interaction. J Chem Inf Model 2024; 64:3923-3932. [PMID: 38615325 DOI: 10.1021/acs.jcim.4c00294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The protein PCSK9 (proprotein convertase subtilisin/Kexin type 9) negatively regulates the recycling of LDLR (low-density lipoprotein receptor), leading to an elevated plasma level of LDL. Inhibition of PCSK9-LDLR interaction has emerged as a promising therapeutic strategy to manage hypercholesterolemia. However, the large interaction surface area between PCSK9 and LDLR makes it challenging to identify a small molecule competitive inhibitor. An alternative strategy would be to identify distal cryptic sites as targets for allosteric inhibitors that can remotely modulate PCSK9-LDLR interaction. Using several microseconds long molecular dynamics (MD) simulations, we demonstrate that on binding with LDLR, there is a significant conformational change (population shift) in a distal loop (residues 211-222) region of PCSK9. Consistent with the bidirectional nature of allostery, we establish a clear correlation between the loop conformation and the binding affinity with LDLR. Using a thermodynamic argument, we establish that the loop conformations predominantly present in the apo state of PCSK9 would have lower LDLR binding affinity, and they would be potential targets for designing allosteric inhibitors. We elucidate the molecular origin of the allosteric coupling between this loop and the LDLR binding interface in terms of the population shift in a set of salt bridges and hydrogen bonds. Overall, our work provides a general strategy toward identifying allosteric hotspots: compare the conformational ensemble of the receptor between the apo and bound states of the protein and identify distal conformational changes, if any. The inhibitors should be designed to bind and stabilize the apo-specific conformations.
Collapse
Affiliation(s)
- Krishnendu Sinha
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata 700 106, India
| | - Ipsita Basu
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata 700 106, India
| | - Zacharia Shah
- Hingez Therapeutics Inc., 8000 Towers Crescent Drive, STE 1331, Vienna, Virginia 22182, United States
| | - Salim Shah
- Hingez Therapeutics Inc., 8000 Towers Crescent Drive, STE 1331, Vienna, Virginia 22182, United States
| | - Suman Chakrabarty
- Department of Chemical and Biological Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata 700 106, India
| |
Collapse
|
36
|
Wei A, Border R, Fu B, Cullina S, Brandes N, Jang SK, Sankararaman S, Kenny E, Udler MS, Ntranos V, Zaitlen N, Arboleda V. Investigating the sources of variable impact of pathogenic variants in monogenic metabolic conditions. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.09.14.23295564. [PMID: 37745486 PMCID: PMC10516069 DOI: 10.1101/2023.09.14.23295564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Over three percent of people carry a dominant pathogenic variant, yet only a fraction of carriers develop disease. Disease phenotypes from carriers of variants in the same gene range from mild to severe. Here, we investigate underlying mechanisms for this heterogeneity: variable variant effect sizes, carrier polygenic backgrounds, and modulation of carrier effect by genetic background (marginal epistasis). We leveraged exomes and clinical phenotypes from the UK Biobank and the Mt. Sinai BioMe Biobank to identify carriers of pathogenic variants affecting cardiometabolic traits. We employed recently developed methods to study these cohorts, observing strong statistical support and clinical translational potential for all three mechanisms of variable carrier penetrance and disease severity. For example, scores from our recent model of variant pathogenicity were tightly correlated with phenotype amongst clinical variant carriers, they predicted effects of variants of unknown significance, and they distinguished gain- from loss-of-function variants. We also found that polygenic scores predicted phenotypes amongst pathogenic carriers and that epistatic effects can exceed main carrier effects by an order of magnitude.
