1
|
Zahid S, Schlamp F, Gildea MA, Lin BX, Chaloemtoem A, Falis M, Parikh M, Fisher EA, Hornemann T, Vaisar T, Heffron SP. High-Density Lipoprotein Lipid and Protein Cargo and Cholesterol Efflux Capacity Before and After Bariatric Surgery. Arterioscler Thromb Vasc Biol 2025; 45:e48-e62. [PMID: 39744840 DOI: 10.1161/atvbaha.124.321686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/02/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Cholesterol efflux capacity (CEC) of HDL (high-density lipoprotein) is inversely associated with incident cardiovascular events, independent of HDL cholesterol. Obesity is characterized by low HDL cholesterol and impaired HDL function, such as CEC. Bariatric surgery, including Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG), broadly leads to improved cardiovascular outcomes, but impacts on risk factors differ by procedure, with greater improvements in weight loss, blood pressure, and glycemic control after RYGB, but greater improvements in HDL cholesterol and CEC levels after SG. This study sought to determine effects of RYGB and SG on HDL protein and lipid cargo and investigate associations with CEC changes. METHODS We prospectively studied nondiabetic, premenopausal Hispanic women with severe obesity not using lipid medications undergoing RYGB (n=31) or SG (n=36). Anthropometric measurements and blood sampling were obtained before and at 6 and 12 months after surgery. HDL was isolated from plasma, and quantitative proteomic and lipidomic assessments were performed with LC-MS/MS (liquid chromatography with tandem mass spectrometry). CEC was assessed ex vivo using apoB-depleted serum. RESULTS Participants experienced similar, significant weight loss over 12 months following bariatric surgery (38.0±10.4 kg) regardless of the procedure. Relative quantities of 47 proteins (34 increased, 13 decreased) and 150 lipids (71 increased, 79 decreased) carried on HDL were significantly altered following either surgical procedure. Proteins with similar aggregate response patterns were clustered into 15 groups (5 increased, 5 decreased, 5 minimal change) and lipids with similar aggregate responses into 25 groups (7 increased, 11 decreased, 7 minimal change). Network mediation analyses suggested that changes in 4 protein and 2 lipid clusters mediated changes in ABCA1 (ATP-binding cassette transporter A1) CEC and that 1 lipid cluster mediated changes in non-ABCA1 CEC. The protein and lipid clusters that mediated changes in CEC were distinct between SG and RYGB. CONCLUSIONS Bariatric surgery produces substantial changes in HDL lipid and protein cargo, and specific changes may mediate changes in HDL function in CEC. Further study of these mechanisms may lead to improved interventions to reduce cardiovascular risk in patients with obesity.
Collapse
Affiliation(s)
- Sohail Zahid
- Department of Medicine, Leon H. Charney Division of Cardiology (S.Z., B.-X.L., A.C., M.F., E.A.F., S.P.H.), New York University Langone Medical Center
| | - Florencia Schlamp
- NYU Cardiovascular Research Center (F.S., M.A.G., E.A.F., S.P.H.), New York University Langone Medical Center
| | - Michael A Gildea
- NYU Cardiovascular Research Center (F.S., M.A.G., E.A.F., S.P.H.), New York University Langone Medical Center
| | - Bing-Xue Lin
- Department of Medicine, Leon H. Charney Division of Cardiology (S.Z., B.-X.L., A.C., M.F., E.A.F., S.P.H.), New York University Langone Medical Center
| | - Ariya Chaloemtoem
- Department of Medicine, Leon H. Charney Division of Cardiology (S.Z., B.-X.L., A.C., M.F., E.A.F., S.P.H.), New York University Langone Medical Center
| | - Marcin Falis
- Department of Medicine, Leon H. Charney Division of Cardiology (S.Z., B.-X.L., A.C., M.F., E.A.F., S.P.H.), New York University Langone Medical Center
| | - Manish Parikh
- Department of Surgery, New York University Langone Medical Center (M.P.)
| | - Edward A Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology (S.Z., B.-X.L., A.C., M.F., E.A.F., S.P.H.), New York University Langone Medical Center
- NYU Cardiovascular Research Center (F.S., M.A.G., E.A.F., S.P.H.), New York University Langone Medical Center
- NYU Center for the Prevention of Cardiovascular Disease (E.A.F., S.P.H.), New York University Langone Medical Center
| | | | - Tomas Vaisar
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine, Seattle (T.V.)
| | - Sean P Heffron
- Department of Medicine, Leon H. Charney Division of Cardiology (S.Z., B.-X.L., A.C., M.F., E.A.F., S.P.H.), New York University Langone Medical Center
- NYU Cardiovascular Research Center (F.S., M.A.G., E.A.F., S.P.H.), New York University Langone Medical Center
- NYU Center for the Prevention of Cardiovascular Disease (E.A.F., S.P.H.), New York University Langone Medical Center
| |
Collapse
|
2
|
Liu X, Zhang Z, Aguirre T, Shipton ML, Fu L, Du J, Furkert D, Qi J, Chin AC, Riley AM, Liu T, Zhang X, Potter BVL, Fiedler D, Zhu Y, Fu C. Inhibiting IP6K1 confers atheroprotection by elevating circulating apolipoprotein A-I. Metabolism 2025; 163:156098. [PMID: 39643078 PMCID: PMC7617243 DOI: 10.1016/j.metabol.2024.156098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/29/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND AND AIMS Atherosclerotic cardiovascular diseases are the leading cause of death. Apolipoprotein A-I (apoA-I) mediates cholesterol efflux to lower the risks of atherosclerosis. Elevating circulating apoA-I is an effective strategy for atheroprotection. However, the regulatory mechanisms of apoA-I have been elusive. METHODS Protein-protein interactions were examined by co-immunoprecipitations. Chemical biology tools were used to determine the binding of 5PP-InsP5 to its target proteins and its roles in mediating protein-protein interactions. The mouse atherosclerotic model was generated by injecting AAV-PCSK9 and feeding a Western diet. Atherosclerotic plaques were determined by Oil Red O and H&E staining. RESULTS We show that blocking IP6K1 activity increases apoA-I production in hepatocytes. IP6K1 binds to apoA-I and via its product 5PP-InsP5 to induce apoA-I degradation, which requires ubiquitination factor E4A (UBE4A). Depleting 5PP-InsP5 by deleting IP6K1 or blocking IP6K1 activity disrupts the interaction between UBE4A and apoA-I, preventing apoA-I degradation, leading to increased production of apoA-I. Hepatocyte-specific deletion of IP6K1 elevates circulating apoA-I levels, which augments cholesterol efflux and lowers the burden of atherosclerosis. Mice with both apoA-I KO and hepatocyte-specific IP6K1 KO were generated to validate that IP6K1 deletion-induced atheroprotection requires apoA-I. CONCLUSIONS Our findings reveal a mechanism by which blocking IP6K1 boosts apoA-I production. Blocking IP6K1 represents a potential treatment strategy to elevate circulating apoA-I for atheroprotection.
Collapse
Affiliation(s)
- Xiaoqi Liu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Tianjin 300070, China
| | - Zixuan Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Tianjin 300070, China
| | - Tim Aguirre
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin 13125, Germany
| | - Megan L Shipton
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Lin Fu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Jimin Du
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Tianjin 300070, China
| | - David Furkert
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin 13125, Germany
| | - Ji Qi
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Alfred C Chin
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Andrew M Riley
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Tianjin 300070, China
| | - Barry V L Potter
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Dorothea Fiedler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin 13125, Germany
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Tianjin 300070, China.
| | - Chenglai Fu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Tianjin 300070, China; Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| |
Collapse
|
3
|
Tsuchigauchi S, Matsushige T, Hashimoto Y, Hosogai M, Takahashi H, Kobayashi S, Shishido T, Hara N, Kaneyoshi K, Uchida S, Maki R, Yamashita H. Potential of low apolipoprotein A-I as a surrogate marker of vulnerable carotid artery plaques. J Stroke Cerebrovasc Dis 2025; 34:108231. [PMID: 39837394 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVE Recent studies suggested that the medical control of atherogenic lipoproteins is not sufficient for stroke prevention. A low apolipoprotein A-I (apoA-I) level may play a crucial role in the anti-atherogenic effects of high-density lipoprotein (HDL-C) and may also be associated with symptomatic vulnerable plaques in carotid artery stenosis. Therefore, the present study investigated the relationship between apoA-I levels and the status of carotid artery stenosis. METHODS Ninety-one patients with carotid artery stenosis were examined. The status of carotid artery plaques was divided into symptomatic (n = 47) and asymptomatic (n = 44). We examined patient profiles, including comorbidities, and laboratory lipid data, and plaque features visualized by ultrasonography, MRI, and digital subtraction angiography. The relationships between plaque instability and risk factors for carotid artery stenosis were investigated. RESULTS No significant differences were observed in the profiles of symptomatic and asymptomatic patients. Regarding plaque features, ulceration, low echo luminance, and a high signal intensity in plaques on T1-weighted images correlated with symptomatic plaques. ApoA-I, total cholesterol, and non-HDL-C levels were significantly lower in symptomatic patients than in asymptomatic patients. A multivariate logistic regression analysis identified low ApoA-I levels, ulceration, and low echo luminance as predictive factors for symptomatic carotid artery stenosis. Diagnostic accuracy for predicting symptomatic carotid stenosis was 0.84 when the following four factors were combined: ulceration, low echo luminance, a high signal intensity on T1-weighted images, and the level of apoA-I. CONCLUSIONS A low apoA-I level was associated with symptomatic carotid artery stenosis. Therefore, ApoA-I levels have potential as a surrogate marker to detect unstable carotid artery plaques.
Collapse
Affiliation(s)
- Saya Tsuchigauchi
- Department of Neurology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Toshinori Matsushige
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan.
| | - Yukishige Hashimoto
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan; Department of Neurosurgery, JA Onomichi general hospital, Hiroshima, Japan
| | - Masahiro Hosogai
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Hiroki Takahashi
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Shohei Kobayashi
- Department of Neurosurgery and Interventional Neuroradiology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Takeo Shishido
- Department of Neurology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Naoyuki Hara
- Department of Neurology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Kenta Kaneyoshi
- Department of Neurology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan; Department of Internal medicine, Aki Ota Hospital, Hiroshima, Japan
| | - Shota Uchida
- Department of Neurology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Ryuga Maki
- Department of Neurology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Hiroshi Yamashita
- Department of Neurology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| |
Collapse
|
4
|
Chen E, Chen H, Yang Y, Liu M, Wang J, Zhang X, Xiao H, Li J, Feng H, Xu Y. High-density lipoprotein alleviates ocular inflammation by downregulating M1 microglia and pyroptosis through regulating lipid accumulation and Caveolin-1 expression. Int Immunopharmacol 2025; 144:113592. [PMID: 39566385 DOI: 10.1016/j.intimp.2024.113592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024]
Abstract
Uveitis encompasses a group of intraocular inflammatory diseases that are often associated with low levels of high-density lipoprotein (HDL). The role of HDL in intraocular inflammatory diseases remains unclear. In our research, we established an endotoxin-induced uveitis (EIU) model to investigate the role of HDL. Our study indicated that HDL could suppress ocular inflammation and restore retinal function in EIU mice. Specifically, HDL intervention effectively inhibited microglial activation and promoted the transformation of microglia from the M1 phenotype to the M2 phenotype. Furthermore, HDL intervention reduced microglial pyroptosis. Additionally, HDL was found to inhibit lipid accumulation in LPS-induced microglia, which is associated with inflammation, M1 polarization, and pyroptosis, by enhancing the expression of Caveolin-1 (CAV-1). Finally, we demonstrated that the function of HDL may be partially dependent on CAV-1 expression. We conclude that HDL inhibits pathological ocular inflammation by regulating M1/M2 phenotype polarization and pyroptosis through the modulation of lipid accumulation and CAV-1 expression. This suggests that HDL may represent a novel therapeutic strategy for ocular inflammation.
Collapse
Affiliation(s)
- Enguang Chen
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Han Chen
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yuan Yang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Miaomiao Liu
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jianhui Wang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xuerui Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Haodong Xiao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jing Li
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Huazhang Feng
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Yu Xu
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
5
|
Yelamanchili D, Gillard BK, Gotto AM, Achirica MC, Nasir K, Remaley AT, Rosales C, Pownall HJ. HDL-free cholesterol influx into macrophages and transfer to LDL correlate with HDL-free cholesterol content. J Lipid Res 2025; 66:100707. [PMID: 39566848 PMCID: PMC11696839 DOI: 10.1016/j.jlr.2024.100707] [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: 09/04/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/22/2024] Open
Abstract
High-density lipoprotein (HDL)-free cholesterol (FC) transfers to other lipoproteins and cells, the former by a spontaneous mechanism and the latter by both spontaneous and receptor-mediated mechanisms. Macrophages are an important cell type in all stages of atherosclerotic cardiovascular disease (ASCVD), and the magnitude of FC efflux from macrophages to HDL, a metric of HDL function, inversely associated with several metrics of ASCVD. Very high plasma HDL concentrations are associated with increased all-cause and ASCVD mortality, suggesting that the reverse process, FC influx from HDL into macrophages, is atherogenic. We hypothesize that HDL-FC is a metric of dysfunctional HDL, and when combined with HDL particle number (HDL-P), is an ASCVD risk factor. The magnitude of FC influx from HDL to macrophages is expected to be a function of HDL-P and HDL-FC content. Here we show that plasma HDL-FC content varies 2-fold among normolipidemic human subjects and linearly correlates with low-density lipoprotein (LDL)-FC content. The influx of HDL-FC into macrophages and transfer to LDL increase linearly with HDL-FC. As expected, the influx of HDL-FC into macrophages and the transfer to LDL are positively correlated. These data support the hypothesis that high HDL FC content is a marker for dysfunctional HDL, resulting in greater influx into macrophages and greater HDL-FC transfer to LDL. HDL-FC transfer to LDL is a valid surrogate for influx into macrophages. This study of HDL composition and function of normolipidemic subjects provides the basis for further investigation and establishment of HDL-FC content as an ASCVD risk factor.
Collapse
Affiliation(s)
| | - Baiba K Gillard
- Department of Medicine, Houston Methodist, Houston, TX, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Antonio M Gotto
- Department of Medicine, Houston Methodist, Houston, TX, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Khurram Nasir
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Cardiology and Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist, Houston, TX, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Corina Rosales
- Department of Medicine, Houston Methodist, Houston, TX, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Henry J Pownall
- Department of Medicine, Houston Methodist, Houston, TX, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Garcia-Fernandez H, Alcala-Diaz JF, Quintana-Navarro GM, Lopez-Moreno J, Luque-Cordoba D, Ruiz-Diaz Narvaez E, Arenas-de Larriva AP, Gutierrez-Mariscal FM, Torres-Peña JD, Rodriguez-Cano D, Luque RM, Priego-Capote F, Lopez-Miranda J, Camargo A. Trimethylamine Oxidation into the Proatherogenic Trimethylamine N-Oxide Is Higher in Coronary Heart Disease Men: From the CORDIOPREV Study. World J Mens Health 2025; 43:249-258. [PMID: 39344118 PMCID: PMC11704170 DOI: 10.5534/wjmh.230366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 10/01/2024] Open
Abstract
PURPOSE Cardiovascular disease (CVD) is more prevalent in men than women, but the mechanisms responsible for this are not fully understood. We aimed to evaluate differences in trimethylamine (TMA), a microbial metabolite and its oxidized form, trimethylamine N-oxide (TMAO), which is thought to promote atherosclerosis, between men and women with coronary heart disease (CHD), using as a reference a non-CVD population. MATERIALS AND METHODS This study was carried out within the framework of the CORDIOPREV study (NCT00924937; June 19, 2009), a clinical trial which included 827 men and 175 women with CHD, with a non-CVD population of 375 individuals (270 men and 105 women) as a reference group. Plasma TMA and TMAO were measured by HPLC-MS/MS. The carotid study was ultrasonically assessed bilaterally by the quantification of intima-media thickness of both common carotid arteries (IMT-CC). RESULTS We found higher TMAO levels and TMAO/TMA ratio in CHD men than CHD women (p=0.034 and p=0.026, respectively). No TMA sex differences were found in CHD patients. The TMA and TMAO levels and TMAO/TMA ratio were lower, and no differences between sexes were found in the non-CVD population. TMAO levels in CHD patients were consistent with higher IMT-CC and more carotid plaques (p=0.032 and p=0.037, respectively) and lower cholesterol efflux in CHD men than CHD women (p<0.001). CONCLUSIONS Our results suggest that CHD men have augmented TMAO levels compared with CHD women, presumably as a consequence of higher rate of TMA to TMAO oxidation, which could be associated with CVD, as these sex differences are not observed in a non-CVD population.
