1
|
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
|
2
|
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
|
3
|
Bao X, Liang Y, Chang H, Cai T, Feng B, Gordon K, Zhu Y, Shi H, He Y, Xie L. Targeting proprotein convertase subtilisin/kexin type 9 (PCSK9): from bench to bedside. Signal Transduct Target Ther 2024; 9:13. [PMID: 38185721 PMCID: PMC10772138 DOI: 10.1038/s41392-023-01690-3] [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: 02/23/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 01/09/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) has evolved as a pivotal enzyme in lipid metabolism and a revolutionary therapeutic target for hypercholesterolemia and its related cardiovascular diseases (CVD). This comprehensive review delineates the intricate roles and wide-ranging implications of PCSK9, extending beyond CVD to emphasize its significance in diverse physiological and pathological states, including liver diseases, infectious diseases, autoimmune disorders, and notably, cancer. Our exploration offers insights into the interaction between PCSK9 and low-density lipoprotein receptors (LDLRs), elucidating its substantial impact on cholesterol homeostasis and cardiovascular health. It also details the evolution of PCSK9-targeted therapies, translating foundational bench discoveries into bedside applications for optimized patient care. The advent and clinical approval of innovative PCSK9 inhibitory therapies (PCSK9-iTs), including three monoclonal antibodies (Evolocumab, Alirocumab, and Tafolecimab) and one small interfering RNA (siRNA, Inclisiran), have marked a significant breakthrough in cardiovascular medicine. These therapies have demonstrated unparalleled efficacy in mitigating hypercholesterolemia, reducing cardiovascular risks, and have showcased profound value in clinical applications, offering novel therapeutic avenues and a promising future in personalized medicine for cardiovascular disorders. Furthermore, emerging research, inclusive of our findings, unveils PCSK9's potential role as a pivotal indicator for cancer prognosis and its prospective application as a transformative target for cancer treatment. This review also highlights PCSK9's aberrant expression in various cancer forms, its association with cancer prognosis, and its crucial roles in carcinogenesis and cancer immunity. In conclusion, this synthesized review integrates existing knowledge and novel insights on PCSK9, providing a holistic perspective on its transformative impact in reshaping therapeutic paradigms across various disorders. It emphasizes the clinical value and effect of PCSK9-iT, underscoring its potential in advancing the landscape of biomedical research and its capabilities in heralding new eras in personalized medicine.
Collapse
Affiliation(s)
- Xuhui Bao
- Institute of Therapeutic Cancer Vaccines, Fudan University Pudong Medical Center, Shanghai, China.
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
- Department of Oncology, Fudan University Pudong Medical Center, Shanghai, China.
- Center for Clinical Research, Fudan University Pudong Medical Center, Shanghai, China.
- Clinical Research Center for Cell-based Immunotherapy, Fudan University, Shanghai, China.
- Department of Pathology, Duke University Medical Center, Durham, NC, USA.
| | - Yongjun Liang
- Center for Medical Research and Innovation, Fudan University Pudong Medical Center, Shanghai, China
| | - Hanman Chang
- Institute for Food Safety and Health, Illinois Institute of Technology, Chicago, IL, USA
| | - Tianji Cai
- Department of Sociology, University of Macau, Taipa, Macau, China
| | - Baijie Feng
- Department of Oncology, Fudan University Pudong Medical Center, Shanghai, China
| | - Konstantin Gordon
- Medical Institute, Peoples' Friendship University of Russia, Moscow, Russia
- A. Tsyb Medical Radiological Research Center, Obninsk, Russia
| | - Yuekun Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Zhangjiang Hi-tech Park, Shanghai, China
| | - Yundong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Liyi Xie
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Bianconi V, Cafaro G, Mannarino MR, Perricone C, Cosentini E, Bistoni O, Paltriccia R, Lombardini R, Gerli R, Pirro M, Bartoloni E. Exploring the Link between Plasma Levels of PCSK9, Immune Dysregulation and Atherosclerosis in Patients with Primary Sjögren's Syndrome. Biomolecules 2023; 13:1384. [PMID: 37759784 PMCID: PMC10527459 DOI: 10.3390/biom13091384] [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: 06/18/2023] [Revised: 08/12/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates lipid metabolism contributing to cardiovascular (CV) risk in the general population. The relationship between PCSK9 and CV risk in systemic autoimmune diseases has been poorly explored. We investigated the association between plasma PCSK9, measures of immune-inflammatory status and markers of atherosclerosis in 52 consecutive patients with primary Sjögren's syndrome (pSS) in comparison to healthy controls (HCs). Median plasma PCSK9 levels were significantly higher in pSS patients versus HCs (162 (79-255) vs. 53 (39-99) ng/mL). Significantly higher prevalence of subclinical atherosclerosis and lower of dyslipidaemia (61% vs. 85%, p = 0.042) characterized pSS patients versus HCs. In pSS, no significant correlation emerged between PCSK9 and disease activity, atherosclerosis and lipid levels. In HCs, PCSK9 significantly correlated with lipid levels and atherosclerosis. Interestingly, significantly higher PCSK9 levels were found in HCs with high-to-very-high as compared to low-to-moderate CV risk (p = 0.018) while a non-significant trend towards higher PCSK9 levels was detected in pSS patients with low-to-moderate as compared to high-to-very-high CV risk (p = 0.060). This is the first demonstration that pSS patients, despite lower prevalence of dyslipidaemia and higher CV risk profile, are characterized by a 3-fold increase in PCSK9 levels in comparison to HCs. As PCSK9 does not correlate with measures of CV risk, its role in CV morbidity in pSS needs further investigation.
Collapse
Affiliation(s)
- Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (V.B.); (M.R.M.); (E.C.); (R.P.); (R.L.); (M.P.)
| | - Giacomo Cafaro
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (G.C.); (C.P.); (O.B.); (E.B.)
| | - Massimo Raffaele Mannarino
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (V.B.); (M.R.M.); (E.C.); (R.P.); (R.L.); (M.P.)
| | - Carlo Perricone
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (G.C.); (C.P.); (O.B.); (E.B.)
| | - Elena Cosentini
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (V.B.); (M.R.M.); (E.C.); (R.P.); (R.L.); (M.P.)
| | - Onelia Bistoni
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (G.C.); (C.P.); (O.B.); (E.B.)
| | - Rita Paltriccia
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (V.B.); (M.R.M.); (E.C.); (R.P.); (R.L.); (M.P.)
| | - Rita Lombardini
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (V.B.); (M.R.M.); (E.C.); (R.P.); (R.L.); (M.P.)
| | - Roberto Gerli
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (G.C.); (C.P.); (O.B.); (E.B.)
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (V.B.); (M.R.M.); (E.C.); (R.P.); (R.L.); (M.P.)
| | - Elena Bartoloni
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy; (G.C.); (C.P.); (O.B.); (E.B.)
| |
Collapse
|
5
|
Artin J, Elsabagh YA, Rashed L, Hussein MA. Proprotein convertase subtilisin/kexin 9 (PCSK9) in patients with diffuse systemic sclerosis: A marker of disease activity and severe disease manifestations with potential therapeutic implementations. Arch Rheumatol 2023; 38:249-256. [PMID: 37680510 PMCID: PMC10481689 DOI: 10.46497/archrheumatol.2023.9638] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/30/2022] [Indexed: 09/09/2023] Open
Abstract
Objectives This study aims to investigate proprotein convertase subtilisin/kexin 9 (PCSK9) in patients with diffuse systemic sclerosis (d-SSc) and its relation to disease activity, severity and subclinical atherosclerosis in such group of patients. Patients and methods Between December 2019 and July 2021, a total of 41 patients with d-SSc (17 males, 24 females; mean age: 36.1±1.9 years; range, 19 to 58 years) and 41- age and sex-matched healthy controls (17 males, 24 females; mean age: 40.1±1.7 years; range, 20 to 60 years) were included. Disease activity and skin thickness of the patients were evaluated using the European Scleroderma Study Group (EScSG) score and modified Rodnan skin score (mRSS), respectively. Serum PCSK9 and carotid intima-media thickness (CIMT) were measured using enzyme-linked immunosorbent assay (ELISA) and Duplex ultrasound, respectively. Results Serum PCSK9 was higher in patients compared to controls (p=0.003), particularly in those with digital ulcer (DU) and interstitial lung disease (ILD) (p<0.001). The PCSK9 positively correlated with the mean pulmonary artery pressure, EScSG, mRSS, C-reactive protein (p<0.001), erythrocyte sedimentation rate (p<0.05), lipid profile, and mean CIMT (p<0.01). In the multivariate analysis, EScSG, mRSS, lipid profile, and waist circumference were significantly correlated with PCSK9. Serum PCSK9 levels of (182.6 ng/mL) had 77.7% sensitivity and 81.2% specificity for diagnosing DU versus (172.8 ng/mL) 90.1% and 73.5% for ILD (p<0.001). Conclusion Serum PCSK9 is upregulated in d-SSc with higher levels in severe disease manifestations such as DU and ILD. It is correlated well with disease activity, more severe disease manifestations, and CIMT. The PCSK9 inhibitors may be a target of therapy in diseases with premature atherosclerosis such as d-SSc regardless of its anti-cholesterol effect, at least in more severe manifestations.
Collapse
Affiliation(s)
- Joy Artin
- Rheumatology and Clinical Immunology Unit of Internal Medicine Department, Cairo University, Cairo, Egypt
| | - Yumn A Elsabagh
- Rheumatology and Clinical Immunology Unit of Internal Medicine Department, Cairo University, Cairo, Egypt
| | - Laila Rashed
- Medical Biochemistry and Molecular Biology, Cairo University, Cairo, Egypt
| | - Mohamed A Hussein
- Rheumatology and Clinical Immunology Unit of Internal Medicine Department, Cairo University, Cairo, Egypt
| |
Collapse
|
6
|
An D, Zhai D, Wan C, Yang K. The role of lipid metabolism in cancer radioresistance. Clin Transl Oncol 2023:10.1007/s12094-023-03134-4. [PMID: 37079212 DOI: 10.1007/s12094-023-03134-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/24/2023] [Indexed: 04/21/2023]
Abstract
Radiotherapy is one of the main therapies for cancer. The process leading to radioresistance is still not fully understood. Cancer radiosensitivity is related to the DNA reparation of cancer cells and the tumor microenvironment (TME), which supports cancer cell survival. Factors that affect DNA reparation and the TME can directly or indirectly affect the radiosensitivity of cancer. Recent studies have shown that lipid metabolism in cancer cells, which is involved in the stability of cell membrane structure, energy supply and signal transduction of cancer cells, can also affect the phenotype and function of immune cells and stromal cells in the TME. In this review, we discussed the effects of lipid metabolism on the radiobiological characteristics of cancer cells and the TME. We also summarized recent advances in targeted lipid metabolism as a radiosensitizer and discussed how these scientific findings could be translated into clinical practice to improve the radiosensitivity of cancer.
Collapse
Affiliation(s)
- Dandan An
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Danyi Zhai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
7
|
Arnold N, Koenig W. PCSK9 Inhibitor Wars: How Does Inclisiran Fit in with Current Monoclonal Antibody Inhibitor Therapy? Considerations for Patient Selection. Curr Cardiol Rep 2022; 24:1657-1667. [PMID: 36087240 PMCID: PMC9729136 DOI: 10.1007/s11886-022-01782-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Treatment of dyslipidemia represents one of the most crucial strategies to reduce risk of atherosclerotic cardiovascular (CV) disease (ASCVD). In this review, we critically summarize our knowledge on emerging cholesterol-lowering therapy, targeting PCSK9, paying particular attention on treatment allocation of two drug groups, currently available for clinical use, namely, anti-PCSK9 monoclonal antibodies (mAbs) and inclisiran, a first-in-class small interfering RNA against PCSK9. RECENT FINDINGS Although both drug classes show a pronounced, but fairly similar reduction in LDL-cholesterol, their long-term safety is still unknown. Compared to mAbs, inclisiran has a more favorable dosing regimen with biannual application that might improve therapeutic adherence significantly. However, a CV outcome trial (CVOT) for inclisiran is still missing. If inclisiran will be safe and effective in ongoing/future CVOTs, it has a huge potential to overcome medication non-compliance, thereby providing a powerful therapeutic option to decrease the burden of ASCVD.