Collapse
|
37
|
Velidakis N, Stachteas P, Gkougkoudi E, Papadopoulos C, Kadoglou NPE. Classical and Novel Lipid-Lowering Therapies for Diabetic Patients with Established Coronary Artery Disease or High Risk of Coronary Artery Disease-A Narrative Clinical Review. Pharmaceuticals (Basel) 2024; 17:568. [PMID: 38794138 PMCID: PMC11124492 DOI: 10.3390/ph17050568] [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: 03/19/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Diabetic atherosclerosis is a complex process that is characterized by diffuse and unstable lesions increasing 2-4-fold the risk of adverse cardiovascular (CV) events. Diabetic dyslipidemia has a predominant role in coronary artery disease (CAD) and has been the target of classical and emerging pharmaceutical agents with established or promising CV benefits. The aim of the present narrative review was to summarize the effects of classical and novel lipid-lowering pharmaceutical agents on lipid profile and CV outcomes in diabetic patients with established CAD or high risk of CAD. Statins remain the first-line treatment for all diabetic patients since they considerably ameliorate lipid parameters and non-lipid CV risk factors, leading to reduced CV morbidity and mortality. Complementary to statins, ezetimibe exerts lipid-lowering properties with modest but significant reductions in major adverse cardiovascular events (MACEs) and CV mortality. PCSK9 inhibitors considerably reduce LDL-C levels and lower MACEs in diabetic patients. On the other hand, fibrates may confer a very modest decline in MACE incidence, while the CV impact of omega-3 fatty acids is promising but remains questionable. Bempedoic acid and inclisiran have a potential therapeutic role in the management of diabetic dyslipidemia, but this is still not adequately documented. Given the heightened CV risk among individuals with diabetes, more decisive results would be of great importance in the utility of all these drugs.
Collapse
Affiliation(s)
- Nikolaos Velidakis
- Medical School, University of Cyprus, 2029 Nicosia, Cyprus; (N.V.); (E.G.)
| | - Panagiotis Stachteas
- Third Department of Cardiology, Aristotle University of Thessaloniki, General Hospital “Hippokration”, 541 24 Thessaloniki, Greece; (P.S.); (C.P.)
| | | | - Christodoulos Papadopoulos
- Third Department of Cardiology, Aristotle University of Thessaloniki, General Hospital “Hippokration”, 541 24 Thessaloniki, Greece; (P.S.); (C.P.)
| | | |
Collapse
|
38
|
Fassi EMA, Citarella A, Albani M, Milano EG, Legnani L, Lammi C, Silvani A, Grazioso G. PCSK9 inhibitors: a patent review 2018-2023. Expert Opin Ther Pat 2024; 34:245-261. [PMID: 38588538 DOI: 10.1080/13543776.2024.2340569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/28/2024] [Indexed: 04/10/2024]
Abstract
INTRODUCTION Proprotein convertase subtilisin/kexin 9 (PCSK9) plays a crucial role in breaking down the hepatic low-density lipoprotein receptor (LDLR), thereby influencing the levels of circulating low-density lipoprotein cholesterol (LDL-C). Consequently, inhibiting PCSK9 through suitable ligands has been established as a validated therapeutic strategy for combating hypercholesterolemia and cardiovascular diseases. AREA COVERED Patent literature claiming novel compounds inhibiting PCSK9 disclosed from 2018 to June 2023 available in the espacenet database, which contains more than 150 million patent documents from over 100 patent-granting authorities worldwide. EXPERT OPINION The undisputable beneficial influence of PCSK9 as a pharmacological target has prompted numerous private and public institutions to patent chemical frameworks as inhibitors of PCSK9. While several compounds have advanced to clinical trials for treating hypercholesterolemia, they have not completed these trials yet. These compounds must contend in a complex market where new, costly, and advanced drugs, such as monoclonal antibodies and siRNA, are prescribed instead of inexpensive and less potent statins.
