51
|
Yamamoto T, Wada F, Harada-Shiba M. Development of Antisense Drugs for Dyslipidemia. J Atheroscler Thromb 2016; 23:1011-25. [PMID: 27466159 PMCID: PMC5090806 DOI: 10.5551/jat.rv16001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Abnormal elevation of low-density lipoprotein (LDL) and triglyceride-rich lipoproteins in plasma as well as dysfunction of anti-atherogenic high-density lipoprotein (HDL) have both been recognized as essential components of the pathogenesis of atherosclerosis and are classified as dyslipidemia. This review describes the arc of development of antisense oligonucleotides for the treatment of dyslipidemia. Chemically-armed antisense candidates can act on various kinds of transcripts, including mRNA and miRNA, via several different endogenous antisense mechanisms, and have exhibited potent systemic anti-dyslipidemic effects. Here, we present specific cutting-edge technologies have recently been brought into antisense strategies, and describe how they have improved the potency of antisense drugs in regard to pharmacokinetics and pharmacodynamics. In addition, we discuss perspectives for the use of armed antisense oligonucleotides as new clinical options for dyslipidemia, in the light of outcomes of recent clinical trials and safety concerns indicated by several clinical and preclinical studies.
Collapse
|
52
|
Hegele RA. Multidimensional regulation of lipoprotein lipase: impact on biochemical and cardiovascular phenotypes. J Lipid Res 2016; 57:1601-7. [PMID: 27412676 DOI: 10.1194/jlr.c070946] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
53
|
Drenos F, Davey Smith G, Ala-Korpela M, Kettunen J, Würtz P, Soininen P, Kangas AJ, Dale C, Lawlor DA, Gaunt TR, Casas JP, Timpson NJ. Metabolic Characterization of a Rare Genetic Variation Within APOC3 and Its Lipoprotein Lipase-Independent Effects. CIRCULATION. CARDIOVASCULAR GENETICS 2016; 9:231-9. [PMID: 27114411 PMCID: PMC4920206 DOI: 10.1161/circgenetics.115.001302] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 04/21/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Plasma triglyceride levels have been implicated in atherosclerosis and coronary heart disease. Apolipoprotein C-III (APOC3) plays a key role in the hydrolysis of triglyceride-rich lipoproteins to remnant particles by lipoprotein lipase (LPL) and their uptake by the liver. A rare variant in APOC3(rs138326449) has been associated with triglyceride, very low-density lipoprotein, and high-density lipoprotein levels, as well as risk of coronary heart disease. We aimed to characterize the impact of this locus across a broad set of mainly lipids-focused metabolic measures. METHODS AND RESULTS A high-throughput serum nuclear magnetic resonance metabolomics platform was used to quantify 225 metabolic measures in 13 285 participants from 2 European population cohorts. We analyzed the effect of the APOC3 variant on the metabolic measures and used the common LPL(rs12678919) polymorphism to test for LPL-independent effects. Eighty-one metabolic measures showed evidence of association with APOC3(rs138326449). In addition to previously reported triglyceride and high-density lipoprotein associations, the variant was also associated with very low-density lipoprotein and high-density lipoprotein composition measures, other cholesterol measures, and fatty acids. Comparison of the APOC3 and LPL associations revealed that APOC3 association results for medium and very large very low-density lipoprotein composition are unlikely to be solely predictable by the action of APOC3 through LPL. CONCLUSIONS We characterized the effects of the rare APOC3(rs138326449) loss of function mutation in lipoprotein metabolism, as well as the effects of LPL(rs12678919). Our results improve our understanding of the role of APOC3 in triglyceride metabolism, its LPL independent action, and the complex and correlated nature of human metabolites.
Collapse
Affiliation(s)
- Fotios Drenos
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.).
| | - George Davey Smith
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Mika Ala-Korpela
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Johannes Kettunen
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Peter Würtz
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Pasi Soininen
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Antti J Kangas
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Caroline Dale
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Debbie A Lawlor
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Tom R Gaunt
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Juan-Pablo Casas
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.)
| | - Nicholas J Timpson
- From the MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol (F.D., G.D.S., M.A.-K., D.A.L., T.R.G., N.J.T.); Institute of Cardiovascular Science, University College London, London, United Kingdom (F.D., C.D., J.-P.C.); Computational Medicine, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu (M.A.-K., J.K., P.W., P.S., A.J.K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio (M.A.-K., J.K., P.S.); Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland (J.K.); and Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom (C.D., J.-P.C.).