Collapse
Affiliation(s)
- Helena Garcia-Fernandez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Gracia M Quintana-Navarro
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Lopez-Moreno
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Diego Luque-Cordoba
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- Department of Analytical Chemistry and Nanochemistry University Institute, University of Cordoba, Cordoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Eugenia Ruiz-Diaz Narvaez
- Department of Clinical Nutrition and Diet Therapy, Clinics Hospital, Faculty of Medical Sciences, National University of Asuncion, San Lorenzo, Paraguay
| | - Antonio P Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco M Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Raul M Luque
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Feliciano Priego-Capote
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- Department of Analytical Chemistry and Nanochemistry University Institute, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Antonio Camargo
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
7
|
Qiu L, Xu F, Dong B. Association Between High-Density Lipoprotein Cholesterol and Length of Hospital Stay in Acute Pancreatitis: A Retrospective Cohort Study. Int J Gen Med 2024; 17:6545-6556. [PMID: 39759892 PMCID: PMC11697649 DOI: 10.2147/ijgm.s487993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/14/2024] [Indexed: 01/07/2025] Open
Abstract
Background Acute pancreatitis (AP) is a complex inflammatory disorder with varying degrees of severity, impacting patient recovery and healthcare resource utilization. The length of hospital stay (LOS) is a pivotal indicator of recovery, and identifying factors influencing LOS can offer insights into AP management. High-density lipoprotein cholesterol (HDL-C), known for its cardioprotective properties, has been posited to influence AP outcomes; however, its relationship with LOS remains unclear. Objective This study aimed to investigate the potential correlation between HDL-C levels and LOS in patients with AP, considering the effects of demographic factors, comorbidities, and other clinical parameters. Methods A retrospective cohort study was conducted. Data collection adhered to the STROBE guidelines, and baseline clinical and laboratory variables were analyzed. Statistical analysis comprised univariate and multivariate regression models, Generalized Additive Models (GAM), and stratified linear regression models to assess the relationship between HDL-C and LOS, while accounting for confounding factors. Results After adjusting for key confounders, including age, sex, BMI, WBC, HB, PLT, CRP, ALT, AMY, TB, GLU, LDL-C, SCR, BUN, ALB, Ca2+, and the presence of comorbidities such as hypertension, gallstones, diabetes mellitus, liver dysfunction, renal insufficiency, smoking and alcohol consumption, the study revealed a nonlinear relationship between HDL-C levels and LOS, with an inflection point at 1.5 mmol/L. Below this threshold, HDL-C was significantly and inversely correlated with LOS, whereas above this threshold, HDL-C was positively correlated with LOS. Subgroup analyses emphasized that in non-diabetic, non-alcoholic and non-hyperlipidemic pancreatitis patients, there is a negative correlation between HDL-C levels and LOS. Conclusion HDL-C exhibits a U-shaped relationship with LOS in patients with AP, suggesting that both low and high levels of HDL-C may influence hospital stay duration. These findings underscore the importance of considering HDL-C levels in the clinical management of AP. Especially in patients who are non-diabetic, non-hyperlipidemic, and non-alcoholic, the management of HDL-C may significantly reduce hospital stay.
Collapse
Affiliation(s)
- Lingyan Qiu
- Department of Gastroenterology, Ningbo No. 2 Hospital, Ningbo, Zhejiang Province, 315010, People’s Republic of China
| | - Fanfan Xu
- Department of Gastroenterology, Shengzhou People’s Hospital (Shengzhou Branch of the First Affiliated Hospital of Zhejiang University School of Medicine), Shaoxing, Zhejiang Province, 312400, People’s Republic of China
| | - Buyuan Dong
- Department of Gastroenterology, Ningbo No. 2 Hospital, Ningbo, Zhejiang Province, 315010, People’s Republic of China
| |
Collapse
|
8
|
Huang X, Hu L, Li J, Wang X. U-shaped association of uric acid to HDL cholesterol ratio (UHR) with ALL-cause and cardiovascular mortality in diabetic patients: NHANES 1999-2018. BMC Cardiovasc Disord 2024; 24:744. [PMID: 39725874 DOI: 10.1186/s12872-024-04436-3] [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: 08/07/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVE To investigate the relationship between the uric acid to high-density lipoprotein cholesterol ratio (UHR) and ALL-cause and cardiovascular mortality among diabetic patients. METHODS This study utilized health data from diabetic patients included in the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2018. The Kaplan-Meier curves was employed to preliminarily explore the association between UHR, its components, and all-cause and cardiovascular mortality in diabetic patients, as well as to analyze UHR levels and mortality across different genders. Subsequently, the Cox proportional hazards model was used to further investigate the relationship between UHR, its components, and mortality in diabetic patients. Restricted cubic spline (RCS) curves were applied to examine the nonlinear relationship between UHR, its components, and mortality, with a particular focus on the association between UHR and mortality across different genders. RESULTS This longitudinal cohort study included a total of 6,370 participants, comprising 3,268 males and 3,102 females. Kaplan-Meier analysis revealed a positive correlation between UHR, UA, and mortality in diabetic patients, while the association between HDL and mortality was negligible. The Cox proportional hazards model demonstrated a positive association between UHR and mortality in the diabetic population, while the statistical effects of UA and HDL on mortality were less pronounced compared to UHR. When analyzed by gender, no significant linear relationship was observed between UHR and mortality in either males or females. Subsequently, RCS analysis indicated a U-shaped nonlinear relationship between UHR and mortality in the overall diabetic population and among female patients, with a similar trend observed in males. Furthermore, stratified RCS analysis confirmed the persistence of the U-shaped relationship between UHR and prognosis across most subgroups. CONCLUSION This study found a U-shaped relationship between UHR and both ALL-cause and cardiovascular mortality in diabetic population. This suggests that clinicians should control UHR around 9-10 to improve the long-term prognosis of diabetic patients.
Collapse
Affiliation(s)
- Xuanchun Huang
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Lanshuo Hu
- Xiyuan Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Jun Li
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China.
| | - Xuejiao Wang
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China.
| |
Collapse
|
9
|
Andersen CJ, Fernandez ML. Emerging Biomarkers and Determinants of Lipoprotein Profiles to Predict CVD Risk: Implications for Precision Nutrition. Nutrients 2024; 17:42. [PMID: 39796476 PMCID: PMC11722654 DOI: 10.3390/nu17010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Biomarkers constitute a valuable tool to diagnose both the incidence and the prevalence of chronic diseases and may help to inform the design and effectiveness of precision nutrition interventions. Cardiovascular disease (CVD) continues to be the foremost cause of death all over the world. While the reasons that lead to increased risk for CVD are multifactorial, dyslipidemias, plasma concentrations of specific lipoproteins, and dynamic measures of lipoprotein function are strong biomarkers to predict and document coronary heart disease incidence. The aim of this review is to provide a comprehensive evaluation of the biomarkers and emerging approaches that can be utilized to characterize lipoprotein profiles as predictive tools for assessing CVD risk, including the assessment of traditional clinical lipid panels, measures of lipoprotein efflux capacity and inflammatory and antioxidant activity, and omics-based characterization of lipoprotein composition and regulators of lipoprotein metabolism. In addition, we discuss demographic, genetic, metagenomic, and lifestyle determinants of lipoprotein profiles-such as age, sex, gene variants and single-nucleotide polymorphisms, gut microbiome profiles, dietary patterns, physical inactivity, obesity status, smoking and alcohol intake, and stress-which are likely to be essential factors to explain interindividual responses to precision nutrition recommendations to mitigate CVD risk.
Collapse
Affiliation(s)
- Catherine J. Andersen
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
- School of Nutrition and Wellness, University of Arizona, Tucson, AZ 85712, USA
| |
Collapse
|
10
|
Nijssen KMR, Chavez-Alfaro MA, Joris PJ, Plat J, Mensink RP. Effects of Longer-Term Mixed Nut Consumption on Lipoprotein Particle Concentrations in Older Adults with Overweight or Obesity. Nutrients 2024; 17:8. [PMID: 39796442 PMCID: PMC11723242 DOI: 10.3390/nu17010008] [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: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Recently, we reported that longer-term mixed nut intake significantly reduced serum total and low-density lipoprotein (LDL)-cholesterol, but these markers may not fully capture lipoprotein-related cardiovascular disease (CVD) risk. OBJECTIVES This randomized, controlled, single-blinded, crossover trial in older adults with overweight or obesity examined the effects of longer-term mixed nut consumption on lipoprotein particle size, number, and lipid distribution. METHODS Twenty-eight participants (aged 65 ± 3 years; BMI 27.9 ± 2.3 kg/m2) completed two 16-week periods (control [no nuts] vs. mixed nuts (60 g/day: 15 g of walnuts, pistachios, cashews, and hazelnuts), separated by an 8-week washout. Plasma lipoprotein particle numbers, sizes, and lipid distributions across subclasses were analyzed using high-throughput nuclear magnetic resonance (NMR) spectroscopy. RESULTS Mixed nut consumption significantly reduced Apolipoprotein B (ApoB) concentrations (-0.07 g/L; p = 0.009), total cholesterol (-0.27 mmol/L; p = 0.047), non-HDL cholesterol (-0.28 mmol/L; p = 0.022), and total triacylglycerol (TAG) (-0.27 mmol/L; p = 0.008). Total very large-density lipoprotein (VLDL) particle numbers decreased by 24 nmol/L (p < 0.001), with reductions observed across all VLDL subclasses. Total LDL particle numbers (p = 0.044), specifically intermediate-density lipoprotein (IDL) (p = 0.002) and large LDL particles (p = 0.015), were also reduced, while HDL particle numbers and sizes were unaffected. The mixed nut intervention significantly reduced cholesterol concentrations across all VLDL subclasses and IDL (all p < 0.01), with no changes in LDL or HDL subclasses. TAG concentrations showed reductions across all lipoprotein subclasses (all p < 0.05). CONCLUSIONS Longer-term mixed nut consumption may lower CVD risk in older adults and favorable shifts in apoB-containing lipoprotein subclasses towards a less atherogenic profile.
Collapse
Affiliation(s)
- Kevin M. R. Nijssen
- Department of Nutrition and Movement Sciences, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (M.A.C.-A.); (P.J.J.); (J.P.); (R.P.M.)
| | | | | | | | | |
Collapse
|
11
|
Orozco Morales JA, Medina Urrutia AX, Tamayo MT, Reyes Barrera J, Galarza EJ, Juárez Rojas JG, Dies Suarez P, Méndez Sánchez N, Díaz Orozco LE, Velázquez-López L, Medina Bravo P. Impact of metabolic-associated fatty liver disease on the cholesterol efflux capacity of high-density lipoproteins in adolescents with type 2 diabetes. Front Pediatr 2024; 12:1462406. [PMID: 39776642 PMCID: PMC11703661 DOI: 10.3389/fped.2024.1462406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/05/2024] [Indexed: 01/11/2025] Open
Abstract
Context Type 2 diabetes (DM2) is an emerging disease in the pediatric population. DM2 is associated with metabolic-associated fatty liver disease (MAFLD). High-density lipoproteins (HDLs) are lipoproteins that are believed to have atheroprotective properties that reduce the risk of cardiovascular disease (CVD). Current evidence suggests that the physicochemical and functional features of HDLs may play a key role in the pathogenesis of atherosclerosis. Objective We aimed to assess the impact of MAFLD on cholesterol efflux capacity (CEC) in adolescents with DM2. Design A cross-sectional study. Setting Attention clinic for Children with Diabetes of the Hospital Infantil de México Federico Gómez. Patients or other participants This study included a total of 70 adolescents, 47 of which had DM2 and 23 were healthy individuals. Interventions The presence of MAFLD was determined by MR spectroscopy with proton density fat fraction. We compared the distribution of HDL subtypes (HDL2b, HDL2a, HDL3a, HDL3b, and HDL3c) and the chemical composition of HDLs (total protein, triglycerides, phospholipids, cholesteryl esters, and free cholesterol). HDL functionality was determined by the CEC, measuring the fluorescent cholesterol efflux from J774 macrophage cells. Main outcome measures We were expecting to observe a decrease in HDL efflux capacity in adolescents with type 2 diabetes and MAFLD. Results In our study, we observed a prevalence of MAFLD in 66% of adolescents with DM2, similar to that reported in other international studies (60%-80%). In the population with DM2 and MAFLD, we did not observe a decrease in CEC. Initially we found a slight elevation of CEC in adolescents with DM2, however, with the increase in liver fat, a little decrease is observed, which could explain a probable metabolic phenomenon, since the physicochemical composition and distribution of the particles is associated with the percentage of liver fat. A positive correlation between the percentage of liver fat and the concentration of HDL2b (p = 0.011), HDL2a (p = 0.014) and average particle size (p = 0.011) and the proportion of triglycerides inside the particles (p = 0.007). Likewise, negative correlation were found with the percentage of liver fat, cholesterol esters (p = 0.010) and free cholesterol of the particles (p < 0.001). We observed a positive correlation between CEC and the percentage of triglycerides (p = 0.007), and a negative correlation with the percentage of cholesterol esters (p = 0.05) inside the HDL's particles. Conclusions In this group of adolescents with DM2, the presence of MAFLD was not associated with CEC; however, it is associated with abnormalities in the distribution and lipid composition of HDL particles. The momentum generated by the original proposal for MAFLD in the adult population and following the recommendations for pediatric MAFLD will be a step forward in helping to study the impact of MAFLD on the atheroprotective properties of HDL in the pediatric population.