Collapse
Affiliation(s)
| | - Andrea Citarella
- Dipartimento di Chimica, Università degli Studi di Milano, Milano, Italy
| | - Marco Albani
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| | - Erica Ginevra Milano
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| | - Laura Legnani
- Dipartimento di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Milano, Italy
| | - Carmen Lammi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| | - Alessandra Silvani
- Dipartimento di Chimica, Università degli Studi di Milano, Milano, Italy
| | - Giovanni Grazioso
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
39
|
Hu X, Chen F, Jia L, Long A, Peng Y, Li X, Huang J, Wei X, Fang X, Gao Z, Zhang M, Liu X, Chen YG, Wang Y, Zhang H, Wang Y. A gut-derived hormone regulates cholesterol metabolism. Cell 2024; 187:1685-1700.e18. [PMID: 38503280 DOI: 10.1016/j.cell.2024.02.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/18/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024]
Abstract
The reciprocal coordination between cholesterol absorption in the intestine and de novo cholesterol synthesis in the liver is essential for maintaining cholesterol homeostasis, yet the mechanisms governing the opposing regulation of these processes remain poorly understood. Here, we identify a hormone, Cholesin, which is capable of inhibiting cholesterol synthesis in the liver, leading to a reduction in circulating cholesterol levels. Cholesin is encoded by a gene with a previously unknown function (C7orf50 in humans; 3110082I17Rik in mice). It is secreted from the intestine in response to cholesterol absorption and binds to GPR146, an orphan G-protein-coupled receptor, exerting antagonistic downstream effects by inhibiting PKA signaling and thereby suppressing SREBP2-controlled cholesterol synthesis in the liver. Therefore, our results demonstrate that the Cholesin-GPR146 axis mediates the inhibitory effect of intestinal cholesterol absorption on hepatic cholesterol synthesis. This discovered hormone, Cholesin, holds promise as an effective agent in combating hypercholesterolemia and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoli Hu
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fengyi Chen
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Liangjie Jia
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Aijun Long
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ying Peng
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xu Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junfeng Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xueyun Wei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinlei Fang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zihua Gao
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Mengxian Zhang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiao Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Ye-Guang Chen
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Guangzhou Laboratory, Guangzhou 510005, China; School of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yan Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Yiguo Wang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
40
|
Wang Y, Tinsley B, Spolitu S, Zadroga JA, Agarwal H, Sarecha AK, Ozcan L. Geranylgeranyl isoprenoids and hepatic Rap1a regulate basal and statin-induced expression of PCSK9. J Lipid Res 2024; 65:100515. [PMID: 38309417 PMCID: PMC10910342 DOI: 10.1016/j.jlr.2024.100515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
LDL-C lowering is the main goal of atherosclerotic cardiovascular disease prevention, and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition is now a validated therapeutic strategy that lowers serum LDL-C and reduces coronary events. Ironically, the most widely used medicine to lower cholesterol, statins, has been shown to increase circulating PCSK9 levels, which limits their efficacy. Here, we show that geranylgeranyl isoprenoids and hepatic Rap1a regulate both basal and statin-induced expression of PCSK9 and contribute to LDL-C homeostasis. Rap1a prenylation and activity is inhibited upon statin treatment, and statin-mediated PCSK9 induction is dependent on geranylgeranyl synthesis and hepatic Rap1a. Accordingly, treatment of mice with a small-molecule activator of Rap1a lowered PCSK9 protein and plasma cholesterol and inhibited statin-mediated PCSK9 induction in hepatocytes. The mechanism involves inhibition of the downstream RhoA-ROCK pathway and regulation of PCSK9 at the post-transcriptional level. These data further identify Rap1a as a novel regulator of PCSK9 protein and show that blocking Rap1a prenylation through lowering geranylgeranyl levels contributes to statin-mediated induction of PCSK9.