Collapse
Affiliation(s)
- Natalie Arnold
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Wolfgang Koenig
- German Heart Center, Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| |
Collapse
|
8
|
Ayoub C, Azar Y, Maddah D, Ghaleb Y, Elbitar S, Abou-Khalil Y, Jambart S, Varret M, Boileau C, El Khoury P, Abifadel M. Low circulating PCSK9 levels in LPL homozygous children with chylomicronemia syndrome in a syrian refugee family in Lebanon. Front Genet 2022; 13:961028. [PMID: 36061186 PMCID: PMC9437297 DOI: 10.3389/fgene.2022.961028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/26/2022] [Indexed: 12/17/2022] Open
Abstract
Familial chylomicronemia syndrome is a rare autosomal recessive disorder of lipoprotein metabolism characterized by the presence of chylomicrons in fasting plasma and an important increase in plasma triglycerides (TG) levels that can exceed 22.58 mmol/l. The disease is associated with recurrent episodes of abdominal pain and pancreatitis, eruptive cutaneous xanthomatosis, lipemia retinalis, and hepatosplenomegaly. A consanguineous Syrian family who migrated to Lebanon was referred to our laboratory after perceiving familial chylomicronemia syndrome in two children. The LPL and PCSK9 genes were sequenced and plasma PCSK9 levels were measured. Sanger sequencing of the LPL gene revealed the presence of the p.(Val227Phe) pathogenic variant in exon 5 at the homozygous state in the two affected children, and at the heterozygous state in the other recruited family members. Interestingly, PCSK9 levels in homozygous carriers of the p.(Val227Phe) were ≈50% lower than those in heterozygous carriers of the variant (p-value = 0.13) and ranged between the 5th and the 7.5th percentile of PCSK9 levels in a sample of Lebanese children of approximately the same age group. Moreover, this is the first reported case of individuals carrying simultaneously an LPL pathogenic variant and PCSK9 variants, the L10 and L11 leucine insertion, which can lower and raise low-density lipoprotein cholesterol (LDL-C) levels respectively. TG levels fluctuated concomitantly between the two children, were especially high following the migration from a country to another, and were reduced under a low-fat diet. This case is crucial to raise public awareness on the risks of consanguineous marriages to decrease the emergence of inherited autosomal recessive diseases. It also highlights the importance of the early diagnosis and management of these diseases to prevent serious complications, such as recurrent pancreatitis in the case of familial hyperchylomicronemia.
Collapse
Affiliation(s)
- Carine Ayoub
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie- Santé, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Yara Azar
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie- Santé, Saint Joseph University of Beirut, Beirut, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM, Paris Cité University and Sorbonne Paris Nord University, Paris, France
| | - Dina Maddah
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Youmna Ghaleb
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie- Santé, Saint Joseph University of Beirut, Beirut, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM, Paris Cité University and Sorbonne Paris Nord University, Paris, France
| | - Sandy Elbitar
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie- Santé, Saint Joseph University of Beirut, Beirut, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM, Paris Cité University and Sorbonne Paris Nord University, Paris, France
| | - Yara Abou-Khalil
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie- Santé, Saint Joseph University of Beirut, Beirut, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM, Paris Cité University and Sorbonne Paris Nord University, Paris, France
| | - Selim Jambart
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Mathilde Varret
- Laboratory for Vascular Translational Science (LVTS), INSERM, Paris Cité University and Sorbonne Paris Nord University, Paris, France
| | - Catherine Boileau
- Laboratory for Vascular Translational Science (LVTS), INSERM, Paris Cité University and Sorbonne Paris Nord University, Paris, France
- Genetic Department, AP-HP, Hôpital Bichat, Paris, France
| | - Petra El Khoury
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie- Santé, Saint Joseph University of Beirut, Beirut, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM, Paris Cité University and Sorbonne Paris Nord University, Paris, France
| | - Marianne Abifadel
- Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie- Santé, Saint Joseph University of Beirut, Beirut, Lebanon
- Laboratory for Vascular Translational Science (LVTS), INSERM, Paris Cité University and Sorbonne Paris Nord University, Paris, France
- *Correspondence: Marianne Abifadel,
| |
Collapse
|
9
|
Ponticelli C, Citterio F. Non-Immunologic Causes of Late Death-Censored Kidney Graft Failure: A Personalized Approach. J Pers Med 2022; 12:1271. [PMID: 36013220 PMCID: PMC9410103 DOI: 10.3390/jpm12081271] [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: 04/01/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022] Open
Abstract
Despite continuous advances in surgical and immunosuppressive protocols, the long-term survival of transplanted kidneys is still far from being satisfactory. Antibody-mediated rejection, recurrent autoimmune diseases, and death with functioning graft are the most frequent causes of late-kidney allograft failure. However, in addition to these complications, a number of other non-immunologic events may impair the function of transplanted kidneys and directly or indirectly lead to their failure. In this narrative review, we will list and discuss the most important nonimmune causes of late death-censored kidney graft failure, including quality of the donated kidney, adherence to prescriptions, drug toxicities, arterial hypertension, dyslipidemia, new onset diabetes mellitus, hyperuricemia, and lifestyle of the renal transplant recipient. For each of these risk factors, we will report the etiopathogenesis and the potential consequences on graft function, keeping in mind that in many cases, two or more risk factors may negatively interact together.
Collapse
Affiliation(s)
| | - Franco Citterio
- Renal Transplant Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
10
|
Cardiovascular Risk after Kidney Transplantation: Causes and Current Approaches to a Relevant Burden. J Pers Med 2022; 12:jpm12081200. [PMID: 35893294 PMCID: PMC9329988 DOI: 10.3390/jpm12081200] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/11/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Background. Cardiovascular disease is a frequent complication after kidney transplantation and represents the leading cause of mortality in this population. Material and Methods. We searched for the relevant articles in the National Institutes of Health library of medicine, transplant, cardiologic and nephrological journals. Results. The pathogenesis of cardiovascular disease in kidney transplant is multifactorial. Apart from non-modifiable risk factors, such as age, gender, genetic predisposition and ethnicity, several traditional and non-traditional modifiable risk factors contribute to its development. Traditional factors, such as diabetes, hypertension and dyslipidemia, may be present before and may worsen after transplantation. Immunosuppressants and impaired graft function may strongly influence the exacerbation of these comorbidities. However, in the last years, several studies showed that many other cardiovascular risk factors may be involved in kidney transplantation, including hyperuricemia, inflammation, low klotho and elevated Fibroblast Growth Factor 23 levels, deficient levels of vitamin D, vascular calcifications, anemia and poor physical activity and quality of life. Conclusions. The timely and effective treatment of time-honored and recently discovered modifiable risk factors represent the basis of the prevention of cardiovascular complications in kidney transplantation. Reduction of cardiovascular risk can improve the life expectancy, the quality of life and the allograft function and survival.
Collapse
|
11
|
Zhu W, Ding C, Huang P, Ran J, Lian P, Tang Y, Dai W, Huang X. Metformin Ameliorates Hepatic Steatosis induced by olanzapine through inhibiting LXRα/PCSK9 pathway. Sci Rep 2022; 12:5639. [PMID: 35379885 PMCID: PMC8979948 DOI: 10.1038/s41598-022-09610-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/17/2022] [Indexed: 11/09/2022] Open
Abstract
Studies have confirmed that olanzapine, the mainstay treatment for schizophrenia, triggers metabolic diseases, including non-alcoholic fatty liver disease (NAFLD). However, the etiology of olanzapine-induced NAFLD is poorly understood. Proprotein convertase subtilisin kexin type 9 (PCSK9) is involved in NAFLD pathogenesis, and metformin can significantly decrease circulating PCSK9. The purpose of this study was to investigate the role of PCSK9 and explore the therapeutic effect of metformin for olanzapine-associated NAFLD. Olanzapine significantly upregulated PCSK9 and promoted lipid accumulation in mouse livers and HepG2 and AML12 cells. Metformin ameliorated these pathological alterations. PCSK9 upstream regulator liver X receptor α (LXRα) was significantly upregulated in olanzapine-induced NAFLD. LXRα antagonist treatment and LXRα overexpression resulted in a decrease and increase of PCSK9, respectively. Hepatic lipogenesis-associated genes FAS and SCD1 were significantly upregulated in olanzapine-induced NAFLD mice and HepG2 cells overexpressing PCSK9, and genes related to lipid β-oxidation (SCAD and PPARα) were downregulated, while metformin reversed these changes. In addition, we found that LXRα overexpression compromised the effect of metformin on PCSK9 levels and intracellular lipid droplet formation. Taken together, our findings suggest that olanzapine enhances hepatic PCSK9 expression by upregulating LXRα, thereby increasing FAS and SCD1 expression as well as decreasing SCAD and PPARα, and promoting lipid accumulation, and, subsequently, NAFLD, which is ameliorated by metformin.
Collapse
Affiliation(s)
- Wenqiang Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Chen Ding
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Piaopiao Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Juanli Ran
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Pingan Lian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yaxin Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Wen Dai
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.,Department of Medicine, Columbia University Medical Center, New York, USA
| | - Xiansheng Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
12
|
Derežanin L, Blažytė A, Dobrynin P, Duchêne DA, Grau JH, Jeon S, Kliver S, Koepfli KP, Meneghini D, Preick M, Tomarovsky A, Totikov A, Fickel J, Förster DW. Multiple types of genomic variation contribute to adaptive traits in the mustelid subfamily Guloninae. Mol Ecol 2022; 31:2898-2919. [PMID: 35334142 DOI: 10.1111/mec.16443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/06/2022] [Accepted: 03/14/2022] [Indexed: 11/28/2022]
Abstract
Species of the mustelid subfamily Guloninae inhabit diverse habitats on multiple continents, and occupy a variety of ecological niches. They differ in feeding ecologies, reproductive strategies and morphological adaptations. To identify candidate loci associated with adaptations to their respective environments, we generated a de novo assembly of the tayra (Eira barbara), the earliest diverging species in the subfamily, and compared this with the genomes available for the wolverine (Gulo gulo) and the sable (Martes zibellina). Our comparative genomic analyses included searching for signs of positive selection, examining changes in gene family sizes, as well as searching for species-specific structural variants (SVs). Among candidate loci associated with phenotypic traits, we observed many related to diet, body condition and reproduction. For example, for the tayra, which has an atypical gulonine reproductive strategy of aseasonal breeding, we observe species-specific changes in many pregnancy-related genes. For the wolverine, a circumpolar hypercarnivore that must cope with seasonal food scarcity, we observed many changes in genes associated with diet and body condition. All types of genomic variation examined (single nucleotide polymorphisms, gene family expansions, structural variants) contributed substantially to the identification of candidate loci. This strongly argues for consideration of variation other than single nucleotide polymorphisms in comparative genomics studies aiming to identify loci of adaptive significance.