Collapse
Affiliation(s)
- José Antonio Orozco Morales
- Department of Endocrinology, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
- Odontological and Health Sciences, UniversidadNacional Autónoma de México, Mexico City, Mexico
| | | | - Margarita Torres Tamayo
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
- Supervision Coordination of IMSS-BIENESTAR, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Mexico City, Mexico
| | - Juan Reyes Barrera
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Esteban Jorge Galarza
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | - Pilar Dies Suarez
- Department of Imaging, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Nahum Méndez Sánchez
- Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Liver Research Unit, Fundación Clínica Médica Sur, Mexico City, Mexico
| | - Luis Enrique Díaz Orozco
- Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Liver Research Unit, Fundación Clínica Médica Sur, Mexico City, Mexico
| | - Lubia Velázquez-López
- Clinical Epidemiology Research Unit, Hospital Carlos Mac Gregor Sánchez Navarro, Mexican Social Security Institute (Instituto Mexicano del Seguro Social, IMSS), Mexico City, Mexico
| | - Patricia Medina Bravo
- Department of Endocrinology, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
- Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
12
|
Yin J, Zheng C, Li Z, Chang Y, Cao L, Qu Y. Gender differences in the association between the uric acid to high-density lipoprotein cholesterol ratio and diabetes risk: a mediation analysis of c-reactive protein, triglycerides, and insulin resistance. Lipids Health Dis 2024; 23:409. [PMID: 39696515 DOI: 10.1186/s12944-024-02404-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The uric acid to high-density lipoprotein cholesterol ratio (UHR) has emerged as a novel metabolic marker and is proven to be associated with diabetes risk. However, there is still a lack of systematic research regarding its role in gender differences and underlying mechanisms. This study aims to assess the association of UHR with diabetes risk in the context of gender differences and to investigate its mediation effects through metabolic and inflammatory pathways. METHODS This study utilized data from NHANES 2005-2010 and included 6,843 adult participants. Multivariate logistic regression was employed to assess the association between UHR and diabetes risk, and restricted cubic spline (RCS) along with correlation analysis was applied to explore its relationship with metabolic risk factors. Multiple mediation analysis was conducted to evaluate the mediating effects of homeostasis model assessment of insulin resistance (HOMA-IR), triglycerides (TG), and C-reactive protein (CRP) on the association between UHR and diabetes risk. RESULTS In the overall population, UHR was significantly positively associated with diabetes risk, but gender-stratified analysis revealed a stronger predictive effect in women. In the unadjusted model, every unit increase in UHR was linked to an 18.6% increase in diabetes risk in women (p < 0.001). In the quartile analysis, women in the highest quartile showed an 8.49-fold increased risk of diabetes (OR = 8.494, 95% CI: 5.542-13.019, p < 0.001), whereas no significant association was observed in men (p > 0.05). Mediation analysis revealed that HOMA-IR was the main mediator of the relationship between UHR and diabetes risk, with mediation effects of 64.55%, 118.38%, and 39.09% in the overall population, men, and women, respectively. Additionally, the mediation effect of TG was stronger in men (36.78%) and weaker in women (17.31%). The mediation effect of CRP was relatively minimal across all groups, accounting for 7.62% in men and 2.67% in women. CONCLUSION This study demonstrates that the association between UHR and diabetes risk exhibits gender differences, with higher diabetes risk observed in women, while men show stronger mediation effects in insulin resistance, lipid metabolism, and inflammatory response.
Collapse
Affiliation(s)
- Jianming Yin
- School of Basic Medical Sciences , Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chuanjie Zheng
- School of Basic Medical Sciences , Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zhan Li
- School of Basic Medical Sciences , Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ying Chang
- School of Basic Medical Sciences , Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lingyong Cao
- School of Basic Medical Sciences , Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yiqian Qu
- School of Basic Medical Sciences , Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
13
|
Sun X, Chen Y, Yang C, Yang S, Lin W, Quan B, Pan X, Ding Q, Chen X, Wang C, Qin W. Chemical Recording of Pump-Specific Drug Efflux in Living Cells. Angew Chem Int Ed Engl 2024; 63:e202409282. [PMID: 39324755 DOI: 10.1002/anie.202409282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
Drug efflux-a process primarily facilitated by efflux pumps such as multidrug resistance proteins (MRPs)-plays a pivotal role in cellular resistance to chemotherapies. Conventional approaches to assess drug efflux are predominantly conducted in vitro and often lack pump specificity. Here we report the bioorthogonal reporter inhibiting efflux (BRIEF) strategy, which enables the recording of pump-specific drug efflux in living cells. In BRIEF, a specific substrate is engineered as a bioorthogonal efflux probe (BEP) for specific pumps. The cellular concentration and protein labeling level of the probe can be augmented when the test drug is transported by the same pumps. Serendipitously, we discovered that per-O-acetylated unnatural monosaccharides, initially designed for metabolic glycan labeling, are exported by some MRPs. Using Ac4GlcNAl as a BEP, we studied the structure-efflux relationship of flavonoids and identified small molecules, including tannic acid, cholesterol and gallic acid, as novel MRP substrates in high-throughput screening. Tannic acid, known for anti-tumor and anti-SARS-CoV-2 properties, showed increased efficacy upon MRP inhibition. Additionally, BRIEF was adapted to assess p-glycoprotein-mediated efflux using Rhodamine 123 as a BEP, leveraging its light-activatable proximity labeling ability. BRIEF provides a versatile approach to investigate drug efflux and enhance chemotherapy strategies.
Collapse
Affiliation(s)
- Xuege Sun
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, The State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, 100871, China
| | - Ying Chen
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences and MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Chen Yang
- School of Medicine, Tsinghua University, Beijing, 100871, China
| | - Song Yang
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences and MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Wei Lin
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences and MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Baiyi Quan
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences and MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Xuanzhen Pan
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, The State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, 100871, China
| | - Qiang Ding
- School of Medicine, Tsinghua University, Beijing, 100871, China
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences and MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Chu Wang
- College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences and MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Wei Qin
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, The State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, 100871, China
| |
Collapse
|
14
|
Rhodes CH, Hong BV, Tang X, Weng CY, Kang JW, Agus JK, Lebrilla CB, Zivkovic AM. Absorption, anti-inflammatory, antioxidant, and cardioprotective impacts of a novel fasting mimetic containing spermidine, nicotinamide, palmitoylethanolamide, and oleoylethanolamide: A pilot dose-escalation study in healthy young adult men. Nutr Res 2024; 132:125-135. [PMID: 39549554 DOI: 10.1016/j.nutres.2024.10.006] [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: 06/05/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/18/2024]
Abstract
This pilot dose-escalation study evaluated the absorption and metabolism of a novel fasting mimetic formulation containing spermidine, nicotinamide, palmitoylethanolamide (PEA), and oleoylethanolamide (OEA) taken as oral supplements in young adults. Five healthy men consumed a standardized breakfast, followed by control (wheat flour) or low, medium, or high doses of supplements containing spermidine, nicotinamide, PEA, and OEA 2 hours later. Blood was drawn at 0, 1, 2, and 4 hours after the supplement (2, 3, 4, and 6 hours postprandial). Plasma concentrations of spermidine, 1-methylnicotinamide, PEA and OEA were quantified by liquid chromatography-mass spectrometry. The secretion of tumor necrosis factor alpha and production of reactive oxygen species by stimulated macrophages incubated with plasma, and cholesterol efflux capacity of plasma were analyzed. Plasma 1-methylnicotinamide, PEA, and OEA concentrations increased after supplement intake (P < .05). Spermidine concentrations decreased in the control arm (P < .05) but not the supplement arms. Net incremental area under the curve for tumor necrosis factor alpha and reactive oxygen species in stimulated macrophages decreased when incubated with plasma following supplement intake (P < .05). Intake of the combined supplements showed they were bioavailable and increased in plasma in a dose-dependent manner and provide preliminary data showing enhanced plasma anti-inflammatory and antioxidant functions. This trial was registered at clinicaltrials.gov (NCT05017428).
Collapse
Affiliation(s)
| | - Brian V Hong
- Department of Nutrition, University of California, Davis, California
| | - Xinyu Tang
- Department of Nutrition, University of California, Davis, California
| | - Cheng-Yu Weng
- Department of Chemistry, University of California, Davis, California
| | - Jea Woo Kang
- Department of Nutrition, University of California, Davis, California
| | - Joanne K Agus
- Department of Nutrition, University of California, Davis, California
| | | | - Angela M Zivkovic
- Department of Nutrition, University of California, Davis, California.
| |
Collapse
|
15
|
Dunne S, Gibney ER, McGillicuddy FC, Feeney EL. The effects of saturated fat intake from dairy on CVD markers: the role of food matrices. Proc Nutr Soc 2024; 83:236-244. [PMID: 38316603 DOI: 10.1017/s0029665124000132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
CVD is the leading cause of death worldwide, and is commonly associated with modifiable risk factors. Most studies to date examining link between food intake and risk of CVD, have focused on modulation of plasma cholesterol concentrations (total cholesterol (TC), LDL-C). However, recent studies suggest LDL particle size is a more sensitive risk marker for CVD with smaller, dense LDL particles reported as more atherogenic than larger, more buoyant LDL. Although dietary guidelines recommend SFA intake of < 10 % of total energy, this does not consider food source, with recent evidence suggesting differing, sometimes beneficial, lipid responses following consumption of SFA from dairy compared to other food sources. This may be from differences in the physical food matrices, the nutrient content of the foods, and/or how these components interact with each other, described as a 'dairy matrix effect'. Dietary fat not only raises LDL-C, but also HDL cholesterol (HDL-C), associated with reduced CVD risk. HDL particles are complex emulsions of lipids, proteins and microRNAs that exhibit atheroprotective properties. In addition, HDL particles exhibit a very heterogeneous proteomic composition, dependent on a person's disease state - with a more pro-inflammatory proteome evident in patients with established CVD. This review will discuss the evidence to date on the importance of the food matrix in modulating response to dietary SFA and impact on CVD risk factors. A focus on potential biomarker properties of lipoprotein particles beyond cholesterol and current use of such biomarkers in human nutrition research will be considered.
Collapse
Affiliation(s)
- Simone Dunne
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- UCD Institute for Food and Health, University College Dublin, Dublin 4, Republic of Ireland
| | - Eileen R Gibney
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- UCD Institute for Food and Health, University College Dublin, Dublin 4, Republic of Ireland
| | - Fiona C McGillicuddy
- UCD Diabetes Complications Research Centre, University College Dublin, Dublin 4, Republic of Ireland
| | - Emma L Feeney
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- UCD Institute for Food and Health, University College Dublin, Dublin 4, Republic of Ireland
| |
Collapse
|
16
|
Perkins ZM, Smith DK, Yancey PG, Linton MF, Smith LE. High-Density Lipoprotein Anti-Inflammatory Capacity and Acute Kidney Injury After Cardiac and Vascular Surgery: A Prospective Observational Study. Crit Care Med 2024; 52:e616-e626. [PMID: 39404489 PMCID: PMC11620948 DOI: 10.1097/ccm.0000000000006440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
OBJECTIVES Acute kidney injury (AKI) predicts death after cardiac and vascular surgery. Higher preoperative high-density lipoprotein (HDL) concentrations are associated with less postoperative AKI. In animals, HDL's anti-inflammatory capacity to suppress endothelial cell adhesion molecule expression reduces kidney damage due to ischemia and hemorrhagic shock. The objective of this study is to evaluate the statistical relationship between HDL anti-inflammatory capacity and AKI after major cardiac and vascular surgery. DESIGN Prospective observational study. SETTING Quaternary medical center. PATIENTS One hundred adults with chronic kidney disease on long-term statin therapy undergoing major elective cardiac and vascular surgery. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Apolipoprotein B-depleted serum collected at anesthetic induction was incubated with tumor necrosis factor alpha stimulated human endothelial cells. Reverse transcriptase-polymerase chain reaction was used to measure intercellular adhesion molecule-1 (ICAM-1) messenger RNA. Enzyme-linked immunosorbent assay assays were used to measure apolipoprotein A-I and postoperative soluble ICAM-1 concentrations in patient plasma. HDL concentration did not correlate with HDL ICAM-1 suppression capacity (Spearman R = 0.05; p = 0.64). Twelve patients (12%) were found to have dysfunctional, pro-inflammatory HDL. Patients with pro-inflammatory HDL had a higher rate of postoperative AKI than patients with anti-inflammatory HDL ( p = 0.046). After adjustment for AKI risk factors, a higher preoperative HDL capacity to suppress endothelial ICAM-1 was independently associated with lower odds of AKI (odds ratio, 0.88; 95% CI, 0.80-0.98; p = 0.016). The association between HDL anti-inflammatory capacity and postoperative AKI was independent of HDL concentration ( p = 0.018). Further, a higher long-term statin dose was associated with higher HDL capacity to suppress endothelial ICAM-1 ( p = 0.045). CONCLUSIONS Patients with chronic kidney disease undergoing cardiac and vascular surgery who have dysfunctional, pro-inflammatory HDL have a higher risk of postoperative AKI compared with patients with anti-inflammatory HDL. Conversely, a higher HDL anti-inflammatory capacity is associated with a lower risk of postoperative AKI, independent of HDL concentration. Higher long-term statin dose is associated with higher HDL anti-inflammatory capacity.
Collapse
Affiliation(s)
| | - Derek K Smith
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Patricia G Yancey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - MacRae F Linton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| | - Loren E Smith
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
17
|
Saenz-Pipaon G, Wacker BK, Bi L, Stamatikos A, Dichek DA. Exosome-Mediated Transfer of X-Motif-Tagged Anti-MiR-33a-5p Antagomirs to the Medial Cells of Transduced Rabbit Carotid Arteries. BIOLOGY 2024; 13:965. [PMID: 39765632 PMCID: PMC11673983 DOI: 10.3390/biology13120965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
Atherosclerosis is caused by the accumulation of cholesterol within intimal smooth muscle cells (SMCs) and macrophages. However, the transporter ATP-binding cassette subfamily A, member 1 (ABCA1), can remove cholesterol from these intimal, cells reducing atherosclerosis. Antagomir-mediated inhibition of miR-33a-5p, a microRNA that represses ABCA1 translation, promotes ABCA1-dependent cholesterol efflux and may impede atherosclerosis development. In our previous work, transducing cultured endothelial cells (ECs) with a helper-dependent adenoviral vector (HDAd) that expresses X-motif-tagged anti-miR-33a-5p enhanced antagomir packaging into EC-derived exosomes, which delivered the antagomir to cultured SMCs and macrophages. In this present study, we tested whether in vivo transduction of rabbit carotid artery endothelium can deliver an X-motif-tagged anti-miR-33a-5p to subendothelial cells. Rabbit carotid endothelial cells were transduced in vivo with an HDAd expressing anti-miR-33a-5p either with or without the X-motif (n = 11 arteries per vector). Contralateral carotids received HDAd that express scrambled oligonucleotides. Three days after transduction, the antagomir-without the X-motif-was detected in the intima but not in the media of transduced carotids (p = 0.062). The X-motif antagomir was detected in 82% of the intimal extracts (9 out of 11 carotids) and 27% of medial samples (3 out of 11 carotids, p = 0.031). However, the X-motif did not significantly enhance antagomir delivery to the media (p = 0.214 vs. non-X-motif antagomir). Expression of the antagomirs-with and without the X-motif-was sub-stoichiometric in ECs and SMCs. No antagomir-related changes in miR-33a-5p or ABCA1 expressions were detected. Despite its potential as a therapeutic strategy, our exosome-targeted gene transfer system requires further improvements to enhance antagomir expression and delivery to the subendothelial cells.
Collapse
Affiliation(s)
- Goren Saenz-Pipaon
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA 98195, USA; (B.K.W.); (L.B.); (D.A.D.)
| | - Bradley K. Wacker
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA 98195, USA; (B.K.W.); (L.B.); (D.A.D.)
| | - Lianxiang Bi
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA 98195, USA; (B.K.W.); (L.B.); (D.A.D.)