Collapse
Affiliation(s)
- Yating Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Brea Tinsley
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Stefano Spolitu
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - John A Zadroga
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Heena Agarwal
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Amesh K Sarecha
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
41
|
Kim HJ, Park SK, Park SH, Lee YG, Park JH, Hwang JT, Chung MY. Schisandrin A in Schisandra chinensis Upregulates the LDL Receptor by Inhibiting PCSK9 Protein Stabilization in Steatotic Model. J Microbiol Biotechnol 2024; 34:425-435. [PMID: 37997262 PMCID: PMC10940739 DOI: 10.4014/jmb.2306.06049] [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: 06/28/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/25/2023]
Abstract
Schisandra chinensis extract (SCE) protects against hypocholesterolemia by inhibiting proprotein convertase subtilisin/kexin 9 (PCSK9) protein stabilization. We hypothesized that the hypocholesterolemic activity of SCE can be attributable to upregulation of the PCSK9 inhibition-associated low-density lipoprotein receptor (LDLR). Male mice were fed a low-fat diet or a Western diet (WD) containing SCE at 1% for 12 weeks. WD increased final body weight and blood LDL cholesterol levels as well as alanine transaminase and aspartate aminotransferase expression. However, SCE supplementation significantly attenuated the increase in blood markers caused by WD. SCE also attenuated WD-mediated increases in hepatic LDLR protein expression in the obese mice. In addition, SCE increased LDLR protein expression and attenuated cellular PCSK9 levels in HepG2 cells supplemented with delipidated serum (DLPS). Non-toxic concentrations of schisandrin A (SA), one of the active components of SCE, significantly increased LDLR expression and tended to decrease PCSK9 protein levels in DLPS-treated HepG2 cells. High levels of SA-mediated PCSK9 attenuation was not attributable to reduced PCSK9 gene expression, but was associated with free PCSK9 protein degradation in this cell model. Our findings show that PCSK9 secretion can be significantly reduced by SA treatment, contributing to reductions in free cholesterol levels.
Collapse
Affiliation(s)
- Hyo-Jin Kim
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
- Department of Food Biotechnology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Seon Kyeong Park
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Soo Hyun Park
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Yu Geon Lee
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Jae-Ho Park
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Jin-Taek Hwang
- Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Min-Yu Chung
- Department of Food and Nutrition, Gangseo University, Seoul 07661, Republic of Korea
| |
Collapse
|
42
|
Arsh H, Manoj Kumar FNU, Simran FNU, Tamang S, Rehman MU, Ahmed G, Khan M, Malik J, Mehmoodi A. Role of PCSK9 inhibition during the inflammatory stage of SARS-COV-2: an updated review. Ann Med Surg (Lond) 2024; 86:899-908. [PMID: 38333263 PMCID: PMC10849418 DOI: 10.1097/ms9.0000000000001601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/28/2023] [Indexed: 02/10/2024] Open
Abstract
The potential role of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition in the management of COVID-19 and other medical conditions has emerged as an intriguing area of research. PCSK9 is primarily known for its impact on cholesterol metabolism, but recent studies have unveiled its involvement in various physiological processes, including inflammation, immune regulation, and thrombosis. In this abstract, the authors review the rationale and potential implications of PCSK9 inhibition during the inflammatory stage of SARS-CoV-2 infection. Severe cases of COVID-19 are characterized by an uncontrolled inflammatory response, often referred to as the cytokine storm, which can lead to widespread tissue damage and organ failure. Preclinical studies suggest that PCSK9 inhibition could dampen this inflammatory cascade by reducing the production of pro-inflammatory cytokines. Additionally, PCSK9 inhibition may protect against acute respiratory distress syndrome (ARDS) through its effects on lung injury and inflammation. COVID-19 has been linked to an increased risk of cardiovascular complications, especially in patients with pre-existing cardiovascular conditions or dyslipidemia. PCSK9 inhibitors are known for their ability to lower low-density lipoprotein (LDL) cholesterol levels by enhancing the recycling of LDL receptors in the liver. By reducing LDL cholesterol, PCSK9 inhibition might protect blood vessels from further damage and lower the risk of atherosclerotic plaque formation. Moreover, PCSK9 inhibitors have shown potential antithrombotic effects in preclinical studies, making them a potential avenue to mitigate the increased risk of coagulation disorders and thrombotic events observed in COVID-19. While the potential implications of PCSK9 inhibition are promising, safety considerations and possible risks need careful evaluation. Hypocholesterolemia, drug interactions, and long-term safety are some of the key concerns that should be addressed. Clinical trials are needed to establish the efficacy and safety of PCSK9 inhibitors in COVID-19 patients and to determine the optimal timing and dosing for treatment. Future research opportunities encompass investigating the immune response, evaluating long-term safety, exploring combination therapy possibilities, and advancing personalized medicine approaches. Collaborative efforts from researchers, clinicians, and policymakers are essential to fully harness the therapeutic potential of PCSK9 inhibition and translate these findings into meaningful clinical outcomes.