Collapse
Affiliation(s)
- Lorena Derežanin
- Leibniz Institute for Zoo and Wildlife Research (IZW, Alfred Kowalke Straße 17, 10315, Berlin, Germany
| | - Asta Blažytė
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST, Ulsan, 44919, Republic of Korea
| | - Pavel Dobrynin
- Computer Technologies Laboratory, ITMO University, 49 Kronverkskiy Pr, 197101, Saint Petersburg, Russia
| | - David A Duchêne
- Center for Evolutionary Hologenomics, The GLOBE Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, 1353, Copenhagen, Denmark
| | - José Horacio Grau
- amedes Genetics, amedes Medizinische Dienstleistungen GmbH, Jägerstr. 61, 10117, Berlin, Germany
| | - Sungwon Jeon
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST, Ulsan, 44919, Republic of Korea.,Clinomics Inc, Ulsan, 44919, Republic of Korea
| | - Sergei Kliver
- Institute of Molecular and Cellular Biology, SB RAS, 8/2 Acad. Lavrentiev Ave, Novosibirsk, 630090, Russia
| | - Klaus-Peter Koepfli
- Computer Technologies Laboratory, ITMO University, 49 Kronverkskiy Pr, 197101, Saint Petersburg, Russia.,Smithsonian-Mason School of Conservation, 1500 Remount Road, Front Royal, VA, 22630, USA.,Smithsonian Conservation Biology Institute, Center for Species Survival, National Zoological Park, 1500 Remount Road, Front Royal, VA, 22630, USA
| | - Dorina Meneghini
- Leibniz Institute for Zoo and Wildlife Research (IZW, Alfred Kowalke Straße 17, 10315, Berlin, Germany
| | - Michaela Preick
- Institute for Biochemistry and Biology, Faculty of Mathematics and Natural Sciences, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, OT, Germany
| | - Andrey Tomarovsky
- Computer Technologies Laboratory, ITMO University, 49 Kronverkskiy Pr, 197101, Saint Petersburg, Russia.,Institute of Molecular and Cellular Biology, SB RAS, 8/2 Acad. Lavrentiev Ave, Novosibirsk, 630090, Russia.,Novosibirsk State University, 1 Pirogova str, Novosibirsk, 630090, Russia
| | - Azamat Totikov
- Computer Technologies Laboratory, ITMO University, 49 Kronverkskiy Pr, 197101, Saint Petersburg, Russia.,Institute of Molecular and Cellular Biology, SB RAS, 8/2 Acad. Lavrentiev Ave, Novosibirsk, 630090, Russia.,Novosibirsk State University, 1 Pirogova str, Novosibirsk, 630090, Russia
| | - Jörns Fickel
- Leibniz Institute for Zoo and Wildlife Research (IZW, Alfred Kowalke Straße 17, 10315, Berlin, Germany.,Institute for Biochemistry and Biology, Faculty of Mathematics and Natural Sciences, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, OT, Germany
| | - Daniel W Förster
- Leibniz Institute for Zoo and Wildlife Research (IZW, Alfred Kowalke Straße 17, 10315, Berlin, Germany
| |
Collapse
|
13
|
Maligłówka M, Kosowski M, Hachuła M, Cyrnek M, Bułdak Ł, Basiak M, Bołdys A, Machnik G, Bułdak RJ, Okopień B. Insight into the Evolving Role of PCSK9. Metabolites 2022; 12:metabo12030256. [PMID: 35323699 PMCID: PMC8951079 DOI: 10.3390/metabo12030256] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is the last discovered member of the family of proprotein convertases (PCs), mainly synthetized in hepatic cells. This serine protease plays a pivotal role in the reduction of the number of low-density lipoprotein receptors (LDLRs) on the surface of hepatocytes, which leads to an increase in the level of cholesterol in the blood. This mechanism and the fact that gain of function (GOF) mutations in PCSK9 are responsible for causing familial hypercholesterolemia whereas loss-of-function (LOF) mutations are associated with hypocholesterolemia, prompted the invention of drugs that block PCSK9 action. The high efficiency of PCSK9 inhibitors (e.g., alirocumab, evolocumab) in decreasing cardiovascular risk, pleiotropic effects of other lipid-lowering drugs (e.g., statins) and the multifunctional character of other proprotein convertases, were the cause for proceeding studies on functions of PCSK9 beyond cholesterol metabolism. In this article, we summarize the current knowledge on the roles that PCSK9 plays in different tissues and perspectives for its clinical use.
Collapse
Affiliation(s)
- Mateusz Maligłówka
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
- Correspondence:
| | - Michał Kosowski
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Marcin Hachuła
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Marcin Cyrnek
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Łukasz Bułdak
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Marcin Basiak
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Aleksandra Bołdys
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Grzegorz Machnik
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Rafał Jakub Bułdak
- Institute of Medical Sciences, University of Opole, 45-040 Opole, Poland;
| | - Bogusław Okopień
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| |
Collapse
|
14
|
Liu H, Pietersz G, Peter K, Wang X. Nanobiotechnology approaches for cardiovascular diseases: site-specific targeting of drugs and nanoparticles for atherothrombosis. J Nanobiotechnology 2022; 20:75. [PMID: 35135581 PMCID: PMC8822797 DOI: 10.1186/s12951-022-01279-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/21/2022] [Indexed: 02/18/2023] Open
Abstract
Atherosclerosis and atherothrombosis, the major contributors to cardiovascular diseases (CVDs), represent the leading cause of death worldwide. Current pharmacological therapies have been associated with side effects or are insufficient at halting atherosclerotic progression effectively. Pioneering work harnessing the passive diffusion or endocytosis properties of nanoparticles and advanced biotechnologies in creating recombinant proteins for site-specific delivery have been utilized to overcome these limitations. Since CVDs are complex diseases, the most challenging aspect of developing site-specific therapies is the identification of an individual and unique antigenic epitope that is only expressed in lesions or diseased areas. This review focuses on the pathological mechanism of atherothrombosis and discusses the unique targets that are important during disease progression. We review recent advances in site-specific therapy using novel targeted drug-delivery and nanoparticle-carrier systems. Furthermore, we explore the limitations and future perspectives of site-specific therapy for CVDs.
Collapse
Affiliation(s)
- Haikun Liu
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Geoffrey Pietersz
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Burnet Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia.,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia. .,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. .,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia. .,Department of Medicine, Monash University, Melbourne, VIC, Australia. .,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
15
|
Coppinger C, Movahed MR, Azemawah V, Peyton L, Gregory J, Hashemzadeh M. A Comprehensive Review of PCSK9 Inhibitors. J Cardiovasc Pharmacol Ther 2022; 27:10742484221100107. [PMID: 35593194 DOI: 10.1177/10742484221100107] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the United States and worldwide. A major risk factor for this condition is increased serum low-density lipoprotein cholesterol (LDL-C) levels for which statins have been successful in reducing serum LDL-C to healthy concentrations. However, patients who are statin intolerant or those who do not achieve their treatment goals while on high-intensity statin therapy, such as those with familial hypercholesterolemia, remain at risk. With the discovery of PCSK9 inhibitors, the ability to provide more aggressive treatment for patients with homozygous and heterozygous familial hypercholesterolemia has increased. Ezetimibe reduces LDL-C by 15%-20% when combined with statin.2,3 Protein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have been found to achieve profound reductions in LDL-C (54%-74%) when added to statins. They have shown dramatic effects at lowering major adverse cardiovascular events (MACE) in high-risk patients4 with LDL-C levels ≥70 mg/dL and can be used in populations that are statin intolerant or not at goal levels with maximally tolerated statin therapy. PCSK9 inhibitors also produce minimal side effects. Myopathy, a common side effect for patients on statins, has been rare in patients on PCSK9 inhibitors. Randomized trials have shown that reduction in LDL-C has translated to clinical benefits even in patients who have not achieved their LDL-C target.
Collapse
Affiliation(s)
- Caroline Coppinger
- 8040Pima Community College, Tucson, AZ, USA.,42283University of Arizona, Tucson, AZ, USA
| | - Mohammad Reza Movahed
- 42283University of Arizona, Tucson, AZ, USA.,42283University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Veronica Azemawah
- 8040Pima Community College, Tucson, AZ, USA.,42283University of Arizona, Tucson, AZ, USA
| | - Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, 12270Mayo Clinic College of Medicine, Rochester, MN, USA
| | - James Gregory
- 8040Pima Community College, Tucson, AZ, USA.,42283University of Arizona, Tucson, AZ, USA
| | - Mehrnoosh Hashemzadeh
- 8040Pima Community College, Tucson, AZ, USA.,42283University of Arizona College of Medicine, Phoenix, AZ, USA
| |
Collapse
|
16
|
Ponticelli C, Reggiani F, Moroni G. Old and New Calcineurin Inhibitors in Lupus Nephritis. J Clin Med 2021; 10:jcm10214832. [PMID: 34768354 PMCID: PMC8584552 DOI: 10.3390/jcm10214832] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 12/27/2022] Open
Abstract
Calcineurin inhibitors (CNIs) are drugs that inhibit calcineurin, a key phosphatase that dephosphorylates a transcription factor called the nuclear factor of activated T cells (NFAT), allowing its translocation into the nucleus of quiescent T cells. In the nucleus, NFAT activates interleukin 2, which stimulates the proliferation and differentiation of T-cells. CNIs can also stabilize the actin cytoskeleton of podocytes reducing proteinuria. Thanks to these characteristics, CNIs have been often used in the treatment of autoimmune diseases. However, the therapeutic index of CNIs is narrow, and their interactions with other drugs can increase toxicity or reduce efficacy. In lupus nephritis, cyclosporine and tacrolimus have been used both in induction and maintenance therapies. Observational studies and randomized controlled trials showed that both cyclosporine and tacrolimus can increase efficacy. Tolerance is satisfactory if low doses are used and the patient is carefully monitored. More recently, a new CNI, called voclosporin (VCS), has been approved by the Food and Drug Administration for use in lupus nephritis. VCS offers potential advantages over other CNIs. In two large multiethnic trials, VCS was not associated with adverse renal and metabolic events and obtained positive results despite a novel and rapid corticosteroid tapering regime.
Collapse
Affiliation(s)
- Claudio Ponticelli
- Nephrology Division, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence:
| | - Francesco Reggiani
- Nephrology and Dialysis Division, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Milan, Italy; (F.R.); (G.M.)
| | - Gabriella Moroni
- Nephrology and Dialysis Division, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Milan, Italy; (F.R.); (G.M.)
| |
Collapse
|
17
|
Oleaga C, Shapiro MD, Hay J, Mueller PA, Miles J, Huang C, Friz E, Tavori H, Toth PP, Wójcik C, Warden BA, Purnell JQ, Duell PB, Pamir N, Fazio S. Hepatic Sensing Loop Regulates PCSK9 Secretion in Response to Inhibitory Antibodies. J Am Coll Cardiol 2021; 78:1437-1449. [PMID: 34593126 PMCID: PMC8486917 DOI: 10.1016/j.jacc.2021.07.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/08/2021] [Accepted: 07/26/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Monoclonal antibodies against proprotein convertase subtilisin/kexin type 9 (PCSK9i) lower LDL-C by up to 60% and increase plasma proprotein convertase subtilisin/kexin type 9 (PCSK9) levels by 10-fold. OBJECTIVES The authors studied the reasons behind the robust increase in plasma PCSK9 levels by testing the hypothesis that mechanisms beyond clearance via the low-density lipoprotein receptor (LDLR) contribute to the regulation of cholesterol homeostasis. METHODS In clinical cohorts, animal models, and cell-based studies, we measured kinetic changes in PCSK9 production and clearance in response to PCSK9i. RESULTS In a patient cohort receiving PCSK9i therapy, plasma PCSK9 levels rose 11-fold during the first 3 months and then plateaued for 15 months. In a cohort of healthy volunteers, a single injection of PCSK9i increased plasma PCSK9 levels within 12 hours; the rise continued for 9 days until it plateaued at 10-fold above baseline. We recapitulated the rapid rise in PCSK9 levels in a mouse model, but only in the presence of LDLR. In vivo turnover and in vitro pulse-chase studies identified 2 mechanisms contributing to the rapid increase in plasma PCSK9 levels in response to PCSK9i: 1) the expected delayed clearance of the antibody-bound PCSK9; and 2) the unexpected post-translational increase in PCSK9 secretion. CONCLUSIONS PCSK9 re-entry to the liver via LDLR triggers a sensing loop regulating PCSK9 secretion. PCSK9i therapy enhances the secretion of PCSK9, an effect that contributes to the increased plasma PCSK9 levels in treated subjects.