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA;
| | - David A. Dichek
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA 98195, USA; (B.K.W.); (L.B.); (D.A.D.)
| |
Collapse
|
18
|
Baumer Y, Irei J, Boisvert WA. Cholesterol crystals in the pathogenesis of atherosclerosis. Nat Rev Cardiol 2024:10.1038/s41569-024-01100-3. [PMID: 39558130 DOI: 10.1038/s41569-024-01100-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 11/20/2024]
Abstract
The presence of cholesterol crystals (CCs) in tissues was first described more than 100 years ago. CCs have a pathogenic role in various cardiovascular diseases, including myocardial infarction, aortic aneurysm and, most prominently, atherosclerosis. Although the underlying mechanisms and signalling pathways involved in CC formation are incompletely understood, numerous studies have highlighted the existence of CCs at various stages of atheroma progression. In this Review, we summarize the mechanisms underlying CC formation and the role of CCs in cardiovascular disease. In particular, we explore the established links between lipid metabolism across various cell types and the formation of CCs, with a focus on CC occurrence in the vasculature. We also discuss CC-induced inflammation as one of the pathogenic features of CCs in the atheroma. Finally, we summarize the therapeutic strategies aimed at reducing CC-mediated atherosclerotic burden, including approaches to inhibit CC formation in the vasculature or to mitigate the inflammatory response triggered by CCs. Addressing CC formation might emerge as a crucial component in our broader efforts to combat cardiovascular disease.
Collapse
Affiliation(s)
- Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, NIH, NHLBI, Bethesda, MD, USA
| | - Jason Irei
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
| |
Collapse
|
19
|
Castañer O, Pérez-Vega KA, Álvarez S, Vázquez S, Casajoana A, Blanchart G, Gaixas S, Schröder H, Zomeño MD, Subirana I, Muñoz-Aguayo D, Fitó M, Benaiges D, Goday A, Oliveras A. Effect of bariatric surgery on HDL-mediated cholesterol efflux capacity. Front Cardiovasc Med 2024; 11:1469433. [PMID: 39574780 PMCID: PMC11578734 DOI: 10.3389/fcvm.2024.1469433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/10/2024] [Indexed: 11/24/2024] Open
Abstract
Background Bariatric surgery (BS) is the most effective intervention for severe obesity, leading to sustained weight loss, reduced obesity-related comorbidities, and cardiovascular mortality. Aim To assess changes in high-density lipoprotein (HDL) functions [cholesterol efflux capacity (CEC) and anti-inflammatory capacity] at different follow-up times in patients with severe obesity undergoing BS. Methods A prospective observational study within a cohort of consecutively enrolled patients with severe obesity scheduled to undergo BS. In total, 62 participants (77% women), with a mean age of 42.1 years (SD 9.33 years) underwent BS. Regarding the surgical procedure, 27 (43.5%) underwent sleeve gastrectomy and 35 (56.5%) Roux-en-Y gastric bypass. All patients were evaluated preoperatively and at 1, 3, 6, and 12 months after surgery. Results A decrease in body mass index and an improvement in the systemic lipid profile, indicated by reductions in total cholesterol, low-density lipoprotein cholesterol (LDLc), and remnant cholesterol, and an increase in HDL cholesterol (HDLc) was observed (all p trend < 0.001). Time-series comparisons vs. baseline showed that, in general, anthropometric measures, glycemia, total cholesterol, LDLc, and remnant cholesterol decreased at all follow-ups, whereas HDLc and triglyceride concentrations significantly improved vs. baseline from 6 months, reaching at 12 months the highest HDLc levels (29.6%, p < 0.001) and the lowest circulating triglycerides (-30%, p < 0.001). Although HDL's anti-inflammatory ability worsens after surgery, the HDL-mediated CEC linearly increased after surgery (for both p trend < 0.013). Conclusion BS improves the lipid profile both quantitatively and qualitatively after 1 year, specifically enhancing HDL-mediated cholesterol efflux capacity, which may contribute to a reduced cardiovascular risk in individuals with severe obesity.
Collapse
Affiliation(s)
- O. Castañer
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Network Biomedical Research Center Consortium (CIBER), M.P. Epidemiology and Public Health (CIBEResp), Instituto de Salud Carlos III, Madrid, Spain
| | - K. A. Pérez-Vega
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Network Biomedical Research Center Consortium (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - S. Álvarez
- Nephrology Department, Hospital del Mar, Barcelona, Spain
| | - S. Vázquez
- Nephrology Department, Hospital del Mar, Barcelona, Spain
| | - A. Casajoana
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Esophagogastric and Bariatric Surgery Department, General Surgery Service, Hospital del Mar, Barcelona, Spain
| | - G. Blanchart
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
| | - S. Gaixas
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
| | - H. Schröder
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Network Biomedical Research Center Consortium (CIBER), M.P. Epidemiology and Public Health (CIBEResp), Instituto de Salud Carlos III, Madrid, Spain
| | - M. D. Zomeño
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Network Biomedical Research Center Consortium (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Nutrition, Ramon Llull University, Barcelona, Spain
| | - I. Subirana
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Network Biomedical Research Center Consortium (CIBER), M.P. Cardiovascular Diseases (CIBERcv), Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular Epidemiology and Genetics Research Group, IMIM, Barcelona, Spain
| | - D. Muñoz-Aguayo
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Network Biomedical Research Center Consortium (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - M. Fitó
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Network Biomedical Research Center Consortium (CIBER), M.P. Pathophysiology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - D. Benaiges
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Department of Endocrinology and Nutrition, Hospital del Mar, Barcelona, Spain
- Department of Medicine, Universitat Pompeu Fabra, Barcelona, Spain
- Department of Endocrinology and Nutrition, Consorci Sanitari de L'Alt Penedès I Garraf, Vilafranca del Penedès, Spain
| | - A. Goday
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Department of Endocrinology and Nutrition, Hospital del Mar, Barcelona, Spain
- Department of Medicine, Autònoma de Barcelona University, Bellaterra, Spain
| | - A. Oliveras
- Nephrology Department, Hospital del Mar, Barcelona, Spain
- Department of Medicine, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
20
|
de la O V, Fernández-Cruz E, Matía Matin P, Larrad-Sainz A, Espadas Gil JL, Barabash A, Fernández-Díaz CM, Calle-Pascual AL, Rubio-Herrera MA, Martínez JA. Translational Algorithms for Technological Dietary Quality Assessment Integrating Nutrimetabolic Data with Machine Learning Methods. Nutrients 2024; 16:3817. [PMID: 39599604 PMCID: PMC11597732 DOI: 10.3390/nu16223817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Recent advances in machine learning technologies and omics methodologies are revolutionizing dietary assessment by integrating phenotypical, clinical, and metabolic biomarkers, which are crucial for personalized precision nutrition. This investigation aims to evaluate the feasibility and efficacy of artificial intelligence tools, particularly machine learning (ML) methods, in analyzing these biomarkers to characterize food and nutrient intake and to predict dietary patterns. METHODS We analyzed data from 138 subjects from the European Dietary Deal project through comprehensive examinations, lifestyle questionnaires, and fasting blood samples. Clustering was based on 72 h dietary recall, considering sex, age, and BMI. Exploratory factor analysis (EFA) assigned nomenclature to clusters based on food consumption patterns and nutritional indices from food frequency questionnaires. Elastic net regression identified biomarkers linked to these patterns, helping construct algorithms. RESULTS Clustering and EFA identified two dietary patterns linked to biochemical markers, distinguishing pro-Mediterranean (pro-MP) and pro-Western (pro-WP) patterns. Analysis revealed differences between pro-MP and pro-WP clusters, such as vegetables, pulses, cereals, drinks, meats, dairy, fish, and sweets. Markers related to lipid metabolism, liver function, blood coagulation, and metabolic factors were pivotal in discriminating clusters. Three computational algorithms were created to predict the probabilities of being classified into the pro-WP pattern. The first is the main algorithm, followed by a supervised algorithm, which is a simplified version of the main model that focuses on clinically feasible biochemical parameters and practical scientific criteria, demonstrating good predictive capabilities (ROC curve = 0.91, precision-recall curve = 0.80). Lastly, a reduced biochemical-based algorithm is presented, derived from the supervised algorithm. CONCLUSIONS This study highlights the potential of biochemical markers in predicting nutritional patterns and the development of algorithms for classifying dietary clusters, advancing dietary intake assessment technologies.
Collapse
Affiliation(s)
- Víctor de la O
- Cardiometabolic Nutrition Group, Precision Nutrition Program, Research Institute on Food and Health Sciences IMDEA Food, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain; (E.F.-C.); (J.A.M.)
- Faculty of Health Sciences, International University of La Rioja (UNIR), 26004 Logroño, Spain
| | - Edwin Fernández-Cruz
- Cardiometabolic Nutrition Group, Precision Nutrition Program, Research Institute on Food and Health Sciences IMDEA Food, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain; (E.F.-C.); (J.A.M.)
- Faculty of Health Sciences, International University of La Rioja (UNIR), 26004 Logroño, Spain
| | - Pilar Matía Matin
- Endocrinology and Nutrition Department, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (P.M.M.); (A.L.-S.); (A.B.); (A.L.C.-P.); (M.A.R.-H.)
- Department of Medicine II, Faculty of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Angélica Larrad-Sainz
- Endocrinology and Nutrition Department, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (P.M.M.); (A.L.-S.); (A.B.); (A.L.C.-P.); (M.A.R.-H.)
| | - José Luis Espadas Gil
- Endocrinology and Nutrition Department, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (P.M.M.); (A.L.-S.); (A.B.); (A.L.C.-P.); (M.A.R.-H.)
| | - Ana Barabash
- Endocrinology and Nutrition Department, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (P.M.M.); (A.L.-S.); (A.B.); (A.L.C.-P.); (M.A.R.-H.)
- Department of Medicine II, Faculty of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Cristina M. Fernández-Díaz
- GENYAL Platform on Nutrition and Health, Research Institute on Food and Health Sciences IMDEA Food, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain;
| | - Alfonso L. Calle-Pascual
- Endocrinology and Nutrition Department, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (P.M.M.); (A.L.-S.); (A.B.); (A.L.C.-P.); (M.A.R.-H.)
- Department of Medicine II, Faculty of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Miguel A. Rubio-Herrera
- Endocrinology and Nutrition Department, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (P.M.M.); (A.L.-S.); (A.B.); (A.L.C.-P.); (M.A.R.-H.)
- Department of Medicine II, Faculty of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - J. Alfredo Martínez
- Cardiometabolic Nutrition Group, Precision Nutrition Program, Research Institute on Food and Health Sciences IMDEA Food, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain; (E.F.-C.); (J.A.M.)
- Centre of Medicine and Endocrinology, University of Valladolid, 47002 Valladolid, Spain
| |
Collapse
|
21
|
Chen J, Luo Q, Yi Y, Wang J, Chen P, Luo F, Fang Z. ANGPTL3 as a target for treating lipid disorders in type 2 diabetes patients. Lipids Health Dis 2024; 23:356. [PMID: 39487451 PMCID: PMC11528995 DOI: 10.1186/s12944-024-02352-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a globally prevalent metabolic disorder, and cardiovascular disease (CVD) is a significant cause of mortality and morbidity in diabetic individuals. In addition to hyperglycemia, lipid abnormalities associated with T2DM play a crucial role in the development of CVD complications. Diabetic dyslipidemia is characterized by elevated levels of triglyceride (TG)-rich lipoproteins and small dense low-density lipoprotein (LDL) particles, reduced high-density lipoprotein (HDL) cholesterol, and impaired HDL function. Angiopoietin protein-like 3 (ANGPTL3) is a liver-derived protein that plays a crucial role in regulating plasma lipoprotein metabolism by inhibiting lipoprotein lipase and influencing lipid levels. Inhibiting ANGPTL3 has shown promising effects in promoting HDL-mediated cholesterol reverse transport and reducing the levels of TG-rich lipoproteins and LDL cholesterol. Here, we explore the potential of ANGPTL3 as a therapeutic target for lipid management in T2DM patients.
Collapse
Affiliation(s)
- Jingfei Chen
- Research Institute of Blood Lipid and Atherosclerosis, Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Qin Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Yanfeng Yi
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Jiangang Wang
- Department of Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, P.R. China
| | - Pengfei Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China.
| | - Fei Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China.
| | - Zhenfei Fang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China.
| |
Collapse
|
22
|
Nishihara S, Koseki M, Tanaka K, Omatsu T, Sawabe H, Inui H, Saga A, Okada T, Higo T, Ohama T, Nishida M, Sakata Y, Watanabe M. Twin Study: The Factors Affecting the Serum LDL-C and HDL-C Levels and an RNA-Seq Analysis in Mononuclear Cells in Monozygotic Twins. J Atheroscler Thromb 2024; 31:1539-1555. [PMID: 38684403 PMCID: PMC11537789 DOI: 10.5551/jat.64882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/10/2024] [Indexed: 05/02/2024] Open
Abstract
AIM A twin study is a valuable tool for elucidating the acquired factors against lifestyle diseases such as dyslipidemia, diabetes mellitus, and obesity. We aimed 1. to investigate the factors that affect low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) in monozygotic (MZ) twins, and 2. to identify genes which expression levels changed in pairs with large differences in LDL-C or HDL-C levels. METHODS The registered database at the Center for Twin Research, Osaka University, containing 263 pairs of MZ twins, was analyzed. 1. The effects of smoking, exercise, nutritional factors, and anthropometric and biochemical parameters on LDL-C or HDL-C levels were examined in MZ twins. 2. RNA sequencing in the peripheral blood mononuclear cells of 59 pairs was analyzed for large differences of LDL-C or HDL-C groups. RESULTS 1. The ΔLDL-C levels were significantly associated with an older age, the ΔTG levels, and ΔBMI. ΔHDL-C levels were associated with the ΔBMI, ΔTG, ΔTP, and ΔLDL-C levels. The HDL-C levels were affected by smoking and exercise habits. The intakes of cholesterol and saturated fatty acids were not associated with the LDL-C or HDL-C levels. 2. An RNA sequencing analysis revealed that the expression of genes related to the TLR4 and IFNG pathways was suppressed in accordance with the HDL-C levels in the larger ΔHDL-C group among the 59 pairs. CONCLUSION We identified the factors affecting the LDL-C or HDL-C levels in monozygotic twins. In addition, some types of inflammatory gene expression in peripheral blood mononuclear cells were suppressed in accordance with the HDL-C levels, thus suggesting the importance of weight management and exercise habits in addition to dietary instructions to control the LDL-C or HDL-C levels.
Collapse
Affiliation(s)
- Sae Nishihara
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masahiro Koseki
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Katsunao Tanaka
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takashi Omatsu
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Sawabe
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroyasu Inui
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ayami Saga
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Okada
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoaki Higo
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tohru Ohama
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Makoto Nishida
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Health Care Division, Health and Counseling Center, Osaka University, Osaka, Japan
| | - Yasushi Sakata
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Mikio Watanabe
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
- Center for Twin Research, Osaka University Graduate School of Medicine, Osaka, Japan
| | | |
Collapse
|
23
|
Al Zein M, Khazzeka A, El Khoury A, Al Zein J, Zoghaib D, Eid AH. Revisiting high-density lipoprotein cholesterol in cardiovascular disease: Is too much of a good thing always a good thing? Prog Cardiovasc Dis 2024; 87:50-59. [PMID: 39442601 DOI: 10.1016/j.pcad.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Cardiovascular disease (CVD) continues to be a leading cause of global mortality and morbidity. Various established risk factors are linked to CVD, and modifying these risk factors is fundamental in CVD management. Clinical studies underscore the association between dyslipidemia and CVD, and therapeutic interventions that target low-density lipoprotein cholesterol elicit clear benefits. Despite the correlation between low high-density lipoprotein cholesterol (HDLC) and heightened CVD risk, HDL-raising therapies have yet to showcase significant clinical benefits. Furthermore, evidence from epidemiological and genetic studies reveals that not only low HDL-C levels, but also very high levels of HDL-C are linked to increased risk of CVD. In this review, we focus on HDL metabolism and delve into the relationship between HDL and CVD, exploring HDL functions and the observed alterations in its roles in disease. Altogether, the results discussed herein support the conventional wisdom that "too much of a good thing is not always a good thing". Thus, our recommendation is that a careful reconsideration of the impact of high HDL-C levels is warranted, and shall be revisited in future research.