Collapse
Affiliation(s)
- Hina Arsh
- Department of Medicine, THQ Hospital, Pasrur
| | - FNU Manoj Kumar
- Department of Medicine, Jinnah Sindh Medical College, Karachi
| | - FNU Simran
- Department of Medicine, Jinnah Sindh Medical College, Karachi
| | - Sweta Tamang
- Department of Medicine, Nepal Medical College and Teaching Hospital, Kathmandu, Nepal
| | | | - Gulfam Ahmed
- Department of Medicine, Muhammad Hospital, Lahore
| | - Masood Khan
- Department of Cardiology, Armed Forces Institute of Cardiology, Rawalpindi, Pakistan
| | - Jahanzeb Malik
- Department of Cardiovascular Medicine, Cardiovascular Analytics Group, Islamabad
| | - Amin Mehmoodi
- Department of Medicine, Ibn e Seena Hospital, Kabul, Afghanistan
| |
Collapse
|
43
|
Vroom MM, Lu H, Lewis M, Thibodeaux BA, Brooks JK, Longo MS, Ramos MM, Sahni J, Wiggins J, Boyd JD, Wang S, Ding S, Hellerstein M, Ryan V, Powchik P, Dodart JC. VXX-401, a novel anti-PCSK9 vaccine, reduces LDL-C in cynomolgus monkeys. J Lipid Res 2024; 65:100497. [PMID: 38216056 PMCID: PMC10875594 DOI: 10.1016/j.jlr.2024.100497] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/20/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of disease burden in the world and is highly correlated with chronic elevations of LDL-C. LDL-C-lowering drugs, such as statins or monoclonal antibodies against proprotein convertase subtilisin/kexin type 9 (PCSK9), are known to reduce the risk of cardiovascular diseases; however, statins are associated with limited efficacy and poor adherence to treatment, whereas PCSK9 inhibitors are only prescribed to a "high-risk" patient population or those who have failed other therapies. Based on the proven efficacy and safety profile of existing monoclonal antibodies, we have developed a peptide-based vaccine against PCSK9, VXX-401, as an alternative option to treat hypercholesterolemia and prevent ASCVD. VXX-401 is designed to trigger a safe humoral immune response against PCSK9, resulting in the production of endogenous antibodies and a subsequent 30-40% reduction in blood LDL-C. In this article, VXX-401 demonstrates robust immunogenicity and sustained serum LDL-C-lowering effects in nonhuman primates. In addition, antibodies induced by VXX-401 bind to human PCSK9 with high affinity and block the inhibitory effect of PCSK9 on LDL-C uptake in a hepatic cell model. A repeat-dose toxicity study conducted in nonhuman primates under good laboratory practices toxicity indicated a suitable safety and tolerability profile, with injection site reactions being the main findings. As a promising safe and effective LDL-C-lowering therapy, VXX-401 may represent a broadly accessible and convenient option to treat hypercholesterolemia and prevent ASCVD.
Collapse
|
44
|
Kozan DW, Farber SA. Is It Ever Wise to Edit Wild-Type Alleles? Engineered CRISPR Alleles Versus Millions of Years of Human Evolution. Arterioscler Thromb Vasc Biol 2024; 44:328-333. [PMID: 38059350 PMCID: PMC10948015 DOI: 10.1161/atvbaha.123.318069] [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] [Indexed: 12/08/2023]
Abstract
The tremendous burden of lipid metabolism diseases, coupled with recent developments in human somatic gene editing, has motivated researchers to propose population-wide somatic gene editing of PCSK9 (proprotein convertase subtilisin/kexin type 9) within the livers of otherwise healthy humans. The best-characterized molecular function of PCSK9 is its ability to regulate plasma LDL (low-density lipoprotein) levels through promoting LDL receptor degradation. Individuals with loss-of-function PCSK9 variants have lower levels of plasma LDL and reduced cardiovascular disease. Gain-of-function variants of PCSK9 are strongly associated with familial hypercholesterolemia. A new therapeutic strategy delivers CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats; CRISPR-associated protein 9) specifically to liver cells to edit the wild-type alleles of PCSK9 with the goal of producing a loss-of-function allele. This direct somatic gene editing approach is being pursued despite the availability of US Food and Drug Administration-approved PCSK9 inhibitors that lower plasma LDL levels. Here, we discuss other characterized functions of PCSK9 including its role in infection and host immunity. We explore important factors that may have contributed to the evolutionary selection of PCSK9 in several vertebrates, including humans. Until such time that more fully understand the multiple biological roles of PCSK9, the ethics of permanently editing the gene locus in healthy, wild-type populations remains highly questionable.