Collapse
Affiliation(s)
- Carlota Oleaga
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Michael D Shapiro
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Joshua Hay
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Paul A Mueller
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Joshua Miles
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Cecilia Huang
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Emily Friz
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Hagai Tavori
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Peter P Toth
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University, and School of Medicine, Baltimore, Maryland, USA; CGH Medical Center, Sterling, Illinois, USA
| | - Cezary Wójcik
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Bruce A Warden
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Jonathan Q Purnell
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - P Barton Duell
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Nathalie Pamir
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA.
| | - Sergio Fazio
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
18
|
Kamaruddin NN, Hajri NA, Andriani Y, Abdul Manan AF, Tengku Muhammad TS, Mohamad H. Acanthaster planci Inhibits PCSK9 and Lowers Cholesterol Levels in Rats. Molecules 2021; 26:5094. [PMID: 34443682 PMCID: PMC8398678 DOI: 10.3390/molecules26165094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is the main cause of cardiovascular diseases which in turn, lead to the highest number of mortalities globally. This pathophysiological condition is developed due to a constant elevated level of plasma cholesterols. Statin is currently the widely used treatment in reducing the level of cholesterols, however, it may cause adverse side effects. Therefore, there is an urgent need to search for new alternative treatment. PCSK9 is an enzyme responsible in directing LDL-receptor (LDL-R)/LDL-cholesterols (LDL-C) complex to lysosomal degradation, preventing the receptor from recycling back to the surface of liver cells. Therefore, PCSK9 offers a potential target to search for small molecule inhibitors which inhibit the function of this enzyme. In this study, a marine invertebrate Acanthaster planci, was used to investigate its potential in inhibiting PCSK9 and lowering the levels of cholesterols. Cytotoxicity activity of A. planci on human liver HepG2 cells was carried out using the MTS assay. It was found that methanolic extract and fractions did not exhibit cytotoxicity effect on HepG2 cell line with IC50 values of more than 30 µg/mL. A compound deoxythymidine also did not exert any cytotoxicity activity with IC50 value of more than 4 µg/mL. Transient transfection and luciferase assay were conducted to determine the effects of A. planci on the transcriptional activity of PCSK9 promoter. Methanolic extract and Fraction 2 (EF2) produced the lowest reduction in PCSK9 promoter activity to 70 and 20% of control at 12.5 and 6.25 μg/mL, respectively. In addition, deoxythymidine also decreased PCSK9 promoter activity to the lowest level of 60% control at 3.13 μM. An in vivo study using Sprague Dawley rats demonstrated that 50 and 100 mg/kg of A. planci methanolic extract reduced the total cholesterols and LDL-C levels to almost similar levels of untreated controls. The level of serum glutamate oxalate transaminase (SGOT) and serum glutamate pyruvate transaminase (SGPT) showed that the administration of the extract did not produce any toxicity effect and cause any damage to rat liver. The results strongly indicate that A. planci produced a significant inhibitory activity on PCSK9 gene expression in HepG2 cells which may be responsible for inducing the uptake of cholesterols by liver, thus, reducing the circulating levels of total cholesterols and LDL-C. Interestingly, A. planci also did show any adverse hepato-cytotoxicity and toxic effects on liver. Thus, this study strongly suggests that A. planci has a vast potential to be further developed as a new class of therapeutic agent in lowering the blood cholesterols and reducing the progression of atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | - Tengku Sifzizul Tengku Muhammad
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Nerus 21030, Malaysia; (N.N.K.); (N.A.H.); (Y.A.); (A.F.A.M.)
| | - Habsah Mohamad
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Nerus 21030, Malaysia; (N.N.K.); (N.A.H.); (Y.A.); (A.F.A.M.)
| |
Collapse
|
19
|
Kajingulu FPM, Lepira FB, Nkodila AN, Makulo JRR, Mokoli VM, Ekulu PM, Bukabau JB, Nlandu YM, Longo AL, Nseka NM, Sumaili EK. Circulating Proprotein Convertase Subtilisin/Kexin Type 9 Levels Predict Future Cardiovascular Event Risks in Hemodialyzed Black African Patients. Rambam Maimonides Med J 2021; 12:RMMJ.10443. [PMID: 34270402 PMCID: PMC8284989 DOI: 10.5041/rmmj.10443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
CONTEXT AND OBJECTIVE Cardiovascular diseases are the leading cause of mortality in patients. In this context, proprotein convertase subtilisin/kexin type 9 (PCSK9) appears to be the new biomarker identified as interfering in lipid homeostasis. This study aimed to investigate the association between PCSK9, dyslipidemia, and future risk of cardiovascular events in a population of black Africans. METHODS A cross-sectional study was conducted between August 2016 and July 2020 in six hemodialysis centers in the city of Kinshasa, Democratic Republic of the Congo. Serum PCSK9 was measured by ELISA; lipid levels of 251 chronic kidney disease grade 5 (CKD G5) hemodialysis patients and the Framingham predictive instrument were used for predicting cardiac events. RESULTS Total cholesterol (TC), low-density lipoprotein cholesterol (LDL-c), and triglycerides (TG) were significantly increased in the tertile with the highest PCSK9. By contrast, high-density lipoprotein cholesterol (HDL-c) was significantly decreased in the same tertile. A strong positive and significant correlation was found between PCSK9 and TC, TG, and LDL-c. Negative and significant correlation was observed between PCSK9 and HDL-c. The levels of PCSK9, smoking, overweight, and atherogenic dyslipidemia were associated with future risks for cardiovascular events in univariate analysis. After adjustment, all these variables persisted as independent determinants of future risk for cardiovascular events. The probability of having a cardiovascular event in this population was independently associated with PCSK9 levels. Compared to the patients in the lowest PCSK9 tertile, patients with PCSK9 levels in the middle (aOR 5.9, 95% CI 2.06-17.3, P<0.001) and highest tertiles (aOR 8.9, 95% CI 3.02-25.08, P<0.001) presented a greater risk of cardiac event. CONCLUSION Increased PCSK9 serum levels are associated with higher levels of TC, LDL-c, and TG and lower levels of HDL-c in black African hemodialysis patients. Serum PCSK9 levels in these patients predict increased risk of cardiovascular events, independent of traditional potential confounders.
Collapse
Affiliation(s)
- François-Pantaléon Musungayi Kajingulu
- Department of Internal Medicine, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
- To whom correspondence should be addressed. E-mail:
| | - François Bompeka Lepira
- Department of Internal Medicine, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
| | - Aliocha Natuhoyila Nkodila
- Faculty of Family Medicine and Primary Care, Protestant University of Congo, Kinshasa, Democratic Republic of the Congo
| | - Jean-Robert Rissassy Makulo
- Department of Internal Medicine, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
| | - Vieux Momeme Mokoli
- Department of Internal Medicine, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
| | - Pepe Mfutu Ekulu
- Department of Pediatrics, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
| | - Justine Busanga Bukabau
- Department of Internal Medicine, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
| | - Yannick Mayamba Nlandu
- Department of Internal Medicine, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
| | - Augustin Luzayadio Longo
- Department of Internal Medicine, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
| | - Nazaire Mangani Nseka
- Department of Internal Medicine, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
| | - Ernest Kiswaya Sumaili
- Department of Internal Medicine, Division of Nephrology–Dialysis, University of Kinshasa Hospital, Kinshasa, Democratic Republic of the Congo
| |
Collapse
|
20
|
Nuclear magnetic resonance reveals postprandial low-density lipoprotein cholesterol determined by enzymatic method could be a misleading indicator. Clin Chim Acta 2020; 514:59-65. [PMID: 33333042 DOI: 10.1016/j.cca.2020.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/05/2020] [Accepted: 12/08/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Serum concentration of low-density lipoprotein cholesterol (LDL-C) is markedly reduced after a meal. Does postprandial cholesterol in LDL truly decline via clearance of LDL particles or is there simply a redistribution of cholesterol in LDL subclasses? Thus, we sought to evaluate whether postprandial decline of LDL-C reflects a reduction of LDL particle and to assess the correlation between proprotein convertase subtilisin/kexin type 9 (PCSK9) concentration and postprandial atherogenic lipoproteins profile. METHODS Eighty-seven persons were enrolled in this study. We measured lipid profiles by enzymatic and nuclear magnetic resonance (NMR)-based methods and serum PCSK9 concentration by enzyme-linked immunosorbent assays before and after a meal. Plasma samples were collected after a 10-h fasting and 2 and 4 h post-meal. RESULTS Compared to the fasting status, there was significant postprandial decline of LDL-C measured enzymatically (LDL-Ce) at 2nd and 4th h [99.38 (80.43, 120.65) vs 95.51 (74.25, 117.17) vs 87.01 (69.99, 108.28) mg/dl, p < 0.000]. But there was no significant reduction in LDL particle and its cholesterol content (LDL-Cn) determined by NMR. Just the postprandial large LDL particle [186.45 (151.36, 229.42) vs 176.92 (147.43, 220.91) vs 181.77 (149.05, 224.17), p < 0.000] and its cholesterol content [19.10 (15.09, 22.37) vs 18.28 (14.59, 21.84) vs 17.79 (14.62, 22.14), p < 0.000] were greatly decreased at 2nd and 4th h compared to the fasting one. Interestingly, postprandial serum PCSK9 was decreased at 2nd and 4th h compared with fasting concentration [298.75 (233.25, 396.92) vs 257.34 (207.52, 342.36) vs 250.57 (215.02, 339.66) ng/ml, p < 0.000]. The postprandial percent decrease in serum PCSK9 at 4th h was positively correlated to the percent decline in postprandial LDL-Ce (r = 0.252, p = 0.019) but was independently associated with the percent increase in remnant cholesterol (r = 0.262, p = 0.016). CONCLUSIONS Postprandial decline of LDL-C determined enzymatically was not confirmed by NMR-based methods. Indeed, there exists cholesterol redistribution in LDL subclasses following a meal. The decrease of postprandial PCSK9 may be secondary to the increase in intrahepatic lipids following food intake.
Collapse
|
21
|
Qin Y, Ting F, Kim MJ, Strelnikov J, Harmon J, Gao F, Dose A, Teng BB, Alipour MA, Yao Z, Crooke R, Krauss RM, Medina MW. Phosphatidylinositol-(4,5)-Bisphosphate Regulates Plasma Cholesterol Through LDL (Low-Density Lipoprotein) Receptor Lysosomal Degradation. Arterioscler Thromb Vasc Biol 2020; 40:1311-1324. [PMID: 32188273 DOI: 10.1161/atvbaha.120.314033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE TMEM55B (transmembrane protein 55B) is a phosphatidylinositol-(4,5)-bisphosphate (PI[4,5]P2) phosphatase that regulates cellular cholesterol, modulates LDLR (low-density lipoprotein receptor) decay, and lysosome function. We tested the effects of Tmem55b knockdown on plasma lipids in mice and assessed the roles of LDLR lysosomal degradation and change in (PI[4,5]P2) in mediating these effects. Approach and Results: Western diet-fed C57BL/6J mice were treated with antisense oligonucleotides against Tmem55b or a nontargeting control for 3 to 4 weeks. Hepatic Tmem55b transcript and protein levels were reduced by ≈70%, and plasma non-HDL (high-density lipoprotein) cholesterol was increased ≈1.8-fold (P<0.0001). Immunoblot analysis of fast protein liquid chromatography (FPLC) fractions revealed enrichment of ApoE-containing particles in the LDL size range. In contrast, Tmem55b knockdown had no effect on plasma cholesterol in Ldlr-/- mice. In primary hepatocytes and liver tissues from Tmem55b knockdown mice, there was decreased LDLR protein. In the hepatocytes, there was increased lysosome staining and increased LDLR-lysosome colocalization. Impairment of lysosome function (incubation with NH4Cl or knockdown of the lysosomal proteins LAMP1 or RAB7) abolished the effect of TMEM55B knockdown on LDLR in HepG2 (human hepatoma) cells. Colocalization of the recycling endosome marker RAB11 (Ras-related protein 11) with LDLR in HepG2 cells was reduced by 50% upon TMEM55B knockdown. Finally, knockdown increased hepatic PI(4,5)P2 levels in vivo and in HepG2 cells, while TMEM55B overexpression in vitro decreased PI(4,5)P2. TMEM55B knockdown decreased, whereas overexpression increased, LDL uptake in HepG2 cells. Notably, the TMEM55B overexpression effect was reversed by incubation with PI(4,5)P2. Conclusions: These findings indicate a role for TMEM55B in regulating plasma cholesterol levels by affecting PI(4,5)P2-mediated LDLR lysosomal degradation.