Collapse
Affiliation(s)
- Mohammad Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Alicia Khazzeka
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Jana Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Dima Zoghaib
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
24
|
Niemelä A, Giorgi L, Nouri S, Yurttaş B, Rauniyar K, Jeltsch M, Koivuniemi A. Gliflozins, sucrose and flavonoids are allosteric activators of lecithin-cholesterol acyltransferase. Sci Rep 2024; 14:26085. [PMID: 39478139 PMCID: PMC11525561 DOI: 10.1038/s41598-024-77104-3] [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: 07/24/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Lecithin-cholesterol acyltransferase (LCAT) serves as a pivotal enzyme in preserving cholesterol homeostasis via reverse cholesterol transport, a process closely associated with the onset of atherosclerosis. Impaired LCAT function can lead to severe LCAT deficiency disorders for which no pharmacological treatment exists. LCAT-based therapies, such as small molecule positive allosteric modulators (PAMs), against LCAT deficiencies and atherosclerosis hold promise, although their efficacy against atherosclerosis remains challenging. Herein we utilized a quantitative in silico metric to predict the activity of novel PAMs and tested their potencies with in vitro enzymatic assays. As predicted, sodium-glucose cotransporter 2 (SGLT2) inhibitors (gliflozins), sucrose and flavonoids activate LCAT. This has intriguing implications for the mechanism of action of gliflozins, which are commonly used in the treatment of type 2 diabetes, and for the endogenous activation of LCAT. Our results underscore the potential of molecular dynamics simulations in rational drug design.
Collapse
Affiliation(s)
- Akseli Niemelä
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| | - Laura Giorgi
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Sirine Nouri
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Betül Yurttaş
- Department of Biotechnology and Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Khushbu Rauniyar
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Michael Jeltsch
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Artturi Koivuniemi
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
25
|
Davidson WS, Vaisar T, Heinecke JW, Bornfeldt KE. Distinct roles of size-defined HDL subpopulations in cardiovascular disease. Curr Opin Lipidol 2024:00041433-990000000-00093. [PMID: 39450930 DOI: 10.1097/mol.0000000000000959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW Doubts about whether high-density lipoprotein-cholesterol (HDL-C) levels are causally related to atherosclerotic cardiovascular disease (CVD) risk have stimulated research on identifying HDL-related metrics that might better reflect its cardioprotective functions. HDL is made up of different types of particles that vary in size, protein and lipid composition, and function. This review focuses on recent findings on the specific roles of HDL subpopulations defined by size in CVD. RECENT FINDINGS Small HDL particles are more effective than larger particles at promoting cellular cholesterol efflux because apolipoprotein A-I on their surface better engages ABCA1 (ATP binding cassette subfamily A member 1). In contrast, large HDL particles bind more effectively to scavenger receptor class B type 1 on endothelial cells, which helps prevent LDL from moving into the artery wall. The specific role of medium-sized HDL particles, the most abundant subpopulation, is still unclear. SUMMARY HDL is made up of subpopulations of different sizes of particles, with selective functional roles for small and large HDLs. The function of HDL may depend more on the size and composition of its subpopulations than on HDL-C levels. Further research is required to understand how these different HDL subpopulations influence the development of CVD.
Collapse
Affiliation(s)
- W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Tomas Vaisar
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
| | - Jay W Heinecke
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
| | - Karin E Bornfeldt
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
26
|
Futatsugi A, Tozuka M, Horiuchi Y, Ohkawa R, Kosho T. High-density lipoprotein functionality in cholesterol efflux in early childhood is related to the content ratio of triglyceride to cholesterol. Sci Rep 2024; 14:23323. [PMID: 39375444 PMCID: PMC11458590 DOI: 10.1038/s41598-024-74699-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024] Open
Abstract
Cholesterol efflux capacity (CEC), commonly measured as a useful risk marker of atherosclerotic cardiovascular disease, depends on high-density lipoprotein (HDL) functionality and its concentration. We defined the relative HDL functionality in cholesterol efflux, not influenced by HDL concentration, as the ratio of measured CEC to standardized CEC (stCEC) based on HDL-cholesterol (HDL-C) of each individual using the curve regression equation obtained from the correlation. HDL-C, CEC, and CEC/stCEC levels in the < 28-day-old participants (neonates) were significantly low compared to those of the ≥ 28-day-old participants, indicating that the low CEC levels in the neonates depend on not only lower HDL-C but also lower HDL functionality. The low level of CEC/stCEC was remarkable in neonates born at < 34 weeks of gestation and did not improved to the reference level (1.000) until the infantile period. The relatively low or high CEC/stCEC ratios in neonates and infants were associated with lower or higher HDL-TG and HDL-TG/HDL-C ratio, respectively. However, no apparent effect of HDL-TG and HDL-TG/HDL-C ratio on CEC/stCEC was observed in the ≥ 1-year-old participants, indicating that HDL functionality in cholesterol efflux could be associated with the various HDL particles with various lipid compositions, but not just with HDL-TG and HDL-TG/HDL-C ratio.
Collapse
Affiliation(s)
- Akiko Futatsugi
- Department of Medical Genetics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Department of Clinical Laboratory, Nagano Children's Hospital, 3100 Toyoshina, Azumino, 399-8288, Japan
| | - Minoru Tozuka
- Department of Medical Genetics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
- Life Science Research Center, Nagano Children's Hospital, 3100, Toyoshina, Azumino, 399-8288, Japan.
| | - Yuna Horiuchi
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, 6-8-1 Hinode, Urayasu, 279-0013, Japan
| | - Ryunosuke Ohkawa
- Department of Clinical Bioanalysis and Molecular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Tomoki Kosho
- Department of Medical Genetics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Center for Medical Genetics, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Research Center for Supports to Advanced Science, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- BioBank Shinshu, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| |
Collapse
|
27
|
Vaisar T, Babenko I, Horvath KV, Niisuke K, Asztalos BF. Relationships between HDL subpopulation proteome and HDL function in overweight/obese people with and without coronary heart disease. Atherosclerosis 2024; 397:118565. [PMID: 39260003 PMCID: PMC11539851 DOI: 10.1016/j.atherosclerosis.2024.118565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND AIMS The structure-function relationships of high-density lipoprotein (HDL) subpopulations are not well understood. Our aim was to examine the interrelationships between HDL particle proteome and HDL functionality in subjects with and without coronary heart disease (CHD). METHODS We isolated 5 different HDL subpopulations based on charge, size, and apolipoprotein A1 (APOA1) content from the plasma of 33 overweight/obese CHD patients and 33 age-and body mass index (BMI)-matched CHD-free subjects. We measured the relative molar concentration of HDL-associated proteins by liquid chromatography tandem mass spectrometry (LC-MS/MS) and assessed particle functionality. RESULTS We quantified 110 proteins associated with the 5 APOA1-containing HDL subpopulations. The relative molar concentration of these proteins spanned five orders of magnitude. Only 10 proteins were present in >1% while 73 were present in <0.1% concentration. Only 6 of the 10 most abundant proteins were apolipoproteins. Interestingly, the largest (α-1) and the smallest (preβ-1) HDL particles contained the most diverse proteomes. The protein composition of each HDL subpopulation was altered in CHD cases as compared to controls with the most prominent differences in preβ-1 and α-1 particles. APOA2 concentration was positively correlated with preβ-1 particle functionality (ABCA1-CEC/mg APOA1 in preβ-1) (R2 = 0.42, p = 0.005), while APOE concentration was inversely correlated with large-HDL particle functionality (SRBI-CEC/mg APOA1 in α-1+α-2) (R2 = 0.18, p = 0.01). CONCLUSIONS The protein composition of the different HDL subpopulations was altered differentially in CHD patients. The functionality of the small and large HDL particles correlated with the protein content of APOA2 and APOE, respectively. Our data indicate that distinct particle subspecies and specific particle associated proteins provide new information about the role of HDL in CHD.
Collapse
Affiliation(s)
- Tomas Vaisar
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Ilona Babenko
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Katalin V Horvath
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Katrin Niisuke
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Bela F Asztalos
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
28
|
Davies B, Trelfa L, Rashbrook VS, Drydale E, Martin R, Bai B, Golebka J, Biggs DS, Channon KM, Bhattacharya S, Douglas G. Mutagenesis on a complex mouse genetic background by site-specific nucleases. Transgenic Res 2024; 33:415-426. [PMID: 39088185 PMCID: PMC11588839 DOI: 10.1007/s11248-024-00399-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Mouse models with complex genetic backgrounds are increasingly used in preclinical research to accurately model human disease and to enable temporal and cell-specific evaluation of genetic manipulations. Backcrossing mice onto these complex genetic backgrounds takes time and leads to significant wastage of animals. In this study, we aimed to evaluate whether site-specific nucleases could be used to generate additional genetic mutations in a complex genetic background, using the REVERSA mouse model of atherosclerosis, a model harbouring four genetically altered alleles. The model is comprised of a functional null mutation in the Ldlr gene in combination with a ApoB100 allele, which, after high-fat diet, leads to the rapid development of atherosclerosis. The regression of the pathology is achieved by inducible knock-out of the Mttp gene. Here we report an investigation to establish if microinjection of site-specific nucleases directly into zygotes prepared from the REVERSA could be used to investigate the role of the ATP binding cassette transporter G1 (ABCG1) in atherosclerosis regression. We show that using this approach we could successfully generate two independent knockout lines on the REVERSA background, both of which exhibited the expected phenotype of a significant reduction in cholesterol efflux to HDL in bone marrow-derived macrophages. However, loss of Abcg1 did not impact atherosclerosis regression in either the aortic root or in aortic arch, demonstrating no important role for this transporter subtype. We have demonstrated that site-specific nucleases can be used to create genetic modifications directly onto complex disease backgrounds and can be used to explore gene function without the need for laborious backcrossing of independent strains, conveying a significant 3Rs advantage.
Collapse
Affiliation(s)
- Benjamin Davies
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
- Francis Crick Institute, 1 Midland Road, London, UK
| | - Lucy Trelfa
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Victoria S Rashbrook
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Edward Drydale
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Rachel Martin
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Boyan Bai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Jedrzej Golebka
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Daniel Stephen Biggs
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Shoumo Bhattacharya
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Gillian Douglas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, BHF Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK.
| |
Collapse
|
29
|
Nishida T, Ayaori M, Arakawa J, Suenaga Y, Shiotani K, Uto-Kondo H, Komatsu T, Nakaya K, Endo Y, Sasaki M, Ikewaki K. Liver-specific Lxr inhibition represses reverse cholesterol transport in cholesterol-fed mice. Atherosclerosis 2024; 397:117578. [PMID: 38797615 DOI: 10.1016/j.atherosclerosis.2024.117578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/26/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND AND AIMS High density lipoprotein (HDL) exerts an anti-atherosclerotic effect via reverse cholesterol transport (RCT). Several phases of RCT are transcriptionally controlled by Liver X receptors (Lxrs). Although macrophage Lxrs reportedly promote RCT, it is still uncertain whether hepatic Lxrs affect RCT in vivo. METHODS To inhibit Lxr-dependent pathways in mouse livers, we performed hepatic overexpression of sulfotransferase family cytosolic 2B member 1 (Sult2b1) using adenoviral vector (Ad-Sult2b1). Ad-Sult2b1 or the control virus was intravenously injected into wild type mice and Lxrα/β double knockout mice, under a normal or high-cholesterol diet. A macrophage RCT assay and an HDL kinetic study were performed. RESULTS Hepatic Sult2b1 overexpression resulted in reduced expression of Lxr-target genes - ATP-binding cassette transporter G5/G8, cholesterol 7α hydroxylase and Lxrα itself - respectively reducing or increasing cholesterol levels in HDL and apolipoprotein B-containing lipoproteins (apoB-L). A macrophage RCT assay revealed that Sult2b1 overexpression inhibited fecal excretion of macrophage-derived 3H-cholesterol only under a high-cholesterol diet. In an HDL kinetic study, Ad-Sult2b1 promoted catabolism/hepatic uptake of HDL-derived cholesterol, thereby reducing fecal excretion. Finally, in Lxrα/β double knockout mice, hepatic Sult2b1 overexpression increased apoB-L levels, but there were no differences in HDL levels or RCT compared to the control, indicating that Sult2b1-mediated effects on HDL/RCT and apoB-L were distinct: the former was Lxr-dependent, but not the latter. CONCLUSIONS Hepatic Lxr inhibition negatively regulates circulating HDL levels and RCT by reducing Lxr-target gene expression.
Collapse
MESH Headings
- Animals
- Liver X Receptors/metabolism
- Liver X Receptors/genetics
- Liver/metabolism
- Mice, Knockout
- Biological Transport
- Mice
- Cholesterol/metabolism
- Macrophages/metabolism
- Mice, Inbred C57BL
- Sulfotransferases/metabolism
- Sulfotransferases/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics
- Cholesterol, Dietary
- ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics
- Male
- Cholesterol, HDL/blood
- Cholesterol, HDL/metabolism
- Lipoproteins, HDL/metabolism
- Lipoproteins
Collapse
Affiliation(s)
- Takafumi Nishida
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan.
| | - Makoto Ayaori
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan; Tokorozawa Heart Center, Tokorozawa, Japan
| | - Junko Arakawa
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yumiko Suenaga
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Kazusa Shiotani
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Harumi Uto-Kondo
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Tomohiro Komatsu
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Kazuhiro Nakaya
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yasuhiro Endo
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Makoto Sasaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Katsunori Ikewaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
30
|
Pammer A, Klobučar I, Stadler JT, Meissl S, Habisch H, Madl T, Frank S, Degoricija V, Marsche G. Impaired HDL antioxidant and anti-inflammatory functions are linked to increased mortality in acute heart failure patients. Redox Biol 2024; 76:103341. [PMID: 39244794 PMCID: PMC11406013 DOI: 10.1016/j.redox.2024.103341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024] Open
Abstract
AIMS Acute heart failure (AHF) is typified by inflammatory and oxidative stress responses, which are associated with unfavorable patient outcomes. Given the anti-inflammatory and antioxidant properties of high-density lipoprotein (HDL), this study sought to examine the relationship between impaired HDL function and mortality in AHF patients. The complex interplay between various HDL-related biomarkers and clinical outcomes remains poorly understood. METHODS HDL subclass distribution was quantified by nuclear magnetic resonance spectroscopy. Lecithin-cholesterol acyltransferase (LCAT) activity, cholesterol ester transfer protein (CETP) activity, and paraoxonase (PON-1) activity were assessed using fluorometric assays. HDL-cholesterol efflux capacity (CEC) was assessed in a validated assay using [3H]-cholesterol-labeled J774 macrophages. RESULTS Among the study participants, 74 (23.5 %) out of 315 died within three months after hospitalization due to AHF. These patients exhibited lower activities of the anti-oxidant enzymes PON1 and LCAT, impaired CEC, and lower concentration of small HDL subclasses, which remained significant after accounting for potential confounding factors. Smaller HDL particles, particularly HDL3 and HDL4, exhibited a strong association with CEC, PON1 activity, and LCAT activity. CONCLUSIONS In patients with AHF, impaired HDL CEC, HDL antioxidant and anti-inflammatory function, and impaired HDL metabolism are associated with increased mortality. Assessment of HDL function and subclass distribution could provide valuable clinical information and help identify patients at high risk.