Collapse
Affiliation(s)
- Darby W. Kozan
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States
| | - Steven A. Farber
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
45
|
Ouyang Z, Ma M, Zhang Z, Wu H, Xue Y, Jian Y, Yin K, Yu S, Zhao C, Guo W, Gu X. Targeted Degradation of PCSK9 In Vivo by Autophagy-Tethering Compounds. J Med Chem 2024; 67:433-449. [PMID: 38112492 DOI: 10.1021/acs.jmedchem.3c01634] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Proprotein convertase subtilisin/kexin type-9 (PCSK9), a secreted protein that is synthesized and spontaneously cleaved in the endoplasmic reticulum, has become a hot lipid-lowering target chased by pharmaceutical companies in recent years. Autophagosome-tethering compounds (ATTECs) represent a new strategy to degrade targeted biomolecules. Here, we designed and synthesized PCSK9·ATTECs that are capable of lowering PCSK9 levels via autophagy in vivo, providing the first report of the degradation of a secreted protein by ATTECs. OY3, one of the PCSK9·ATTECs synthesized, shows greater potency to reduce plasma low-density lipoprotein cholesterol (LDL-C) levels and improve atherosclerosis symptoms than treatment with the same dose of simvastatin. OY3 also significantly reduces the high expression of PCSK9 caused by simvastatin administration in atherosclerosis model mice and subsequently increases the level of low-density lipoprotein receptor, promoting simvastatin to clear plasma LDL-C and alleviate atherosclerosis symptoms. Thus, we developed a new candidate compound to treat atherosclerosis that could also promote statin therapy.
Collapse
Affiliation(s)
- Zhirong Ouyang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201301, China
| | - Muye Ma
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201301, China
| | - Ziwen Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201301, China
| | - Hongyu Wu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201301, China
| | - Yongxing Xue
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201301, China
| | - Yuting Jian
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201301, China
| | - Kai Yin
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201301, China
| | - Shaokun Yu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201301, China
| | - Chunchang Zhao
- Key Laboratory for Advanced Materials and Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Wei Guo
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201301, China
| | - Xianfeng Gu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201301, China
| |
Collapse
|
46
|
Agnello F, Mauro MS, Rochira C, Landolina D, Finocchiaro S, Greco A, Ammirabile N, Raffo C, Mazzone PM, Spagnolo M, Occhipinti G, Imbesi A, Giacoppo D, Capodanno D. PCSK9 inhibitors: current status and emerging frontiers in lipid control. Expert Rev Cardiovasc Ther 2024; 22:41-58. [PMID: 37996219 DOI: 10.1080/14779072.2023.2288169] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 11/22/2023] [Indexed: 11/25/2023]
Abstract
INTRODUCTION Atherosclerotic cardiovascular disease (ASCVD) is a leading cause of global mortality, imposing substantial healthcare economic burdens. Among the modifiable risk factors, hypercholesterolemia, especially elevated low-density lipoprotein cholesterol (LDL-C), plays a pivotal role in ASCVD development. Novel therapies such as PCSK9 (Proprotein Convertase Subtilisin/Kexin type 9) inhibitors are emerging to address this concern. These inhibitors offer the potential to reduce ASCVD risk by directly targeting LDL-C levels. AREAS COVERED The article reviews the structural and functional aspects of PCSK9, highlighting its role in LDL receptor regulation. The pharmacological strategies for PCSK9 inhibition, including monoclonal antibodies, binding peptides, gene silencing, and active immunization, are explored. Clinical evidence from various trials underscores the safety and efficacy of PCSK9 inhibitors in reducing LDL-C levels and potentially improving cardiovascular outcomes. Despite these promising results, challenges such as cost-effectiveness and long-term safety considerations are addressed. EXPERT OPINION Among PCSK9 inhibitors, monoclonal antibodies represent a cornerstone. Many trials have showed their efficacy in reducing LDL-C and the risk for major adverse clinical events, revealing long-lasting effects, with special benefits particularly for statin-intolerant and familial hypercholesterolemia patients. However, long-term impacts, high costs, and patient selection necessitate further research.