Collapse
Affiliation(s)
- Yuanyuan Qin
- From the Department of Pediatrics, University of California San Francisco, Oakland (Y.Q., F.T., R.M.K., M.W.M.)
| | - Flora Ting
- From the Department of Pediatrics, University of California San Francisco, Oakland (Y.Q., F.T., R.M.K., M.W.M.)
| | - Mee J Kim
- Children's Hospital Oakland Research Institute, CA (M.J.K., J.S., J.H., F.G., A.D.)
| | - Jacob Strelnikov
- Children's Hospital Oakland Research Institute, CA (M.J.K., J.S., J.H., F.G., A.D.)
| | - Joseph Harmon
- Children's Hospital Oakland Research Institute, CA (M.J.K., J.S., J.H., F.G., A.D.)
| | - Feng Gao
- Children's Hospital Oakland Research Institute, CA (M.J.K., J.S., J.H., F.G., A.D.)
| | - Andrea Dose
- Children's Hospital Oakland Research Institute, CA (M.J.K., J.S., J.H., F.G., A.D.)
| | - Ba-Bie Teng
- Center for Human Genetics, University of Texas Health Science Center, Houston (B.-B.T.)
| | - Mohsen Amir Alipour
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ontario, Canada (M.A.A., Z.Y.)
| | - Zemin Yao
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ontario, Canada (M.A.A., Z.Y.)
| | | | - Ronald M Krauss
- From the Department of Pediatrics, University of California San Francisco, Oakland (Y.Q., F.T., R.M.K., M.W.M.)
| | - Marisa W Medina
- From the Department of Pediatrics, University of California San Francisco, Oakland (Y.Q., F.T., R.M.K., M.W.M.)
| |
Collapse
|
22
|
Ponticelli C, Arnaboldi L, Moroni G, Corsini A. Treatment of dyslipidemia in kidney transplantation. Expert Opin Drug Saf 2020; 19:257-267. [DOI: 10.1080/14740338.2020.1732921] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Claudio Ponticelli
- Divisione di Nefrologia, Istituto Scientifico Ospedale Maggiore, Milano, Italy (retired)
| | - Lorenzo Arnaboldi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DISFeB), Università degli Studi di Milano, Milano, Italy
| | - Gabriella Moroni
- Nefrologia e Dialisi, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Alberto Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DISFeB), Università degli Studi di Milano, Milano, Italy
- IRCCS Multimedica, Milano, Italy
| |
Collapse
|
23
|
Sarkar SK, Foo ACY, Matyas A, Asikhia I, Kosenko T, Goto NK, Vergara-Jaque A, Lagace TA. A transient amphipathic helix in the prodomain of PCSK9 facilitates binding to low-density lipoprotein particles. J Biol Chem 2020; 295:2285-2298. [PMID: 31949048 PMCID: PMC7039556 DOI: 10.1074/jbc.ra119.010221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 01/13/2020] [Indexed: 01/07/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type-9 (PCSK9) is a ligand of low-density lipoprotein (LDL) receptor (LDLR) that promotes LDLR degradation in late endosomes/lysosomes. In human plasma, 30–40% of PCSK9 is bound to LDL particles; however, the physiological significance of this interaction remains unknown. LDL binding in vitro requires a disordered N-terminal region in PCSK9's prodomain. Here, we report that peptides corresponding to a predicted amphipathic α-helix in the prodomain N terminus adopt helical structure in a membrane-mimetic environment. This effect was greatly enhanced by an R46L substitution representing an atheroprotective PCSK9 loss-of-function mutation. A helix-disrupting proline substitution within the putative α-helical motif in full-length PCSK9 lowered LDL binding affinity >5-fold. Modeling studies suggested that the transient α-helix aligns multiple polar residues to interact with positively charged residues in the C-terminal domain. Gain-of-function PCSK9 mutations associated with familial hypercholesterolemia (FH) and clustered at the predicted interdomain interface (R469W, R496W, and F515L) inhibited LDL binding, which was completely abolished in the case of the R496W variant. These findings shed light on allosteric conformational changes in PCSK9 required for high-affinity binding to LDL particles. Moreover, the initial identification of FH-associated mutations that diminish PCSK9's ability to bind LDL reported here supports the notion that PCSK9-LDL association in the circulation inhibits PCSK9 activity.
Collapse
Affiliation(s)
- Samantha K Sarkar
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Alexander C Y Foo
- Department of Chemistry and Biomolecular Sciences, Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Angela Matyas
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Ikhuosho Asikhia
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Tanja Kosenko
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Natalie K Goto
- Department of Chemistry and Biomolecular Sciences, Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Ariela Vergara-Jaque
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, Talca, Chile; Millennium Nucleus of Ion Channels-associated Diseases (MiNICAD), 3460000 Talca, Chile
| | - Thomas A Lagace
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada.
| |
Collapse
|
24
|
Iqbal Z, Dhage S, Mohamad JB, Abdel-Razik A, Donn R, Malik R, Ho JH, Liu Y, Adam S, Isa B, Stefanutti C, Soran H. Efficacy and safety of PCSK9 monoclonal antibodies. Expert Opin Drug Saf 2019; 18:1191-1201. [DOI: 10.1080/14740338.2019.1681395] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Zohaib Iqbal
- Cardiovascular Trials Unit, The Old St Mary’s Hospital, Central Manchester University Hospitals, Manchester, UK
| | - Shaishav Dhage
- Cardiovascular Trials Unit, The Old St Mary’s Hospital, Central Manchester University Hospitals, Manchester, UK
| | | | - Alaa Abdel-Razik
- Cardiovascular Trials Unit, The Old St Mary’s Hospital, Central Manchester University Hospitals, Manchester, UK
| | - Rachelle Donn
- Cardiovascular Research Group, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Rayaz Malik
- Department of Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Jan Hoong Ho
- Cardiovascular Trials Unit, The Old St Mary’s Hospital, Central Manchester University Hospitals, Manchester, UK
| | - Yifen Liu
- Cardiovascular Research Group, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Safwaan Adam
- Cardiovascular Trials Unit, The Old St Mary’s Hospital, Central Manchester University Hospitals, Manchester, UK
| | - Basil Isa
- Department of Endocrinology and Diabetes, Wythenshawe Hospital, Manchester, UK
| | - Claudia Stefanutti
- Department of Molecular Medicine, Sapienza’ University of Rome, Rome, Italy
| | - Handrean Soran
- Cardiovascular Trials Unit, The Old St Mary’s Hospital, Central Manchester University Hospitals, Manchester, UK
| |
Collapse
|
25
|
Hopkins PN, Krempf M, Bruckert E, Donahue S, Yang F, Zhang Y, DiCioccio AT. Pharmacokinetic and pharmacodynamic assessment of alirocumab in patients with familial hypercholesterolemia associated with proprotein convertase subtilisin/kexin type 9 gain-of-function or apolipoprotein B loss-of-function mutations. J Clin Lipidol 2019; 13:970-978. [PMID: 31767518 DOI: 10.1016/j.jacl.2019.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/10/2019] [Accepted: 10/13/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Familial hypercholesterolemia is characterized by high levels of low-density lipoprotein cholesterol (LDL-C), and causes of familial hypercholesterolemia include apolipoprotein B (APOB) loss-of-function mutations (LOFm) and proprotein convertase subtilisin/kexin type 9 (PCSK9) gain-of-function mutations (GOFm). OBJECTIVE The aim of this study was to compare the pharmacokinetics and pharmacodynamics of alirocumab between patients with APOB LOFm vs PCSK9 GOFm. METHODS Patients (6 APOB LOFm and 17 PCSK9 GOFm carriers) with LDL-C ≥70 mg/dL on maximally tolerated lipid-lowering therapies received alirocumab 150 mg at Weeks 0, 2, 4, and 6, placebo at Week 8, alirocumab at Week 10, placebo at Weeks 12 and 14, then completed a follow-up period at Week 22. RESULTS At Week 8, mean ± standard error (SE) alirocumab concentration was lower in APOB LOFm carriers compared with PCSK9 GOFm carriers (12.12 ± 1.81 vs 16.74 ± 2.53 mg/L). APOB LOFm carriers had higher mean ± SE total PCSK9 (6.56 ± 0.73 mg/L) and lower mean ± SE free PCSK9 (0.025 ± 0.016 mg/L) at Week 8 compared with PCSK9 GOFm carriers (4.21 ± 0.35 and 0.11 ± 0.035 mg/L for total and free PCSK9, respectively). Despite this observed greater PCSK9 suppression, mean ± SE percent LDL-C reduction was lower in APOB LOFm (55.3 ± 1.0%) compared with PCSK9 GOFm carriers (73.1 ± 0.9%). Treatment-emergent adverse events occurred in 16 patients (94.1%) in the PCSK9 GOFm group and 5 patients (83.3%) in the APOB LOFm group. CONCLUSIONS Overall, PCSK9 inhibition with alirocumab results in clinically meaningful reductions in LDL-C in both APOB LOFm and PCSK9 GOFm carriers, although reductions were greater in the PCSK9 GOFm carriers. The results indicate a possible underlying contributor to hypercholesterolemia other than PCSK9 in patients with APOB LOFm. CLINICAL TRIAL REGISTRATION NCT01604824; clinicaltrials.gov.
Collapse
Affiliation(s)
- Paul N Hopkins
- School of Medicine, University of Utah, Salt Lake City, UT, USA.
| | - Michel Krempf
- CHU de Nantes - Hôpital Nord Laennec, Saint-Herblain, France
| | | | | | - Feng Yang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Yi Zhang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | |
Collapse
|
26
|
Wang M, Zhao D, Xu L, Guo W, Nie L, Lei Y, Long Y, Liu M, Wang Y, Zhang X, Zhang L, Li H, Zhang J, Yuan D, Yue L. Role of PCSK9 in lipid metabolic disorders and ovarian dysfunction in polycystic ovary syndrome. Metabolism 2019; 94:47-58. [PMID: 30768966 DOI: 10.1016/j.metabol.2019.02.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/03/2019] [Accepted: 02/09/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a critical role in the cholesterol metabolism by negatively regulating the low-density lipoprotein receptor (LDLR). Lipid metabolic and ovarian disorders are the common clinical manifestation of polycystic ovary syndrome (PCOS). Here, we intended to elucidate the role of PCSK9 in the pathogenesis of PCOS conducted on a human population in case-control design and animal part in an interventional study. METHODS We firstly investigated the serum levels of PCSK9 in 46 PCOS patients compared with 49 healthy women as controls, and then developed a PCOS mouse model induced by dehydroepiandrosterone (DHEA) and a high-fat diet (HFD) to determine the role of PCSK9 in abnormal lipid metabolism and ovarian dysfunction of PCOS in four groups (n = 40 per group): control, PCOS mice, PCOS plus alirocumab group, and PCOS plus vehicle group. The expression of PCSK9 in their serum, hepatic and ovarian tissues, serum lipid profiles and hormones were measured. Additionally, mRNA and protein expression levels of LDLR in hepatic and ovarian tissues, ovarian morphology and function were determined. Finally, we used freshly isolated theca-interstitial cells (TICs) and granulosa cells (GCs) from prepubertal normal mice to explore the effect of PCSK9 on LDL uptake of the cells. RESULTS Serum PCSK9 concentrations were higher in PCOS patients than normal controls (P < 0.05). The PCOS model mice exhibited significantly increased serum levels of total cholesterol (TC), LDL-C and high-density lipoprotein-cholesterol (HDL-C; P < 0.001, P < 0.001, P = 0.0004, respectively). Moreover, the serum PCSK9 protein level was significantly increased in PCOS mice (P = 0.0002), which positively correlated with serum LDL-C (r = 0.5279, P = 0.0004) and TC (r = 0.4151, P = 0.035). In both liver and ovary of PCOS mice, PCSK9 mRNA and protein levels were significantly increased (P < 0.05), but LDLR levels were significantly decreased (P < 0.05). Furthermore, alirocumab inhibiting PCSK9 partly increased in LDLR expression in both liver and ovary in PCOS mice, also ameliorated the lipid metabolic disorders and pathological changes of ovarian morphology and function and serum reproductive hormones but not in the PCOS plus vehicle group. In vitro experiment, recombinant PCSK9 decreased LDL uptake in TICs and GCs (P < 0.001, P = 0.0011, respectively), which were partly reversed by alirocumab (P < 0.001, P = 0.012, respectively). CONCLUSION Abnormal high expression of PCSK9 in the blood, liver and ovary may be involved in the pathogenesis of PCOS by affecting lipid metabolism and ovarian function, and the inhibition of PCSK9 may partly reverse the pathological changes of PCOS. Our research suggests a possibility of PCSK9 as a new attractive target for diagnosis and treatment of PCOS.