Collapse
Affiliation(s)
- Anja Pammer
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Iva Klobučar
- Department of Cardiology, Sisters of Charity University Hospital Centre, Zagreb, Croatia
| | - Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Sabine Meissl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Hansjörg Habisch
- Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Tobias Madl
- Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Saša Frank
- BioTechMed-Graz, Graz, Austria; Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria.
| | - Vesna Degoricija
- Department of Medicine, Sisters of Charity University Hospital Centre, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
31
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024:S1885-5857(24)00279-2. [PMID: 39357800 DOI: 10.1016/j.rec.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
32
|
Moon HR, Yun JM. Effect of Siegesbeckia glabrescens Extract on Foam Cell Formation in THP-1 Macrophages. Prev Nutr Food Sci 2024; 29:288-300. [PMID: 39371520 PMCID: PMC11450289 DOI: 10.3746/pnf.2024.29.3.288] [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: 07/16/2024] [Revised: 08/14/2024] [Accepted: 08/17/2024] [Indexed: 10/08/2024] Open
Abstract
The accumulation of cholesterol-bearing macrophage foam cells in the initial stages of atherosclerosis serves as a characteristic feature of atherosclerotic lesions. The inhibitory effect of Siegesbeckia glabrescens, a species of flowering plant in the Asteraceae family, on foam cell formation in THP-1 macrophages has not yet been elucidated. In this study, we explored the effect of S. glabrescens ethanol extract (SGEE) and hot water extract (SGWE) on foam cell formation via co-treatment with oxidized low density lipoprotein (ox-LDL) and lipopolysaccharide (LPS), mimicking the occurrence of atherosclerosis in vitro, and studied the regulation of its underlying mechanisms. THP-1 cells differentiated by PMA (1 μM) for 48 h were subsequently treated with/without SGWE and SGEE for 48 h. THP-1 macrophages were treated with ox-LDL (20 μg/mL) and LPS (500 ng/mL) for 24 h. Treatment with ox-LDL and LPS for 24 h enhanced the lipid accumulation in foam cells compared to in untreated cells, as determined by oil red O staining. In contrast, SGWE and SGEE treatment inhibited lipid accumulation in foam cells. Both extracts significantly upregulated ABCA1, LXRα, and PPARγ expression in ox-LDL- and LPS-treated cells (P<0.05). Moreover, both SGWE and SGEE decreased LOX-1, CD36, and SR-A1 expression. The co-treatment of ox-LDL and LPS increased NF-κB, COX-2, and pro-inflammatory activation and expression compared with untreated cells. However, this increase suppressed NF-κB, COX-2, and pro-inflammatory expression by SGWE and SGEE. The results indicated that both extracts can partially inhibit foam cell formation and contribute to protective effects by suppressing cholesterol accumulation during the onset of atherosclerosis.
Collapse
Affiliation(s)
- Ha-Rin Moon
- Department of Food and Nutrition, Chonam National University, Gwangju 61186, Korea
| | - Jung-Mi Yun
- Department of Food and Nutrition, Chonam National University, Gwangju 61186, Korea
| |
Collapse
|
33
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. Rev Esp Cardiol 2024:S1885-5857(24)00279-2. [PMID: 39357800 DOI: 10.1016/j.recesp.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 01/11/2025]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
34
|
Kloosterman DJ, Erbani J, Boon M, Farber M, Handgraaf SM, Ando-Kuri M, Sánchez-López E, Fontein B, Mertz M, Nieuwland M, Liu NQ, Forn-Cuni G, van der Wel NN, Grootemaat AE, Reinalda L, van Kasteren SI, de Wit E, Ruffell B, Snaar-Jagalska E, Petrecca K, Brandsma D, Kros A, Giera M, Akkari L. Macrophage-mediated myelin recycling fuels brain cancer malignancy. Cell 2024; 187:5336-5356.e30. [PMID: 39137777 PMCID: PMC11429458 DOI: 10.1016/j.cell.2024.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 04/26/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024]
Abstract
Tumors growing in metabolically challenged environments, such as glioblastoma in the brain, are particularly reliant on crosstalk with their tumor microenvironment (TME) to satisfy their high energetic needs. To study the intricacies of this metabolic interplay, we interrogated the heterogeneity of the glioblastoma TME using single-cell and multi-omics analyses and identified metabolically rewired tumor-associated macrophage (TAM) subpopulations with pro-tumorigenic properties. These TAM subsets, termed lipid-laden macrophages (LLMs) to reflect their cholesterol accumulation, are epigenetically rewired, display immunosuppressive features, and are enriched in the aggressive mesenchymal glioblastoma subtype. Engulfment of cholesterol-rich myelin debris endows subsets of TAMs to acquire an LLM phenotype. Subsequently, LLMs directly transfer myelin-derived lipids to cancer cells in an LXR/Abca1-dependent manner, thereby fueling the heightened metabolic demands of mesenchymal glioblastoma. Our work provides an in-depth understanding of the immune-metabolic interplay during glioblastoma progression, thereby laying a framework to unveil targetable metabolic vulnerabilities in glioblastoma.
Collapse
Affiliation(s)
- Daan J Kloosterman
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Johanna Erbani
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Menno Boon
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Martina Farber
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Shanna M Handgraaf
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Masami Ando-Kuri
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Elena Sánchez-López
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Bauke Fontein
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Marjolijn Mertz
- Bioimaging Facility, Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Marja Nieuwland
- Genomics Core Facility, Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Ning Qing Liu
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Gabriel Forn-Cuni
- Institute of Biology Leiden, Leiden University, Leiden, the Netherlands
| | - Nicole N van der Wel
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Anita E Grootemaat
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Luuk Reinalda
- The Institute of Chemical Immunology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Sander I van Kasteren
- The Institute of Chemical Immunology, Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Elzo de Wit
- Division of Gene Regulation, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Brian Ruffell
- Department of Immunology, Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Kevin Petrecca
- Montreal Neurological Institute-Hospital, McGill University Health Centre and Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Dieta Brandsma
- Department of Neuro-Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066CX Amsterdam, the Netherlands
| | - Alexander Kros
- Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Leila Akkari
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands.
| |
Collapse
|
35
|
Kempegowda SN, Sugur K, Thimmulappa RK. Dysfunctional HDL Diagnostic Metrics for Cardiovascular Disease Risk Stratification: Are we Ready to Implement in Clinics? J Cardiovasc Transl Res 2024:10.1007/s12265-024-10559-x. [PMID: 39298091 DOI: 10.1007/s12265-024-10559-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024]
Abstract
Epidemiological studies have revealed that patients with higher levels of high-density lipoprotein cholesterol (HDL-C) were more resistant to cardiovascular diseases (CVD), and yet targeting HDL for CVD prevention, risk assessment, and pharmacological management has not proven to be very effective. The mechanistic investigations have demonstrated that HDL exerts anti-atherogenic functions via mediating reverse cholesterol transport, antioxidant action, anti-inflammatory activity, and anti-thrombotic activity. Contrary to expectations, however, adverse cardiovascular events were reported in clinical trials of drugs that raised HDL levels. This has sparked a debate between HDL quantity and quality. Patients with atherosclerotic CVD are associated with dysfunctional HDL, and the degree of HDL dysfunction is correlated with the severity of the disease, independent of HDL-C levels. This growing body of evidence has underscored the need for integrating HDL functional assays in clinical practice for CVD risk management. Because HDL exerts diverse athero-protective functions, there is no single method for capturing HDL functionality. This review critically evaluates the various techniques currently being used for monitoring HDL functionality and discusses key structural changes in HDL indicative of dysfunctional HDL and the technical challenges that need to be addressed to enable the integration of HDL function-based metrics in clinical practice for CVD risk estimation and the development of newer therapies targeting HDL function.
Collapse
Affiliation(s)
- Swetha N Kempegowda
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Kavya Sugur
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Rajesh K Thimmulappa
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India.
| |
Collapse
|
36
|
Pammer A, Obermayer A, Stadler JT, Pferschy PN, Tripolt NJ, Habisch H, Madl T, Sourij H, Marsche G. Effects of dietary interventions and intermittent fasting on HDL function in obese individuals with T2DM: a randomized controlled trial. Cardiovasc Diabetol 2024; 23:339. [PMID: 39267053 PMCID: PMC11395628 DOI: 10.1186/s12933-024-02426-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/29/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Cardiovascular disease represents a significant risk factor for mortality in individuals with type 2 diabetes mellitus (T2DM). High-density lipoprotein (HDL) is believed to play a crucial role in maintaining cardiovascular health through its multifaceted atheroprotective effects and its capacity to enhance glycemic control. The impact of dietary interventions and intermittent fasting (IF) on HDL functionality remains uncertain. The objective of this study was to assess the effects of dietary interventions and IF as a strategy to safely improve glycemic control and reduce body weight on functional parameters of HDL in individuals with T2DM. METHODS Before the 12-week intervention, all participants (n = 41) of the INTERFAST-2 study were standardized to a uniform basal insulin regimen and randomized to an IF or non-IF group. Additionally, all participants were advised to adhere to dietary recommendations that promoted healthy eating patterns. The IF group (n = 19) followed an alternate-day fasting routine, reducing their calorie intake by 75% on fasting days. The participants' glucose levels were continuously monitored. Other parameters were measured following the intervention: Lipoprotein composition and subclass distribution were measured by nuclear magnetic resonance spectroscopy. HDL cholesterol efflux capacity, paraoxonase 1 (PON1) activity, lecithin cholesterol acyltransferase (LCAT) activity, and cholesterol ester transfer protein (CETP) activity were assessed using cell-based assays and commercially available kits. Apolipoprotein M (apoM) levels were determined by ELISA. RESULTS Following the 12-week intervention, the IF regimen significantly elevated serum apoM levels (p = 0.0144), whereas no increase was observed in the non-IF group (p = 0.9801). ApoM levels correlated with weight loss and fasting glucose levels in the IF group. Both groups exhibited a robust enhancement in HDL cholesterol efflux capacity (p < 0.0001, p = 0.0006) after 12 weeks. Notably, only the non-IF group exhibited significantly elevated activity of PON1 (p = 0.0455) and LCAT (p = 0.0117) following the 12-week intervention. In contrast, the changes observed in the IF group did not reach statistical significance. CONCLUSIONS A balanced diet combined with meticulous insulin management improves multiple metrics of HDL function. While additional IF increases apoM levels, it does not further enhance other aspects of HDL functionality. TRIAL REGISTRATION The study was registered at the German Clinical Trial Register (DRKS) on 3 September 2019 under the number DRKS00018070.
Collapse
Affiliation(s)
- Anja Pammer
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, 8010, Austria
| | - Anna Obermayer
- Interdisciplinary Metabolic Medicine Trials Unit, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, 8036, Austria
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, 8036, Austria
| | - Julia T Stadler
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, 8010, Austria
| | - Peter N Pferschy
- Interdisciplinary Metabolic Medicine Trials Unit, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, 8036, Austria
| | - Norbert J Tripolt
- Interdisciplinary Metabolic Medicine Trials Unit, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, 8036, Austria
| | - Hansjörg Habisch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Graz, 8010, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Graz, 8010, Austria
- BioTechMed Graz, Graz, 8010, Austria
| | - Harald Sourij
- Interdisciplinary Metabolic Medicine Trials Unit, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, 8036, Austria.
- BioTechMed Graz, Graz, 8010, Austria.
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, 8010, Austria.
- BioTechMed Graz, Graz, 8010, Austria.
| |
Collapse
|
37
|
Mamede I, Braga MAP, Martins OC, Franchini AEO, Silveira Filho RB, Santos MCF. Association between very high HDL-C levels and mortality: A systematic review and meta-analysis. J Clin Lipidol 2024; 18:e701-e709. [PMID: 39278774 DOI: 10.1016/j.jacl.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Recent research has raised questions about the assumed cardiovascular (CV) benefits of high-density lipoprotein cholesterol (HDL-C) and the potential for adverse outcomes with extremely high levels. OBJECTIVE We conducted a meta-analysis to investigate the association between very high HDL-C levels (≥80 mg/dL) and mortality outcomes in individuals without coronary artery disease (CAD). METHODS We systematically searched PubMed, Embase, and Cochrane databases for studies comparing very high HDL-C levels to normal levels (40-60 mg/dL) in CAD-free individuals. We assessed heterogeneity using I2 statistics with a random-effects model. RESULTS Our analysis included 1,004,584 individuals from 8 studies, of whom 133,646 (13.3%) had very high HDL-C levels. All-cause mortality did not significantly differ between groups (p = 0.55), nor did cancer mortality (p = 0.45). Cardiovascular mortality showed no change in those with very high HDL-C (hazard ratio [HR] 1.05; 95% confidence interval [CI] 0.94-1.17; p = 0.37). Fatal and non-fatal coronary heart disease events were less frequent in the very high HDL-C group (HR 0.79; 95% CI 0.73-0.86; p < 0.00001). Subgroup dose-response analysis revealed that very high HDL-C levels increased cardiovascular death in women above 116 mg/dL (HR 1.47; 95% CI 1.01-2.15) and in men above 94 mg/dL (HR 1.29; 95% CI 1.01-1.65) (p_nonlinearity <0.01). CONCLUSIONS These findings suggest that very high HDL-C levels are not protective against CV mortality and may, in fact, increase CV mortality risk especially in men.
Collapse
Affiliation(s)
- Isadora Mamede
- Faculty of Medicine, Federal University of Sao Joao del-Rei, Centro Oeste Campus, Divinopolis, Brazil (Dr Mamede).
| | | | - Otavio C Martins
- Faculty of Medicine, Federal University of Juiz de Fora, Juiz de Fora, Brazil (Dr Martins)
| | - Anne E O Franchini
- Faculty of Medicine, President Tancredo de Almeida Neves University Center, Sao Joao del Rei, Brazil (Dr Franchini)
| | | | - Marcel C F Santos
- Department of Internal Medicine, University of Sao Paulo, Sao Paulo, Brazil (Dr Santos)
| |
Collapse
|
38
|
Toh R. Genetic Determinants of High-density Lipoprotein Cholesterol Efflux Capacity: Insights from Paraoxonase 1 Polymorphisms. J Atheroscler Thromb 2024; 31:1260-1262. [PMID: 38910119 PMCID: PMC11374540 DOI: 10.5551/jat.ed267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024] Open
Affiliation(s)
- Ryuji Toh
- Division of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine
| |
Collapse
|
39
|
Zhu J, Wang Y, Li M, Huang D, Li S, Li J. Clinical incidence and relevance of incomplete endothelialization in atrial fibrillation patients with Left Atrial Appendage Closure. BMC Cardiovasc Disord 2024; 24:439. [PMID: 39179989 PMCID: PMC11342651 DOI: 10.1186/s12872-024-04113-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND The objective of this study is to investigate the incidence, potential risk factors, and clinical outcomes of incomplete device endothelialization (IDE) in atrial fibrillation (AF) patients undergoing Watchman left atrial appendage closure (LAAC). METHODS In this study, 68 AF patients who underwent successful implantation of the Watchman device without peri-device leak (PDL) during follow-up were included. The endothelialization status was assessed using Transesophageal echocardiography (TEE) and LAA computed tomography angiography (CTA) at 6 weeks and 6 months post-implantation. Adverse cerebro-cardiac events were documented at one-year follow-up. Baseline characteristics, including age, device sizes, and clinical indicators, were analyzed as potential predictors for IDE. RESULTS IDE was observed in 70.6% and 67.6% of patients at 6 weeks and 6 months after implantation, respectively. Higher levels of high-density lipoprotein cholesterol (HDL-C) [odds ratio (OR): 15.109, 95% confidence interval (CI): 1.637-139.478, p = 0.017 and OR: 11.015, 95% CI: 1.365-88.896, p = 0.024] and lower aspartate aminotransferase (AST) (OR 0.924, 95% CI: 0.865-0.986, p = 0.017 and OR: 0.930, 95% CI: 0.874-0.990, p = 0.023) at baseline were found to be significantly associated with IDE at 6 weeks and 6 months, respectively, although no significant difference in adverse cerebro-cardiac events was noted between incomplete and complete DE groups during 1-year follow-up CONCLUSIONS: IDE is found to be a prevalent occurrence in humans following LAAC. Elevated HDL-C and reduced AST levels are shown to be linked to an increased risk of IDE after LAAC.