Collapse
Affiliation(s)
- Federica Agnello
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Maria Sara Mauro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Carla Rochira
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Davide Landolina
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Simone Finocchiaro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Antonio Greco
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Nicola Ammirabile
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Carmelo Raffo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Placido Maria Mazzone
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Marco Spagnolo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Giovanni Occhipinti
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Antonino Imbesi
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Daniele Giacoppo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico - San Marco", University of Catania, Catania, Italy
| |
Collapse
|
47
|
Agarwal H, Tinsley B, Sarecha AK, Ozcan L. Rap1 in the Context of PCSK9, Atherosclerosis, and Diabetes. Curr Atheroscler Rep 2023; 25:931-937. [PMID: 37979063 DOI: 10.1007/s11883-023-01162-7] [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] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE OF REVIEW The focus of this article is to highlight the importance of the small GTPase, Ras-associated protein 1 (Rap1), in proprotein convertase subtilisin/kexin type 9 (PCSK9) regulation and atherosclerosis and type 2 diabetes etiology and discuss the potential therapeutic implications of targeting Rap1 in these disease areas. REVIEW FINDINGS Cardiometabolic disease characterized by obesity, glucose intolerance, dyslipidemia, and atherosclerotic cardiovascular disease remain an important cause of mortality. Evidence using mouse models of obesity and insulin resistance indicates that Rap1 deficiency increases proatherogenic PCSK9 and low-density lipoprotein cholesterol levels and predisposes these mice to develop obesity- and statin-induced hyperglycemia, which highlights Rap1's role in cardiometabolic dysfunction. Rap1 may also contribute to cardiovascular disease through its effects on vascular wall cells involved in the atherosclerosis progression. Rap1 activation, specifically in the liver, could be beneficial in the prevention of cardiometabolic perturbations, including type 2 diabetes, hypercholesterolemia, and atherosclerosis.
Collapse
Affiliation(s)
- Heena Agarwal
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Brea Tinsley
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Amesh K Sarecha
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
48
|
Cui J, Qiu Y, Kang N, Lu J, Zheng L. Correlations of PCSK9 and LDLR Gene Polymorphisms and Serum PCSK9 Levels With Atherosclerosis and Lipid Metabolism in Patients on Maintenance Hemodialysis. J Clin Pharmacol 2023; 63:1430-1437. [PMID: 37563753 DOI: 10.1002/jcph.2332] [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: 04/20/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
This study is aimed at investigating the correlations of PCSK9 and LDLR gene polymorphisms as well as serum proprotein convertase subtilisin/kexin type 9 (PCSK9) levels with atherosclerosis and lipid metabolism in patients on maintenance hemodialysis (HD). A single nucleotide polymorphism at the E670G locus of the PCSK9 gene and the rs688 locus of the LDLR gene was analyzed by polymerase chain reaction-restriction fragment length polymorphism. All study subjects' blood lipid (triglyceride [TG], total cholesterol [TC], high-density lipoprotein cholesterol [HDL-C], and low-density lipoprotein cholesterol [LDL-C]) concentrations and lipoprotein(a) and PCSK9 levels were measured. The differences in blood lipid levels between different genotypes of the E670G locus of the PCSK9 gene and the rs688 locus of the LDLR gene in patients on maintenance HD with atherosclerosis were compared. Patients on maintenance HD with atherosclerosis at the E670G locus of the PCSK9 gene AG + GG genotype had higher levels of TG, TC, LDL-C, and lipoprotein(a) than the AA genotype, and lower levels of HDL-C than the AA genotype. Patients on maintenance HD with atherosclerosis at the rs688 locus of the LDLR gene CT + TT genotype had higher levels of TG, TC, LDL-C, and lipoprotein(a) than the CC genotype, and lower levels of HDL-C than the CC genotype. Serum PCSK9 contents in patients on maintenance HD with atherosclerosis were positively correlated with lipid indices (TG, TC, LDL-C, and lipoprotein(a)) and carotid ultrasound indices (intima-media thickness and resistance index), and negatively correlated with HDL-C, maximum systolic blood flow velocity, and minimum diastolic blood flow velocity (all P < .05).