Collapse
Affiliation(s)
- Meijiao Wang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Dan Zhao
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Liangzhi Xu
- Reproductive Endocrinology and Regulation Joint Laboratory, West China Second University Hospital, Sichuan University, Sichuan, Chengdu, China; Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan, Chengdu, China
| | - Wenjing Guo
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Li Nie
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Yi Lei
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Yun Long
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Min Liu
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Yichen Wang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Xueqin Zhang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Li Zhang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Hanna Li
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Jinhu Zhang
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China
| | - Dongzhi Yuan
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China.
| | - Limin Yue
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, Chengdu, China.
| |
Collapse
|
27
|
Comparing the serum TAG response to high-dose supplementation of either DHA or EPA among individuals with increased cardiovascular risk: the ComparED study. Br J Nutr 2019; 121:1223-1234. [PMID: 30854986 DOI: 10.1017/s0007114519000552] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Studies have shown that the reduction in serum TAG concentrations with long-chain n-3 fatty acid supplementation is highly variable among individuals. The objectives of the present study were to compare the proportions of individuals whose TAG concentrations lowered after high-dose DHA and EPA, and to identify the predictors of response to both modalities. In a double-blind, controlled, crossover study, 154 men and women were randomised to three supplemented phases of 10 weeks each: (1) 2·7 g/d of DHA, (2) 2·7 g/d of EPA and (3) 3 g/d of maize oil, separated by 9-week washouts. As secondary analyses, the mean intra-individual variation in TAG was calculated using the standard deviation from the mean of four off-treatment samples. The response remained within the intra-individual variation (±0·25 mmol/l) in 47 and 57 % of participants after DHA and EPA, respectively. Although there was a greater proportion of participants with a reduction >0·25 mmol/l after DHA than after EPA (45 υ. 32 %; P 0·25 mmol/l after both DHA and EPA had higher non-HDL-cholesterol, TAG and insulin concentrations compared with other responders at baseline (all P < 0·05). In conclusion, supplementation with 2·7 g/d DHA or EPA had no meaningful effect on TAG concentrations in a large proportion of individuals with normal mean TAG concentrations at baseline. Although DHA lowered TAG in a greater proportion of individuals compared with EPA, the magnitude of TAG lowering among them was similar.
Collapse
|
28
|
Xiao J, Bai XQ, Liao L, Zhou M, Peng J, Xiang Q, Ren Z, Wen HY, Jiang ZS, Tang ZH, Wang MM, Liu LS. Hydrogen sulfide inhibits PCSK9 expression through the PI3K/Akt‑SREBP‑2 signaling pathway to influence lipid metabolism in HepG2 cells. Int J Mol Med 2019; 43:2055-2063. [PMID: 30864739 PMCID: PMC6443339 DOI: 10.3892/ijmm.2019.4118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/25/2019] [Indexed: 12/20/2022] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous gaseous signaling molecule that plays important roles in the cardiovascular system. In our previous studies, we demonstrated that H2S regulates lipid metabolism. In the present study, we aimed to explore the mechanisms through which H2 regulates lipid metabolism in HepG2 cells in vitro. Treatment of the HepG2 cells with H2S inhibited the expression of proprotein convertase subtilisin/kexin type 9 (PCSK9) and increased the level of low-density lipoprotein receptor (LDLR) in a time- and dose-dependent manner. The knockdown of PCSK9 by siRNA effectively increased the levels of LDLR and 1,1′-dioctadecyl-3,3,3′,3′-tetramethyl-indocarbocyanine perchlorate-labeled LDL (DiI-LDL) uptake in the H2S-treated HepG2 cells. Furthermore, the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)-sterol regulatory element binding proteins 2 (SREBP-2) signaling pathway was confirmed to be involved in H2S-regulated PCSK9 expression. Notably, the HepG2 cells were incubated with 30% serum and DiI-LDL for 24 h, and the results revealed that H2S increased lipid uptake, but caused no increase in lipid accumulation. On the whole, the findings of this study demonstrate that H2S is involved in the regulation of lipid metabolism in HepG2 cells through the regulation of the expression of PCSK9 via the PI3K/Akt-SREBP-2 signaling pathway. To the very best of our knowledge, this study is the first to report that H2S can regulate the expression of PCSK9.
Collapse
Affiliation(s)
- Jun Xiao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xue-Qin Bai
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ling Liao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Min Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Juan Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Qiong Xiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong-Yan Wen
- Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Han Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Mei-Mei Wang
- Department of Pediatrics, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lu-Shan Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
29
|
Bojanin D, Vekic J, Milenkovic T, Vukovic R, Zeljkovic A, Stefanovic A, Janac J, Ivanisevic J, Mitrovic K, Miljkovic M, Spasojevic-Kalimanovska V. Association between proprotein convertase subtilisin/kexin 9 (PCSK9) and lipoprotein subclasses in children with type 1 diabetes mellitus: Effects of glycemic control. Atherosclerosis 2019; 280:14-20. [DOI: 10.1016/j.atherosclerosis.2018.11.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/01/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022]
|
30
|
Jerome RN, Pulley JM, Roden DM, Shirey-Rice JK, Bastarache LA, R Bernard G, B Ekstrom L, Lancaster WJ, Denny JC. Using Human 'Experiments of Nature' to Predict Drug Safety Issues: An Example with PCSK9 Inhibitors. Drug Saf 2018; 41:303-311. [PMID: 29185237 DOI: 10.1007/s40264-017-0616-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION When a new drug enters the market, its full array of side effects remains to be defined. Current surveillance approaches targeting these effects remain largely reactive. There is a need for development of methods to predict specific safety events that should be sought for a given new drug during development and postmarketing activities. OBJECTIVE We present here a safety signal identification approach applied to a new set of drug entities, inhibitors of the serine protease proprotein convertase subtilisin/kexin type 9 (PCSK9). METHODS Using phenome-wide association study (PheWAS) methods, we analyzed available genotype and clinical data from 29,722 patients, leveraging the known effects of changes in PCSK9 to identify novel phenotypes in which this protein and its inhibitors may have impact. RESULTS PheWAS revealed a significantly reduced risk of hypercholesterolemia (odds ratio [OR] 0.68, p = 7.6 × 10-4) in association with a known loss-of-function variant in PCSK9, R46L. Similarly, laboratory data indicated significantly reduced beta mean low-density lipoprotein cholesterol (- 14.47 mg/dL, p = 2.58 × 10-23) in individuals carrying the R46L variant. The R46L variant was also associated with an increased risk of spina bifida (OR 5.90, p = 2.7 × 10-4), suggesting that further investigation of potential connections between inhibition of PCSK9 and neural tube defects may be warranted. CONCLUSION This novel methodology provides an opportunity to put in place new mechanisms to assess the safety and long-term tolerability of PCSK9 inhibitors specifically, and other new agents in general, as they move into human testing and expanded clinical use.
Collapse
Affiliation(s)
- Rebecca N Jerome
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Jill M Pulley
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dan M Roden
- Office of Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jana K Shirey-Rice
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa A Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Gordon R Bernard
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Office of Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leeland B Ekstrom
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Nashville Biosciences, Nashville, TN, USA
| | - William J Lancaster
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
31
|
Ruscica M, Simonelli S, Botta M, Ossoli A, Lupo MG, Magni P, Corsini A, Arca M, Pisciotta L, Veglia F, Franceschini G, Ferri N, Calabresi L. Plasma PCSK9 levels and lipoprotein distribution are preserved in carriers of genetic HDL disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:991-997. [DOI: 10.1016/j.bbalip.2018.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 04/18/2018] [Accepted: 05/27/2018] [Indexed: 01/27/2023]
|
32
|
Allaire J, Vors C, Tremblay AJ, Marin J, Charest A, Tchernof A, Couture P, Lamarche B. High-Dose DHA Has More Profound Effects on LDL-Related Features Than High-Dose EPA: The ComparED Study. J Clin Endocrinol Metab 2018; 103:2909-2917. [PMID: 29846653 DOI: 10.1210/jc.2017-02745] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 05/22/2018] [Indexed: 11/19/2022]
Abstract
CONTEXT Supplementation with high-dose docosahexaenoic acid (DHA) increases serum low-density lipoprotein (LDL) cholesterol (LDL-C) concentrations more than high-dose eicosapentaenoic acid (EPA). The mechanisms underlying this difference are unknown. OBJECTIVE To examine the phenotypic change in LDL and mechanisms responsible for the differential LDL-C response to EPA and DHA supplementation in men and women at risk of cardiovascular disease. DESIGN, SETTING, PARTICIPANTS, AND INTERVENTION In a double-blind, controlled, crossover study, 48 men and 106 women with abdominal obesity and subclinical inflammation were randomized to a sequence of three treatment phases: phase 1, 2.7 g/d of EPA; phase 2, 2.7 g/d of DHA; and phase 3, 3 g/d of corn oil. All supplements were provided as three 1-g capsules for a total of 3 g/d. The 10-week treatment phases were separated by a 9-week washout period. MAIN OUTCOME MEASURE In vivo kinetics of apolipoprotein (apo)B100-containing lipoproteins were assessed using primed-constant infusion of deuterated leucine at the end of each treatment in a subset of participants (n = 19). RESULTS Compared with EPA, DHA increased LDL-C concentrations (+3.3%; P = 0.038) and mean LDL particle size (+0.7 Å; P < 0.001) and reduced the proportion of small LDL (-3.2%; P < 0.01). Both EPA and DHA decreased proprotein convertase subtilisin/kexin type 9 concentrations similarly (-18.2% vs -25.0%; P < 0.0001 vs control). Compared with EPA, DHA supplementation increased both the LDL apoB100 fractional catabolic rate (+11.4%; P = 0.008) and the production rate (+9.4%; P = 0.03). CONCLUSIONS The results of the present study have shown that supplementation with high-dose DHA increases LDL turnover and contributes to larger LDL particles compared with EPA.
Collapse
Affiliation(s)
- Janie Allaire
- Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Cécile Vors
- Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec City, Québec, Canada
| | - André J Tremblay
- Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Johanne Marin
- Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec City, Québec, Canada
| | - Amélie Charest
- Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec City, Québec, Canada
| | - André Tchernof
- Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec City, Québec, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
- Institut Universitaire de Cardiologie et de Pneumologie du Québec, Québec City, Québec, Canada
| | - Patrick Couture
- Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec City, Québec, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
| | - Benoît Lamarche
- Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
33
|
Kosmas CE, DeJesus E, Morcelo R, Garcia F, Montan PD, Guzman E. Lipid-lowering interventions targeting proprotein convertase subtilisin/kexin type 9 (PCSK9): an emerging chapter in lipid-lowering therapy. Drugs Context 2017; 6:212511. [PMID: 29209403 PMCID: PMC5706592 DOI: 10.7573/dic.212511] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/12/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease that is mainly expressed in the liver but can also be found in the intestine and kidneys. PCSK9 promotes the degradation of low density lipoprotein receptors (LDLR) by reducing their recycling and targeting the receptors for lysosomal destruction, thereby decreasing the rate of removal of LDL-cholesterol from the circulation. Thus, interventions targeting PCSK9 by reducing its expression may lead to significant reductions of LDL-cholesterol and possibly decrease cardiovascular risk. The present review aims to present and discuss the current clinical and scientific data pertaining to lipid-lowering interventions targeting PCSK9.