Collapse
Affiliation(s)
- Jini Zhu
- Department of Cardiology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanpeng Wang
- Department of Cardiology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meifang Li
- Department of Emergency, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong Huang
- Department of Cardiology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuai Li
- Department of Cardiology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jingbo Li
- Department of Cardiology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
40
|
Chen Q, Abudukeremu A, Li K, Zheng M, Li H, Huang T, Huang C, Wen K, Wang Y, Zhang Y. High-Density Lipoprotein Subclasses and Their Role in the Prevention and Treatment of Cardiovascular Disease: A Narrative Review. Int J Mol Sci 2024; 25:7856. [PMID: 39063097 PMCID: PMC11277419 DOI: 10.3390/ijms25147856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The association between high-density lipoprotein cholesterol (HDL-C) and cardiovascular disease (CVD) is controversial. HDL-C is one content type of high-density lipoprotein (HDL). HDL consists of diverse proteins and lipids and can be classified into different subclasses based on size, shape, charge, and density, and can change dynamically in disease states. Therefore, HDL-C levels alone cannot represent HDLs' cardioprotective role. In this review, we summarized the methods for separating HDL subclasses, the studies on the association between HDL subclasses and cardiovascular risk (CVR), and the impact of lipid-modifying medications and nonpharmacological approaches (exercise training, dietary omega fatty acids, and low-density lipoprotein apheresis) on HDL subclasses. As HDL is a natural nanoplatform, recombinant HDLs (rHDLs) have been used as a delivery system in vivo by loading small interfering RNA, drugs, contrast agents, etc. Therefore, we further reviewed the HDL subclasses used in rHDLs and their advantages and disadvantages. This review would provide recommendations and guidance for future studies on HDL subclasses' cardioprotective roles.
Collapse
Affiliation(s)
- Qiaofei Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Ayiguli Abudukeremu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Kaiwen Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510120, China;
| | - Minglong Zheng
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Hongwei Li
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Tongsheng Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Canxia Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Kexin Wen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Yue Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
| | - Yuling Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; (Q.C.); (A.A.); (M.Z.); (H.L.); (T.H.); (C.H.); (K.W.); (Y.W.)
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou 510080, China
| |
Collapse
|
41
|
Costacou T, Vaisar T, Miller RG, Davidson WS, Heinecke JW, Orchard TJ, Bornfeldt KE. High-Density Lipoprotein Particle Concentration and Size Predict Incident Coronary Artery Disease Events in a Cohort With Type 1 Diabetes. J Am Heart Assoc 2024; 13:e034763. [PMID: 38958152 PMCID: PMC11292758 DOI: 10.1161/jaha.123.034763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/20/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND The cholesterol efflux capacity of high density lipoprotein (HDL) is negatively associated with cardiovascular risk. Small HDL particles account almost quantitatively for cholesterol efflux capacity, perhaps mediated through efflux of cholesterol and outer leaflet plasma membrane phospholipids by ABCA1 (ATP binding cassette subfamily A member 1). People with type 1 diabetes are at increased coronary artery disease (CAD) risk despite normal HDL-cholesterol concentrations. We therefore tested the hypothesis that small HDL particles (HDL-P)-rather than HDL-cholesterol-predict incident CAD in type 1 diabetes. METHODS AND RESULTS Incident CAD (CAD death, myocardial infarction, or coronary revascularization) was determined in 550 individuals with childhood-onset type 1 diabetes. HDL-P was quantified by calibrated ion mobility analysis and cholesterol efflux capacity was quantified with validated assays. During a median follow-up of 26 years, 36.5% of the participants developed incident CAD, for an incidence density of 181.3 per 10 000 person-years. In multivariable Cox models, neither HDL-cholesterol nor apolipoprotein A1 concentration was significantly associated with CAD risk. In contrast, higher extra-small HDL-P concentrations were significantly associated with decreased CAD risk (hazard ratio [HR], 0.26 [95% CI, 0.14-0.50]). Weaker associations were observed for total HDL-P (HR, 0.88 [95% CI, 0.83-0.93]), small HDL (HR, 0.83 [95% CI, 0.68-1.02]), medium HDL (HR, 0.79 [95% CI, 0.71-0.89]), and large HDL (HR, 0.72 [95% CI, 0.59-0.89]). Although cholesterol efflux capacity was negatively associated with incident CAD, this association was no longer significant after adjustment for total HDL-P. CONCLUSIONS Lower concentrations of total HDL-P and HDL subpopulations were positively associated with incident CAD independently of HDL-cholesterol, apolipoprotein A1, and other common CVD risk factors. Extra-small HDL was a much stronger predictor of risk than the other HDLs. Our data are consistent with the proposal that extra-small HDL plays a critical role in cardioprotection in type 1 diabetes, mediated by macrophage cholesterol efflux by the ABCA1 pathway.
Collapse
Affiliation(s)
- Tina Costacou
- Department of EpidemiologyUniversity of PittsburghPittsburghPA
| | - Tomas Vaisar
- Department of MedicineUniversity of WashingtonSeattleWA
| | | | - W. Sean Davidson
- Department of Pathology and Laboratory MedicineUniversity of Cincinnati College of MedicineCincinnatiOH
| | | | | | | |
Collapse
|
42
|
Nasr A, Brooks MM, Barinas-Mitchell E, Orchard T, Billheimer J, Wang NC, McConnell D, Rader DJ, El Khoudary SR. High-density lipoprotein metrics during midlife and future subclinical atherosclerosis in women: the SWAN HDL study. Menopause 2024; 31:567-574. [PMID: 38743910 PMCID: PMC11213666 DOI: 10.1097/gme.0000000000002371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
OBJECTIVE The clinical utility of high-density lipoprotein cholesterol (HDL-C) in risk classification is limited, especially in midlife women. Novel metrics of HDL may better reflect this risk. We clustered a comprehensive profile of HDL metrics into favorable and unfavorable clusters and assessed how these two clusters are related to future subclinical atherosclerosis (carotid intima media thickness [cIMT], interadventitial diameter [IAD], and carotid plaque presence) in midlife women. METHODS Four hundred sixty-one women (baseline age: 50.4 [2.7] years; 272 White, 137 Black, 52 Chinese) from the Study of Women's Health Across the Nation HDL ancillary study who had baseline measures of HDL cholesterol efflux capacity (HDL-CEC), lipid contents (HDL-phospholipids [HDL-PL] and HDL triglycerides [HDL-Tg]), and HDL particle (HDL-P) distribution and size, followed by carotid ultrasound (average 12.9 [SD: 2.6] years later), were included. Using latent cluster analysis, women were clustered into a favorable (high HDL-CEC, HDL-PL, large and medium HDL-P, less HDL-Tg and small HDL-P, larger size) or an unfavorable HDL cluster (low HDL-CEC, HDL-PL, large and medium HDL-P, more HDL-Tg, and small HDL-P, smaller size) and then linked to future subclinical atherosclerosis using linear or logistic regression. RESULTS The favorable HDL cluster was associated with lower cIMT, IAD, and odds of carotid plaque presence. These associations were attenuated by body mass index, except in Chinese women where the association with cIMT persisted (0.72 [0.63, 0.83]). CONCLUSIONS The association between favorable HDL clusters and a better postmenopausal subclinical atherosclerosis profile is largely explained by body mass index; however, racial/ethnic differences may exist.
Collapse
Affiliation(s)
- Alexis Nasr
- University of Pittsburgh School of Public Health, Public Health Building, 130 De Soto St, Pittsburgh, PA 15261, USA
| | - Maria M. Brooks
- University of Pittsburgh School of Public Health, Public Health Building, 130 De Soto St, Pittsburgh, PA 15261, USA
| | - Emma Barinas-Mitchell
- Department of Epidemiology, University of Pittsburgh, 130 N. Bellefield Avenue, Suite 338, Pittsburgh PA 15213
| | - Trevor Orchard
- University of Pittsburgh School of Public Health, Public Health Building, 130 De Soto St, Pittsburgh, PA 15261, USA
| | - Jeffrey Billheimer
- University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Norman C. Wang
- Department of Medicine, University of Pittsburgh School of Medicine, UPMC Presbyterian, 200 Lothrop Street, South Tower, 3 Floor, Room 352.9, Pittsburgh PA, USA
| | - Daniel McConnell
- University of Michigan Department of Epidemiology, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| | - Daniel J. Rader
- University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Samar R. El Khoudary
- University of Pittsburgh School of Public Health, Public Health Building, 130 De Soto St, Pittsburgh, PA 15261, USA
| |
Collapse
|
43
|
Seto Y, Nagao M, Iino T, Harada A, Murakami K, Miwa K, Shinohara M, Nishimori M, Yoshikawa S, Asakura J, Fujioka T, Ishida T, Hirata KI, Toh R. Impaired Cholesterol Uptake Capacity in Patients with Hypertriglyceridemia and Diabetes Mellitus. J Appl Lab Med 2024; 9:728-740. [PMID: 38574000 DOI: 10.1093/jalm/jfae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/06/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Although low high-density lipoprotein cholesterol (HDL-C) levels are a common metabolic abnormality associated with insulin resistance, their role in cardiovascular risk stratification remains controversial. Recently, we developed a simple, high-throughput, cell-free assay system to evaluate the "cholesterol uptake capacity (CUC)" as a novel concept for HDL functionality. In this study, we assessed the CUC in patients with hypertriglyceridemia and diabetes mellitus. METHODS The CUC was measured using cryopreserved serum samples from 285 patients who underwent coronary angiography or percutaneous coronary intervention between December 2014 and May 2019 at Kobe University Hospital. RESULTS The CUC was significantly lower in diabetic patients (n = 125) than in nondiabetic patients (93.0 vs 100.7 arbitrary units (A.U.), P = 0.002). Patients with serum triglyceride (TG) levels >150 mg/dL (n = 94) also had a significantly lower CUC (91.8 vs 100.0 A.U., P = 0.004). Furthermore, the CUC showed a significant inverse correlation with TG, hemoglobin A1c (Hb A1c), homeostasis model assessment of insulin resistance (HOMA-IR), and body mass index (BMI). Finally, the HDL-C/Apolipoprotein A1 (ApoA1) ratio, calculated as a surrogate index of HDL particle size, was significantly positively correlated with the CUC (r2 = 0.49, P < 0.001), but inversely correlated with TG levels (r2 = -0.30, P < 0.001). CONCLUSIONS The CUC decreased in patients with hypertriglyceridemia and diabetes mellitus, and HDL particle size was a factor defining the CUC and inversely correlated with TG levels, suggesting that impaired CUC in insulin-resistant states was partially due to the shift in HDL towards smaller particles. These findings provide a better understanding of the mechanisms underlying impaired HDL functionality.
Collapse
Affiliation(s)
- Yutaro Seto
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Manabu Nagao
- Division of Evidence-Based Laboratory Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Takuya Iino
- Central Research Laboratories, Sysmex Corporation, Nishi-ku, Kobe, Japan
| | - Amane Harada
- Central Research Laboratories, Sysmex Corporation, Nishi-ku, Kobe, Japan
| | - Katsuhiro Murakami
- Central Research Laboratories, Sysmex Corporation, Nishi-ku, Kobe, Japan
| | - Keiko Miwa
- Central Research Laboratories, Sysmex Corporation, Nishi-ku, Kobe, Japan
| | - Masakazu Shinohara
- Division of Molecular Epidemiology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Makoto Nishimori
- Division of Molecular Epidemiology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Sachiko Yoshikawa
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Junko Asakura
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Tomoo Fujioka
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Tatsuro Ishida
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
- Division of Nursing Practice, Kobe University Graduate School of Health Sciences, Suma-ku, Kobe, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
- Division of Evidence-Based Laboratory Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Ryuji Toh
- Division of Evidence-Based Laboratory Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| |
Collapse
|
44
|
Zhang X, van der Vorst EPC. High-Density Lipoprotein Modifications: Causes and Functional Consequences in Type 2 Diabetes Mellitus. Cells 2024; 13:1113. [PMID: 38994965 PMCID: PMC11240616 DOI: 10.3390/cells13131113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024] Open
Abstract
High-density lipoprotein (HDL) is a group of small, dense, and protein-rich lipoproteins that play a role in cholesterol metabolism and various cellular processes. Decreased levels of HDL and HDL dysfunction are commonly observed in individuals with type 2 diabetes mellitus (T2DM), which is also associated with an increased risk for cardiovascular disease (CVD). Due to hyperglycemia, oxidative stress, and inflammation that develop in T2DM, HDL undergoes several post-translational modifications such as glycation, oxidation, and carbamylation, as well as other alterations in its lipid and protein composition. It is increasingly recognized that the generation of HDL modifications in T2DM seems to be the main cause of HDL dysfunction and may in turn influence the development and progression of T2DM and its related cardiovascular complications. This review provides a general introduction to HDL structure and function and summarizes the main modifications of HDL that occur in T2DM. Furthermore, the potential impact of HDL modifications on the pathogenesis of T2DM and CVD, based on the altered interactions between modified HDL and various cell types that are involved in glucose homeostasis and atherosclerotic plaque generation, will be discussed. In addition, some perspectives for future research regarding the T2DM-related HDL modifications are addressed.
Collapse
Affiliation(s)
- Xiaodi Zhang
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), 80336 Munich, Germany
| |
Collapse
|
45
|
Endo Y, Teramoto M, Arakawa J, Ukita S, Toshima G, Suenaga Y, Sasaki K, Ayaori M, Nakayama H, Inoue Y, Ikewaki K. Obstructive sleep apnea syndrome attenuated high-density lipoprotein function. J Clin Lipidol 2024:S1933-2874(24)00199-5. [PMID: 39294021 DOI: 10.1016/j.jacl.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is associated with an increased risk of cardiovascular disease. High-density lipoproteins (HDLs) exert anti-atherogenic effects, even on cholesterol efflux capacity (CEC). The HDL proteome is reportedly altered in patients with coronary artery disease. OBJECTIVE We hypothesized that OSA attenuates HDL function through an altered HDL proteome, which could be alleviated by continuous positive airway pressure (CPAP) therapy. METHODS Patients aged ≥20 years (n = 115) with suspected OSA were enrolled in this cross-sectional study, with 34 patients diagnosed with moderate and severe OSA included in the interventional study and treated with CPAP therapy for 12 weeks. To further investigate the HDL proteome in OSA, we conducted a discovery study by analyzing HDL proteomes in 10 patients. RESULTS In this study, CEC was significantly lower in the sleep apnea syndrome (SAS) group (apnea-hypopnea index [AHI] ≥5) than in the non-SAS group (AHI <5; 0.96 ± 0.14 vs. 1.06 ± 0.15, p = 0.01). Multiple regression analysis revealed that minimal pulse oxygen saturation (MinSpO2) was positively correlated with CEC. In the interventional study, a 12-week CPAP therapy did not affect CEC. We identified orosomucoid 1 (ORM1), an acute-phase inflammatory molecule, as a candidate protein for OSA-induced HDL dysfunction. Further validation study revealed that serum ORM1 levels were inversely associated with CEC, independent of HDL-cholesterol and high-sensitivity C-reactive protein. CONCLUSIONS HDL function was impaired in patients with OSA and a reduced CEC. However, CPAP therapy did not affect CEC. An altered HDL proteome, particularly with increased ORM1 levels, may be associated with impaired HDL function. TRIAL REGISTRATION This clinical study was registered with the University Hospital Medical Information Network Clinical Trials Registry (UMIN000025335 and UMIN000025341).