Collapse
Affiliation(s)
- Jun Cui
- Department of Nephrology, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Yuxiang Qiu
- Department of Nephrology, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Ningsu Kang
- Department of Nephrology, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Jianxun Lu
- Department of Nephrology, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Lu Zheng
- Department of Nephrology, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
49
|
Liao G, Liu W, Dai Y, Shi X, Liu Y, Li D, Xu T. Beneficial effects of flavonoids on animal models of atherosclerosis: A systematic review and meta-analysis. iScience 2023; 26:108337. [PMID: 38026172 PMCID: PMC10665821 DOI: 10.1016/j.isci.2023.108337] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/07/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Atherosclerosis is the main cause of cardiovascular diseases that seriously endanger human health. The existing treatment drugs are effective, but they have some side effects. Accumulating evidence suggests that flavonoids have attracted wide attention due to their multiple cardioprotective effects and fewer side effects. PubMed, Web of Science database, Embase, and Cochrane Library were searched for studies evaluating the effects of flavonoids against atherosclerosis. 119 studies published from August 1954 to April 2023 were included. Random-effects models were performed for synthesis. Compared with the control group, flavonoids significantly reduced longitudinal and cross-sectional plaque area. The findings indicated that flavonoids significantly reduced the concentrations of serum TC, TG, and LDL-C and increased serum HDL-C concentrations. Besides, flavonoids reduced the levels of circulating pro-inflammatory factors, including TNF-α, IL-1β, and IL-6, and increased the serum IL-10 level. This study provides evidence for the potential cardiovascular benefits of flavonoids.
Collapse
Affiliation(s)
- Gege Liao
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Wanlu Liu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Yiming Dai
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Xiangxiang Shi
- Department of Cardiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yang Liu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Tongda Xu
- Department of Cardiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
50
|
Itkonen A, Hakkola J, Rysä J. Adverse outcome pathway for pregnane X receptor-induced hypercholesterolemia. Arch Toxicol 2023; 97:2861-2877. [PMID: 37642746 PMCID: PMC10504106 DOI: 10.1007/s00204-023-03575-4] [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: 05/22/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
Pharmaceuticals and environmental contaminants contribute to hypercholesterolemia. Several chemicals known to cause hypercholesterolemia, activate pregnane X receptor (PXR). PXR is a nuclear receptor, classically identified as a sensor of chemical environment and regulator of detoxification processes. Later, PXR activation has been shown to disrupt metabolic functions such as lipid metabolism and recent findings have shown PXR activation to promote hypercholesterolemia through multiple mechanisms. Hypercholesterolemia is a major causative risk factor for atherosclerosis and greatly promotes global health burden. Metabolic disruption by PXR activating chemicals leading to hypercholesterolemia represents a novel toxicity pathway of concern and requires further attention. Therefore, we constructed an adverse outcome pathway (AOP) by collecting the available knowledge considering the molecular mechanisms for PXR-mediated hypercholesterolemia. AOPs are tools of modern toxicology for systematizing mechanistic knowledge to assist health risk assessment of chemicals. AOPs are formalized and structured linear concepts describing a link between molecular initiating event (MIE) and adverse outcome (AO). MIE and AO are connected via key events (KE) through key event relationships (KER). We present a plausible route of how PXR activation (MIE) leads to hypercholesterolemia (AO) through direct regulation of cholesterol synthesis and via activation of sterol regulatory element binding protein 2-pathway.
Collapse
Affiliation(s)
- Anna Itkonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine and Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|