Collapse
Affiliation(s)
- Constantine E Kosmas
- Department of Medicine, Division of Cardiology, Mount Sinai Hospital, New York, NY, USA
| | - Eddy DeJesus
- Department of Medicine, Bronx-Lebanon Hospital Center, Bronx, NY, USA
| | - Rosmery Morcelo
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | - Frank Garcia
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | - Peter D Montan
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | - Eliscer Guzman
- Department of Medicine, Division of Cardiology, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
34
|
The PCSK9-LDL Receptor Axis and Outcomes in Heart Failure. J Am Coll Cardiol 2017; 70:2128-2136. [DOI: 10.1016/j.jacc.2017.08.057] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 12/17/2022]
|
35
|
Pirro M, Bianconi V, Francisci D, Schiaroli E, Bagaglia F, Sahebkar A, Baldelli F. Hepatitis C virus and proprotein convertase subtilisin/kexin type 9: a detrimental interaction to increase viral infectivity and disrupt lipid metabolism. J Cell Mol Med 2017; 21:3150-3161. [PMID: 28722331 PMCID: PMC5706572 DOI: 10.1111/jcmm.13273] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/07/2017] [Indexed: 12/21/2022] Open
Abstract
From viral binding to the hepatocyte surface to extracellular virion release, the replication cycle of the hepatitis C virus (HCV) intersects at various levels with lipid metabolism; this leads to a derangement of the lipid profile and to increased viral infectivity. Accumulating evidence supports the crucial regulatory role of proprotein convertase subtilisin/kexin type 9 (PCSK9) in lipoprotein metabolism. Notably, a complex interaction between HCV and PCSK9 has been documented. Indeed, either increased or reduced circulating PCSK9 levels have been observed in HCV patients; this discrepancy might be related to several confounders, including HCV genotype, human immunodeficiency virus (HIV) coinfection and the ambiguous HCV‐mediated influence on PCSK9 transcription factors. On the other hand, PCSK9 may itself influence HCV infectivity, inasmuch as the expression of different hepatocyte surface entry proteins and receptors is regulated by PCSK9. The aim of this review is to summarize the current evidence about the complex interaction between HCV and liver lipoprotein metabolism, with a specific focus on PCSK9. The underlying assumption of this review is that the interconnections between HCV and PCSK9 may be central to explain viral infectivity.
Collapse
Affiliation(s)
- Matteo Pirro
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Daniela Francisci
- Unit of Infectious Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Elisabetta Schiaroli
- Unit of Infectious Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Francesco Bagaglia
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Franco Baldelli
- Unit of Infectious Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
36
|
Breslin WJ, Hilbish KG, Cannady EA, Edwards TL. Fertility and Embryo-Fetal Development Assessment in Rats and Rabbits with Evacetrapib: A Cholesteryl Ester Transfer Protein Inhibitor. Birth Defects Res 2017; 109:513-527. [PMID: 28398618 DOI: 10.1002/bdr2.1022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND The purpose of these studies was to evaluate the effects of evacetrapib on male and female fertility and on embryo-fetal development (EFD). METHODS Evacetrapib, a potent and selective inhibitor of cholesteryl ester transfer protein (CETP), was administered daily by oral gavage starting 2 weeks (for female) or 4 weeks (for male) before mating, during cohabitation, and until necropsy in the male rat fertility study or through gestation day (GD) 17 in the female rat combined fertility/EFD study. For rabbit EFD studies, animals were dosed from GDs 7 to 19 or from 1 week before mating through GD 19. Dose levels of evacetrapib ranged from 60 to 600 mg/kg for rats and from 1 to 100 mg/kg/day for rabbits. RESULTS Parental findings in rats included decreased body weight and food consumption and moribund euthanasia in animals given 600 mg/kg/day and decreased food consumption at 300 mg/kg/day. There were no adverse effects on estrus cycling, fertility indices, sperm parameters, maternal reproductive parameters, male reproductive tissue, or fetal viability, growth, or external/visceral morphology. An increase in the incidence of 14th rudimentary ribs, a minor, transient variation considered nonadverse, was the only significant developmental finding in rats given 600 mg/kg/day. Slight decreases in body weight and food consumption at 100 mg/kg/day were the only maternal effects observed in rabbits with no adverse developmental effects noted. CONCLUSION No adverse effects on fertility or EFD were observed in rats at doses up to 600 mg/kg/day and no adverse effects on EFD were noted in rabbits at doses up to 100 mg/kg/day. Birth Defects Research 109:513-527, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- William J Breslin
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana
| | - Kim G Hilbish
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana
| | - Ellen A Cannady
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana
| | | |
Collapse
|
37
|
Abstract
INTRODUCTION Cardiovascular morbidity and mortality are of increasing concern, not only to patients but also to the health care profession and service providers. The preventative benefit of treatment of dyslipidaemia is unquestioned but there is a large, so far unmet need to improve clinical outcome. There are exciting new discoveries of targets that may translate into improved clinical outcome. Areas covered: This review highlights some new pathways in cholesterol and triglyceride metabolism and examines new targets, new drugs and new molecules. The review includes the results of recent trials of relatively new drugs that have shown benefit in cardiovascular endpoint outcomes, drugs that have been licenced without endpoint trials yet available and new drugs that have not yet been licenced but have produced exciting results in animal studies and some in early phase 2 human studies. Expert opinion: The new areas that have been discovered as the cause of dyslipidaemia have opened up a host of new targets for new drugs including antisense RNA's, microRNA's and human monoclonal antibodies. The plethora of new targets and new drugs has made it an extraordinarily exciting time in the development of therapeutics to combat atherosclerosis.
Collapse
Affiliation(s)
- Gerald H Tomkin
- a Diabetes Institute of Ireland , Beacon Clinic and Trinity College , Dublin 2 , Ireland
| | - Daphne Owens
- a Diabetes Institute of Ireland , Beacon Clinic and Trinity College , Dublin 2 , Ireland
| |
Collapse
|
38
|
Scott Kiss R, Sniderman A. Shunts, channels and lipoprotein endosomal traffic: a new model of cholesterol homeostasis in the hepatocyte. J Biomed Res 2017; 31:95-107. [PMID: 28808191 PMCID: PMC5445212 DOI: 10.7555/jbr.31.20160139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The liver directs cholesterol metabolism in the organism. All the major fluxes of cholesterol within the body involve the liver: dietary cholesterol is directed to the liver; cholesterol from peripheral cells goes to the liver; the liver is a major site of cholesterol synthesis for the organism; cholesterol is secreted from the liver within the bile, within apoB lipoproteins and translocated to nascent HDL. The conventional model of cholesterol homeostasis posits that cholesterol from any source enters a common, rapidly exchangeable pool within the cell, which is in equilibrium with a regulatory pool. Increased influx of cholesterol leads rapidly to decreased synthesis of cholesterol. This model was developed based on in vitro studies in the fibroblast and validated only for LDL particles. The challenges the liver must meet in vivo to achieve cholesterol homeostasis are far more complex. Our model posits that the cholesterol derived from three different lipoproteins endosomes has three different fates: LDL-derived cholesterol is largely recycled within VLDL with most of the cholesterol shunted through the hepatocyte without entering the exchangeable pool of cholesterol; high density lipoprotein-derived CE is transcytosed into bile; and chylomicron remnant-derived cholesterol primarily enters the regulatory pool within the hepatocyte. These endosomal channels represent distinct physiological pathways and hepatic homeostasis represents the net result of the outcomes of these distinct channels. Our model takes into account the distinct physiological challenges the hepatocyte must meet, underlie the pathophysiology of many of the apoB dyslipoproteinemias and account for the sustained effectiveness of therapeutic agents such as statins.
Collapse
Affiliation(s)
- Robert Scott Kiss
- Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada.,Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Allan Sniderman
- Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada.,Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
39
|
Unpacking and Understanding the Impact of Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors on Apolipoprotein B Metabolism. Circulation 2017; 135:363-365. [DOI: 10.1161/circulationaha.116.025897] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
The complexity of lipoprotein (a) lowering by PCSK9 monoclonal antibodies. Clin Sci (Lond) 2017; 131:261-268. [DOI: 10.1042/cs20160403] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/04/2023]
Abstract
Since 2012, clinical trials dedicated to proprotein convertase subtilisin kexin type 9 (PCSK9) inhibition with monoclonal antibodies (mAbs) have unambiguously demonstrated robust reductions not only in low-density lipoprotein (LDL) cholesterol (LDL-C) but also in lipoprotein (a) [Lp(a)] levels. The scientific literature published prior to those studies did not provide any evidence for a link between PCSK9 and Lp(a) metabolism. More recent investigations, either in vitro or in vivo, have attempted to unravel the mechanism(s) by which PCSK9 mAbs reduce circulating Lp(a) levels, with some showing a specific implication of the LDL receptor (LDLR) in Lp(a) clearance whereas others found no significant role for the LDLR in that process. This elusive pathway appears clearly distinct from that of the widely prescribed statins that also enhance LDLR function but do not lower circulating Lp (a) levels in humans. So how does PCSK9 inhibition with mAbs reduce Lp(a)? This still remains to be established.
Collapse
|
41
|
Abstract
PURPOSE Chronic kidney disease (CKD) is accompanied by a number of secondary metabolic dysregulations, such as lipid abnormalities, presenting with unique characteristics. Proprotein convertase subtilisin/kexin 9 (PCSK9) inhibitors have been introduced as the new era in the management of dyslipidemia with promising results in groups with refractory lipid abnormalities. Increasing number of studies investigate the possible association of PCSK9 levels with kidney function, especially with nephrotic range proteinuria, as well as its role as a prognostic cardiovascular risk marker in CKD. In this review, we discuss the existing evidence for PCSK9 levels in patient groups with nephrotic syndrome, non-dialysis CKD, end-stage renal disease and kidney transplantation. METHODS Online research was conducted in MEDLINE database to identify articles investigating PCSK9 in all different aspects of CKD. References from relevant studies were screened for supplementary articles. RESULTS Four cross-sectional studies, one secondary analysis, one publication from two independent cohort studies and one multicentre prospective cohort study assessed PCSK9 plasma levels in different subgroups of CKD patients. PCSK9 levels increase in nephrotic syndrome and have a positive correlation with proteinuria. In CKD patients, no correlation was found between PCSK9 levels and estimated GFR. Peritoneal dialysis patients have higher PCSK9 levels compared with hemodialysis and renal transplant patients as well as general population. CONCLUSION Accumulative evidence focuses on the possible association of PCSK9 levels with kidney function. No data are available for the administration of PCSK9 inhibitors in CKD patients. Further research will optimize knowledge on the role of PCSK9 levels and PCSK9 inhibitors in CKD.
Collapse
|
42
|
Inoue I, Kubota R, Yanagi S, Akita M, Nakano T, Katayama S, Shimada A, Noda M. The New Molecular Entity Evolocumab, One Kind of PCSK9 Inhibitor, Reduce Plasma Small Size LDL-Cholesterol Levels by Using a New Standardized Method of Measuring LDL Size. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/ojmip.2017.71001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
43
|
Simha V, Qin S, Shah P, Smith BH, Kremers WK, Kushwaha S, Wang L, Pereira NL. Sirolimus Therapy Is Associated with Elevation in Circulating PCSK9 Levels in Cardiac Transplant Patients. J Cardiovasc Transl Res 2016; 10:9-15. [PMID: 28028691 DOI: 10.1007/s12265-016-9719-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/08/2016] [Indexed: 11/24/2022]
Abstract
Sirolimus used in transplantation is often associated with hypercholesterolemia. We measured serum lipid and PCSK9 levels in 51 heart transplant recipients who had their immunosuppressive therapy switched from calcineurin inhibitors to sirolimus. The switch resulted in a 23% increase in LDL cholesterol, and 46% increase in triglycerides and PCSK9 levels increased from 316 ± 105 ng/mL to 343 ± 107 ng/mL (p = 0.04), however the change in PCSK9 levels did not correlate with an increase in lipid levels (p = 0.2). To investigate the mechanism for the variability in the change in PCSK9 levels, lymphoblastoid cell lines were incubated with both sirolimus and everolimus, resulting in a 2-3 fold increase in PCSK9 expression and protein levels in mTOR inhibitor sensitive but not in mTOR inhibitor resistant cell lines. This first in human study demonstrates that sirolimus therapy is associated with elevation in PCSK9 levels which is not associated with sirolimus-induced hypercholesterolemia.