Collapse
Affiliation(s)
- Yasuhiro Endo
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan (Drs Endo, Teramoto, Arakawa, Suenaga, Sasaki, and Ikewaki); Division of Environmental Medicine, National Defense Medical College Research Institute, 3-2 Namiki, Tokorozawa, Saitama, Japan (Dr Endo).
| | - Manami Teramoto
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan (Drs Endo, Teramoto, Arakawa, Suenaga, Sasaki, and Ikewaki)
| | - Junko Arakawa
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan (Drs Endo, Teramoto, Arakawa, Suenaga, Sasaki, and Ikewaki)
| | - Shoko Ukita
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan (Drs Ukita and Toshima)
| | - Genta Toshima
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan (Drs Ukita and Toshima)
| | - Yumiko Suenaga
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan (Drs Endo, Teramoto, Arakawa, Suenaga, Sasaki, and Ikewaki)
| | - Kei Sasaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan (Drs Endo, Teramoto, Arakawa, Suenaga, Sasaki, and Ikewaki)
| | | | - Hideaki Nakayama
- Department of Somnology, Tokyo Medical University, Tokyo, Japan (Dr Nakayama)
| | - Yuichi Inoue
- Japan Somnology Center, Institute of Neuropsychiatry, Tokyo, Japan (Dr Inoue)
| | - Katsunori Ikewaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan (Drs Endo, Teramoto, Arakawa, Suenaga, Sasaki, and Ikewaki)
| |
Collapse
|
46
|
Jiang Y, Pang S, Liu X, Wang L, Liu Y. The Gut Microbiome Affects Atherosclerosis by Regulating Reverse Cholesterol Transport. J Cardiovasc Transl Res 2024; 17:624-637. [PMID: 38231373 DOI: 10.1007/s12265-024-10480-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/07/2024] [Indexed: 01/18/2024]
Abstract
The human system's secret organ, the gut microbiome, has received considerable attention. Emerging research has yielded substantial scientific evidence indicating that changes in gut microbial composition and microbial metabolites may contribute to the development of atherosclerotic cardiovascular disease. The burden of cardiovascular disease on healthcare systems is exacerbated by atherosclerotic cardiovascular disease, which continues to be the leading cause of mortality globally. Reverse cholesterol transport is a powerful protective mechanism that effectively prevents excessive accumulation of cholesterol for atherosclerotic cardiovascular disease. It has been revealed how the gut microbiota modulates reverse cholesterol transport in patients with atherosclerotic risk. In this review, we highlight the complex interactions between microbes, their metabolites, and their potential impacts in reverse cholesterol transport. We also explore the feasibility of modulating gut microbes and metabolites to facilitate reverse cholesterol transport as a novel therapy for atherosclerosis.
Collapse
Affiliation(s)
- Yangyang Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuchao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Xiaoyu Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lixin Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| |
Collapse
|
47
|
Zheng J, Hong BV, Agus JK, Tang X, Klebaner NR, Chen S, Guo F, Harvey DJ, Lebrilla CB, Zivkovic AM. Lutein and Zeaxanthin Enhance, Whereas Oxidation, Fructosylation, and Low pH Damage High-Density Lipoprotein Biological Functionality. Antioxidants (Basel) 2024; 13:616. [PMID: 38790721 PMCID: PMC11118252 DOI: 10.3390/antiox13050616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
High-density lipoproteins (HDLs) are key regulators of cellular cholesterol homeostasis but are functionally altered in many chronic diseases. The factors that cause HDL functional loss in chronic disease are not fully understood. It is also unknown what roles antioxidant carotenoids play in protecting HDL against functional loss. The aim of this study was to measure how various disease-associated chemical factors including exposure to (1) Cu2+ ions, (2) hypochlorous acid (HOCL), (3) hydrogen peroxide (H2O2), (4) sialidase, (5) glycosidase, (6) high glucose, (7) high fructose, and (8) acidic pH, and the carotenoid antioxidants (9) lutein and (10) zeaxanthin affect HDL functionality. We hypothesized that some of the modifications would have stronger impacts on HDL particle structure and function than others and that lutein and zeaxanthin would improve HDL function. HDL samples were isolated from generally healthy human plasma and incubated with the corresponding treatments listed above. Cholesterol efflux capacity (CEC), lecithin-cholesterol acyl transferase (LCAT) activity, and paraoxonase-1 (PON1) activity were measured in order to determine changes in HDL functionality. Median HDL particle diameter was increased by acidic pH treatment and reduced by HOCl, high glucose, high fructose, N-glycosidase, and lutein treatments. Acidic pH, oxidation, and fructosylation all reduced HDL CEC, whereas lutein, zeaxanthin, and sialidase treatment improved HDL CEC. LCAT activity was reduced by acidic pH, oxidation, high fructose treatments, and lutein. PON1 activity was reduced by sialidase, glycosidase, H2O2, and fructose and improved by zeaxanthin and lutein treatment. These results show that exposure to oxidizing agents, high fructose, and low pH directly impairs HDL functionality related to cholesterol efflux and particle maturation, whereas deglycosylation impairs HDL antioxidant capacity. On the other hand, the antioxidants lutein and zeaxanthin improve or preserve both HDL cholesterol efflux and antioxidant activity but have no effect on particle maturation.
Collapse
Affiliation(s)
- Jingyuan Zheng
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA; (J.Z.); (B.V.H.); (J.K.A.); (X.T.); (N.R.K.)
| | - Brian V. Hong
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA; (J.Z.); (B.V.H.); (J.K.A.); (X.T.); (N.R.K.)
| | - Joanne K. Agus
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA; (J.Z.); (B.V.H.); (J.K.A.); (X.T.); (N.R.K.)
| | - Xinyu Tang
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA; (J.Z.); (B.V.H.); (J.K.A.); (X.T.); (N.R.K.)
| | - Nola R. Klebaner
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA; (J.Z.); (B.V.H.); (J.K.A.); (X.T.); (N.R.K.)
| | - Siyu Chen
- Department of Chemistry, University of California Davis, Davis, CA 95616, USA; (S.C.); (C.B.L.)
| | - Fei Guo
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, USA;
| | - Danielle J. Harvey
- Department of Public Health Sciences, University of California Davis, Davis, CA 95616, USA;
| | - Carlito B. Lebrilla
- Department of Chemistry, University of California Davis, Davis, CA 95616, USA; (S.C.); (C.B.L.)
| | - Angela M. Zivkovic
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA; (J.Z.); (B.V.H.); (J.K.A.); (X.T.); (N.R.K.)
| |
Collapse
|
48
|
Sergi D, Sanz JM, Trentini A, Bonaccorsi G, Angelini S, Castaldo F, Morrone S, Spaggiari R, Cervellati C, Passaro A. HDL-Cholesterol Subfraction Dimensional Distribution Is Associated with Cardiovascular Disease Risk and Is Predicted by Visceral Adiposity and Dietary Lipid Intake in Women. Nutrients 2024; 16:1525. [PMID: 38794763 PMCID: PMC11124017 DOI: 10.3390/nu16101525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
HDL-cholesterol quality, including cholesterol distribution in HDL subfractions, is emerging as a key discriminant in dictating the effects of these lipoproteins on cardiovascular health. This study aims at elucidating the relationship between cholesterol distribution in HDL subfractions and CVD risk factors as well as diet quality and energy density in a population of pre- and postmenopausal women. Seventy-two women aged 52 ± 6 years were characterized metabolically and anthropometrically. Serum HDL-C subfractions were quantified using the Lipoprint HDL System. Cholesterol distribution in large HDL subfractions was lower in overweight individuals and study participants with moderate to high estimated CVD risk, hypertension, or insulin resistance. Cholesterol distribution in large, as opposed to small, HDL subfractions correlated negatively with insulin resistance, circulating triglycerides, and visceral adipose tissue (VAT). VAT was an independent positive and negative predictor of cholesterol distribution in large and small HDL subfractions, respectively. Furthermore, an increase in energy intake could predict a decrease in cholesterol levels in large HDL subfractions while lipid intake positively predicted cholesterol levels in small HDL subfractions. Cholesterol distribution in HDL subfractions may represent an additional player in shaping CVD risk and a novel potential mediator of the effect of diet on cardiovascular health.
Collapse
Affiliation(s)
- Domenico Sergi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (D.S.); (S.A.); (F.C.); (S.M.); (R.S.); (C.C.); (A.P.)
| | - Juana Maria Sanz
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandro Trentini
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Gloria Bonaccorsi
- Department of Translational Medicine, Menopause and Osteoporosis Center, University Center for Studies on Gender Medicine, 44121 Ferrara, Italy;
| | - Sharon Angelini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (D.S.); (S.A.); (F.C.); (S.M.); (R.S.); (C.C.); (A.P.)
| | - Fabiola Castaldo
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (D.S.); (S.A.); (F.C.); (S.M.); (R.S.); (C.C.); (A.P.)
| | - Sara Morrone
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (D.S.); (S.A.); (F.C.); (S.M.); (R.S.); (C.C.); (A.P.)
| | - Riccardo Spaggiari
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (D.S.); (S.A.); (F.C.); (S.M.); (R.S.); (C.C.); (A.P.)
| | - Carlo Cervellati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (D.S.); (S.A.); (F.C.); (S.M.); (R.S.); (C.C.); (A.P.)
| | - Angelina Passaro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (D.S.); (S.A.); (F.C.); (S.M.); (R.S.); (C.C.); (A.P.)
| | | |
Collapse
|
49
|
Machado VA, Santisteban ARN, Martins CM, Damasceno NRT, Fonseca FA, Neto AMF, Izar MC. Effects of phytosterol supplementation on lipoprotein subfractions and LDL particle quality. Sci Rep 2024; 14:11108. [PMID: 38750162 PMCID: PMC11096344 DOI: 10.1038/s41598-024-61897-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 05/10/2024] [Indexed: 05/18/2024] Open
Abstract
Phytosterols are natural components of plant-based foods used as supplements because of their known cholesterol-lowering effect. However, their effects on lipoprotein subfractions and the quality of the LDL particle have not been studied in greater detail. We aimed to evaluate the effects of phytosterols supplements on lipids, lipoproteins subfractions, and on the quality of LDL. A prospective, pilot-type, open label, cross-over study, randomized 23 males in primary prevention of hypercholesterolemia to receive diet or diet plus phytosterol (2.6 g in 2 doses, with meals) for 12 weeks, when treatments were switched for another 12 weeks. Lipoprotein subfractions were analyzed by electrophoresis in polyacrylamide gel (Lipoprint System®). The Sampson equation estimated the small and dense (sd) and large and buoyant (lb) LDL subfractions from the lipid profile. Quality of LDL particle was analyzed by Z-scan and UV-vis spectroscopy. Primary outcome was the comparison of diet vs. diet plus phytosterols. Secondary outcomes assessed differences between baseline, diet and diet plus phytosterol. Non-parametric statistics were performed with p < 0.05. There was a trend to reduction on HDL-7 (p = 0.05) in diet plus phytosterol arm, with no effects on the quality of LDL particles. Heatmap showed strong correlations (ρ > 0.7) between particle size by different methods with both interventions. Diet plus phytosterol reduced TC, increased HDL-c, and reduced IDL-B, whereas diet increased HDL7, and reduced IDL-B vs. baseline (p < 0.05, for all). Phytosterol supplementation demonstrated small beneficial effects on HDL-7 subfraction, compared with diet alone, without effects on the quality of LDL particles.This trial is registered in Clinical Trials (NCT06127732) and can be accessed at https://clinicaltrials.gov .
Collapse
Affiliation(s)
- Valeria Arruda Machado
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, 340 - Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Celma Muniz Martins
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, 340 - Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Francisco A Fonseca
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, 340 - Sao Paulo, Sao Paulo, SP, Brazil
- Institute of Physics, National Institute of Complex Fluids, University of São Paulo, São Paulo, SP, Brazil
| | - Antonio M Figueiredo Neto
- Institute of Physics, National Institute of Complex Fluids, University of São Paulo, São Paulo, SP, Brazil
| | - Maria Cristina Izar
- Cardiology Division, Department of Medicine, Federal University of Sao Paulo, 340 - Sao Paulo, Sao Paulo, SP, Brazil.
- Institute of Physics, National Institute of Complex Fluids, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
50
|
Brewer HB, Schaefer EJ, Foldyna B, Ghoshhajra BB. High-density lipoprotein infusion therapy: A review. J Clin Lipidol 2024; 18:e374-e383. [PMID: 38782655 DOI: 10.1016/j.jacl.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 05/25/2024]
Abstract
Increased cholesterol-rich, low-density, non-calcified atheromas as assessed by computer coronary tomography angiography analyses have been shown to predict myocardial infarction significantly better than coronary artery calcium score or the presence of obstructive coronary artery disease (CAD) as evaluated with standard coronary angiography. Low serum high-density lipoprotein (HDL) cholesterol values are an independent risk factor for CAD. Very small, lipid-poor preβ-1 HDL particles have been shown to be most effective in promoting cellular cholesterol efflux. HDL infusions have been documented to reduce aortic atherosclerosis in cholesterol-fed animal models. However, human studies using infusions of either the HDL mimetic containing recombinant apolipoprotein (apo) A-I Milano or Cerenis Compound-001 with native recombinant apoA-I have been mainly negative in promoting coronary atherosclerosis progression as assessed by intravascular ultrasound. In contrast, a study using 7 weekly infusions of autologous delipidated HDL in six homozygous familial hypercholesterolemic patients was effective in promoting significant regression of low-density non-calcified coronary atheroma regression as assessed by computed coronary angiography. This therapy has received Food and Drug Administration approval. Commonwealth Serum Laboratories has carried out a large clinical endpoint trial using an HDL complex (native apoA-I with phospholipid), and the results were negative. Our purpose is to review animal and human studies using various forms of HDL infusion therapy to promote regression of atherosclerosis. In our view, differences in results may be due to: 1) the HDL preparations used, 2) the subjects studied, and 3) the methods used to assess coronary atherosclerosis.
Collapse
Affiliation(s)
| | - Ernst J Schaefer
- Boston Heart Diagnostics, Framingham, MA, USA (Dr Schaefer); Department of Medicine, Tufts University School of Medicine, Boston, MA, USA (Dr Schaefer).
| | - Borek Foldyna
- Division of Cardiovascular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA (Drs Foldyna and Ghoshhajra)
| | - Brian B Ghoshhajra
- Division of Cardiovascular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA (Drs Foldyna and Ghoshhajra)
| |
Collapse
|