Collapse
Affiliation(s)
- Vinaya Simha
- Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
| | - Sisi Qin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | - Pankaj Shah
- Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
| | - Byron H Smith
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Walter K Kremers
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Sudhir Kushwaha
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Naveen L Pereira
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, 200 First Street SW, Rochester, MN, 55905, USA. .,Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
44
|
Dietary high oleic canola oil supplemented with docosahexaenoic acid attenuates plasma proprotein convertase subtilisin kexin type 9 (PCSK9) levels in participants with cardiovascular disease risk: A randomized control trial. Vascul Pharmacol 2016; 87:60-65. [DOI: 10.1016/j.vph.2016.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/21/2016] [Accepted: 06/26/2016] [Indexed: 12/13/2022]
|
45
|
Villard EF, Thedrez A, Blankenstein J, Croyal M, Tran TTT, Poirier B, Le Bail JC, Illiano S, Nobécourt E, Krempf M, Blom DJ, Marais AD, Janiak P, Muslin AJ, Guillot E, Lambert G. PCSK9 Modulates the Secretion But Not the Cellular Uptake of Lipoprotein(a) Ex Vivo: An Effect Blunted by Alirocumab. ACTA ACUST UNITED AC 2016; 1:419-427. [PMID: 29308438 PMCID: PMC5753417 DOI: 10.1016/j.jacbts.2016.06.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Unlike LDL uptake, Lp(a) uptake is not altered by PCSK9 or PCSK9 inhibition in primary human hepatocytes and in primary dermal fibroblasts isolated from familial hypercholesterolemic and non–familial hypercholesterolemic patients. Lp(a) uptake is occurring in the absence of a functional LDL receptor and is not affected by LDL receptor blockade with monoclonal antibodies. Lp(a) cellular binding and whole particle uptake are not altered by PCSK9. The secretion of Lp(a) from primary human hepatocytes is enhanced by PCSK9, an effect that is blunted by PCSK9 inhibition with alirocumab.
To elucidate how the proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitor alirocumab modulates lipoprotein(a) [Lp(a)] plasma levels, the authors performed a series of Lp(a) uptake studies in primary human hepatocytes and dermal fibroblasts and measured Lp(a) secretion from human hepatocytes. They found that Lp(a) cellular uptake occurred in a low-density lipoprotein receptor–independent manner. Neither PCSK9 nor alirocumab altered Lp(a) internalization. By contrast, the secretion of apolipoprotein (a) from human hepatocytes was sharply increased by PCSK9, an effect that was reversed by alirocumab. They propose that PCSK9 does not significantly modulate Lp(a) catabolism, but rather enhances the secretion of Lp(a) from liver cells.
Collapse
Affiliation(s)
| | - Aurélie Thedrez
- Inra UMR 1280, Nantes, France.,Université de Nantes UMR1280, Faculté de Médecine, Nantes, France
| | | | | | | | - Bruno Poirier
- Sanofi Recherche Développement, Chilly-Mazarin, France
| | | | | | - Estelle Nobécourt
- Inra UMR 1280, Nantes, France.,Université de Nantes UMR1280, Faculté de Médecine, Nantes, France
| | - Michel Krempf
- Inra UMR 1280, Nantes, France.,Université de Nantes UMR1280, Faculté de Médecine, Nantes, France
| | - Dirk J Blom
- Division of Lipidology, Department of Internal Medicine, University of Cape Town, Cape Town, South Africa
| | - A David Marais
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Philip Janiak
- Sanofi Recherche Développement, Chilly-Mazarin, France
| | | | | | - Gilles Lambert
- Inra UMR 1280, Nantes, France.,Inserm UMR 1188, Sainte-Clotilde, France.,Université de la Réunion UMR1188, Faculté de Médecine, Saint-Denis, France.,CHU de la Réunion, Saint Denis, France
| |
Collapse
|
46
|
Cui CJ, Li S, Zhu CG, Sun J, Du Y, Zhang Y, Wu NQ, Guo YL, Xu RX, Gao Y, Li JJ. Enhanced pro-protein convertase subtilisin/kexin type 9 expression by C-reactive protein through p38MAPK-HNF1α pathway in HepG2 cells. J Cell Mol Med 2016; 20:2374-2383. [PMID: 27633999 PMCID: PMC5134380 DOI: 10.1111/jcmm.12931] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/17/2016] [Indexed: 12/25/2022] Open
Abstract
Plasma C‐reactive protein (CRP) concentration is associated positively with cardiovascular risk, including dyslipidemia. We suggested a regulating role of CRP on pro‐protein convertase subtilisin/kexin type 9 (PCSK9), a key regulator of low‐density lipoprotein (LDL) metabolism, and demonstrated the PCSK9 as a pathway linking CRP and LDL regulation. Firstly, experiments were carried out in the presence of human CRP on the protein and mRNA expression of PCSK9 and LDL receptor (LDLR) in human hepatoma cell line HepG2 cells. Treatment with CRP (10 μg/ml) enhanced significantly the mRNA and protein expression of PCSK9 and suppressed the expression of LDLR. Of note, a late return of LDLR mRNA levels occurred at 12 hrs, while the LDLR protein continued to decrease at 24 hrs, suggesting that the late decrease in LDLR protein levels was unlikely to be accounted for the decrease in LDL mRNA. Secondly, the role of PCSK9 in CRP‐induced LDLR decrease and the underlying pathways were investigated. As a result, the inhibition of PCSK9 expression by small interfering RNA (siRNA) returned partly the level of LDLR protein and LDL uptake during CRP treatment; CRP‐induced PCSK9 increase was inhibited by the p38MAPK inhibitor, SB203580, resulting in a significant rescue of LDLR protein expression and LDL uptake; the pathway was involved in hepatocyte nuclear factor 1α (HNF1α) but not sterol responsive element‐binding proteins (SREBPs) preceded by the phosphorylation of p38MAPK. These findings indicated that CRP increased PCSK9 expression by activating p38MAPK‐HNF1α pathway, with a certain downstream impairment in LDL metabolism in HepG2 cells.
Collapse
Affiliation(s)
- Chuan-Jue Cui
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Sha Li
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Cheng-Gang Zhu
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Sun
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ying Du
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yan Zhang
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Na-Qiong Wu
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuan-Lin Guo
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Rui-Xia Xu
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ying Gao
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jian-Jun Li
- Division of Dyslipidemia, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Hosseni B, Shabani M, Mohammadi A, Naseri F, Soltanmohammadi E, Piran S, Najafi M. Plasma PCSK9 level affects passively LAMP-2 expression; an evidence of transcription network. GENE REPORTS 2016. [DOI: 10.1016/j.genrep.2016.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
48
|
Lappegård KT, Enebakk T, Thunhaug H, Hovland A. Transition from LDL apheresis to evolocumab in heterozygous FH is equally effective in lowering LDL, without lowering HDL cholesterol. Atherosclerosis 2016; 251:119-123. [DOI: 10.1016/j.atherosclerosis.2016.06.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/01/2016] [Accepted: 06/08/2016] [Indexed: 11/26/2022]
|
49
|
Zhang L, Wang HH. The essential functions of endoplasmic reticulum chaperones in hepatic lipid metabolism. Dig Liver Dis 2016; 48:709-16. [PMID: 27133206 DOI: 10.1016/j.dld.2016.03.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 03/06/2016] [Accepted: 03/22/2016] [Indexed: 12/11/2022]
Abstract
The endoplasmic reticulum (ER) is an essential organelle for protein and lipid synthesis in hepatocytes. ER homeostasis is vital to maintain normal hepatocyte physiology. Perturbed ER functions causes ER stress associated with accumulation of unfolded protein in the ER that activates a series of adaptive signalling pathways, termed unfolded protein response (UPR). The UPR regulates ER chaperone levels to preserve ER protein-folding environment to protect the cell from ER stress. Recent findings reveal an array of ER chaperones that alter the protein-folding environment in the ER of hepatocytes and contribute to dysregulation of hepatocyte lipid metabolism and liver disease. In this review, we will discuss the specific functions of these chaperones in regulation of lipid metabolism, especially de novo lipogenesis and lipid transport and demonstrate their homeostatic role not only for ER-protein synthesis but also for lipid metabolism in hepatocyte.
Collapse
Affiliation(s)
- LiChun Zhang
- Department of Emergency, Shengjing Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Hong-Hui Wang
- College of Biology, Hunan University, Changsha, Hunan Province, China.
| |
Collapse
|
50
|
Kockx M, Glaros E, Leung B, Ng TW, Berbée JFP, Deswaerte V, Nawara D, Quinn C, Rye KA, Jessup W, Rensen PCN, Meikle PJ, Kritharides L. Low-Density Lipoprotein Receptor-Dependent and Low-Density Lipoprotein Receptor-Independent Mechanisms of Cyclosporin A-Induced Dyslipidemia. Arterioscler Thromb Vasc Biol 2016; 36:1338-49. [PMID: 27150391 DOI: 10.1161/atvbaha.115.307030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 04/20/2016] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Cyclosporin A (CsA) is an immunosuppressant commonly used to prevent organ rejection but is associated with hyperlipidemia and an increased risk of cardiovascular disease. Although studies suggest that CsA-induced hyperlipidemia is mediated by inhibition of low-density lipoprotein receptor (LDLr)-mediated lipoprotein clearance, the data supporting this are inconclusive. We therefore sought to investigate the role of the LDLr in CsA-induced hyperlipidemia by using Ldlr-knockout mice (Ldlr(-/-)). APPROACH AND RESULTS Ldlr(-/-) and wild-type (wt) C57Bl/6 mice were treated with 20 mg/kg per d CsA for 4 weeks. On a chow diet, CsA caused marked dyslipidemia in Ldlr(-/-) but not in wt mice. Hyperlipidemia was characterized by a prominent increase in plasma very low-density lipoprotein and intermediate-density lipoprotein/LDL with unchanged plasma high-density lipoprotein levels, thus mimicking the dyslipidemic profile observed in humans. Analysis of specific lipid species by liquid chromatography-tandem mass spectrometry suggested a predominant effect of CsA on increased very low-density lipoprotein-IDL/LDL lipoprotein number rather than composition. Mechanistic studies indicated that CsA did not alter hepatic lipoprotein production but did inhibit plasma clearance and hepatic uptake of [(14)C]cholesteryl oleate and glycerol tri[(3)H]oleate-double-labeled very low-density lipoprotein-like particles. Further studies showed that CsA inhibited plasma lipoprotein lipase activity and increased levels of apolipoprotein C-III and proprotein convertase subtilisin/kexin type 9. CONCLUSIONS We demonstrate that CsA does not cause hyperlipidemia via direct effects on the LDLr. Rather, LDLr deficiency plays an important permissive role for CsA-induced hyperlipidemia, which is associated with abnormal lipoprotein clearance, decreased lipoprotein lipase activity, and increased levels of apolipoprotein C-III and proprotein convertase subtilisin/kexin type 9. Enhancing LDLr and lipoprotein lipase activity and decreasing apolipoprotein C-III and proprotein convertase subtilisin/kexin type 9 levels may therefore provide attractive treatment targets for patients with hyperlipidemia receiving CsA.
Collapse
Affiliation(s)
- Maaike Kockx
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Elias Glaros
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Betty Leung
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Theodore W Ng
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Jimmy F P Berbée
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Virginie Deswaerte
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Diana Nawara
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Carmel Quinn
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Kerry-Anne Rye
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Wendy Jessup
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Patrick C N Rensen
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Peter J Meikle
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.)
| | - Leonard Kritharides
- From the ANZAC Research Institute (M.K., D.N., W.J., L.K.) and Department of Cardiology (L.K.), Concord Hospital, University of Sydney, Sydney, Australia; Centre for Vascular Research (E.G., C.Q.) and Department of Pathology (B.L.), University of New South Wales, Sydney, Australia; Baker IDI Heart and Diabetes Institute, Melbourne, Australia (T.W.N., P.J.M.); Department of Medicine, Division Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands (J.F.P.B., P.C.N.R.); Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia (V.D.); Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Sydney, Australia (K.-A.R.).
| |
Collapse
|