| |
Collapse
|
54
|
Abstract
Hypertriglyceridemic pancreatitis (HTGP) is often encountered clinically as a common form of recurrent acute pancreatitis (AP). It is important to evaluate the management of severe hypertriglyceridemia (HTG) or anti-inflammation in the prophylaxis of HTGP in the clinic. FTY720 (2-amino-2[2-(4-octylphenyl) ethyl]-1, 3-propanediol) is a new anti-inflammatory agent with low toxicity and reported to ameliorate lung injury with pancreatitis in rat. We evaluated its protective affection on AP induced by seven hourly intraperitoneal injection of cerulein in apolipoprotein CIII transgenic mice with severe HTG. FTY720 at 1.5 mg/kg was administered by gastric lavage daily for 3 days before induction of AP. The effects of FTY720 to protect against HTGP were assessed by serum amylase, pancreatic pathological scores, immunostaining, and the expression of inflammatory cytokine genes. As a result, injection of cerulein resulted in more severe pathological changes of AP and higher monocyte chemoattractant protein 1 expression in the pancreas in transgenic than in nontransgenic mice. FTY720 pretreatment improved the pathological severity of AP and decreased the expression of monocyte chemoattractant protein 1 in the pancreas significantly, especially near fourfold reduction in transgenic mice. However, FTY720 did not affect plasma triglyceride levels, and other inflammatory factors and plasma amylase were not correlated with the extent of pancreatic damage in AP with or without FTY720 administration. In summary, our study in a new model, apolipoprotein CIII transgenic mice, demonstrated that HTG mice are susceptible to induction of AP. Prophylactic treatment of FTY720 can significantly attenuate cerulein-induced AP and hence warrant further investigation of sphingosine-1-phosphate receptors agonist for potential clinical application in recurrent attacks of HTGP.
Collapse
|
55
|
Shi J, Yang H, Duan X, Li L, Sun L, Li Q, Zhang J. Apolipoproteins as Differentiating and Predictive Markers for Assessing Clinical Outcomes in Patients with Small Cell Lung Cancer. Yonsei Med J 2016; 57:549-56. [PMID: 26996551 PMCID: PMC4800341 DOI: 10.3349/ymj.2016.57.3.549] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/11/2015] [Accepted: 04/09/2015] [Indexed: 02/06/2023] Open
Abstract
PURPOSE The present study aimed to investigate the value of apolipoproteins, including ApoA-1, ApoC-III, and ApoE, in patients with small cell lung cancer (SCLC) as potential biomarkers for diagnosis, prognosis, and cancer progression. MATERIALS AND METHODS Lung samples were collected from 89 patients with SCLC. Nineteen lung samples from non-small cell lung cancer (NSCLC) patients and 12 normal lung tissues were used as controls. Expression profiles of ApoA-1, ApoC-III, and ApoE in different samples were examined using immunohistochemical methods, and the expression levels were correlated with cancer types, treatment, and outcomes using chi-square and Mann-Whitney tests. RESULTS Expression of ApoA-1 and ApoC-III in SCLC was significantly different, compared with that in NSCLC and normal lung tissues, and was correlated with recurrence of SCLC. Patients undergoing neoadjuvant chemotherapy before surgery showed significantly reduced expression of ApoA-1 and increased expression of ApoC-III and ApoE. Nevertheless, the expression levels of ApoA-1, ApoC-III, and ApoE were not correlated with SCLC staging. CONCLUSION ApoA-1 and ApoC-III may be used as differentiating and predictive markers for SCLC. ApoA-1, ApoC-III, and ApoE may be used to monitor the efficacy of chemotherapy.
Collapse
Affiliation(s)
- Jian Shi
- Department of Medical Oncology, Forth Hospital of Hebei Medical University, Tumor Hospital of Hebei Province, Shijiazhuang, Hebei, China.
| | - Huichai Yang
- Department of Pathology, Forth Hospital of Hebei Medical University, Tumor Hospital of Hebei Province, Shijiazhuang, Hebei, China
| | - Xiaoyang Duan
- Department of Medical Oncology, Forth Hospital of Hebei Medical University, Tumor Hospital of Hebei Province, Shijiazhuang, Hebei, China
| | - Lihua Li
- Department of Medical Oncology, Forth Hospital of Hebei Medical University, Tumor Hospital of Hebei Province, Shijiazhuang, Hebei, China
| | - Lulu Sun
- Department of Medical Oncology, Forth Hospital of Hebei Medical University, Tumor Hospital of Hebei Province, Shijiazhuang, Hebei, China
| | - Qian Li
- Department of Medical Oncology, Forth Hospital of Hebei Medical University, Tumor Hospital of Hebei Province, Shijiazhuang, Hebei, China
| | - Junjun Zhang
- Department of Medical Oncology, Forth Hospital of Hebei Medical University, Tumor Hospital of Hebei Province, Shijiazhuang, Hebei, China
| |
Collapse
|
56
|
Kassai A, Muniyappa R, Levenson AE, Walter MF, Abel BS, Ring M, Taylor SI, Biddinger SB, Skarulis MC, Gorden P, Brown RJ. Effect of Leptin Administration on Circulating Apolipoprotein CIII levels in Patients With Lipodystrophy. J Clin Endocrinol Metab 2016; 101:1790-7. [PMID: 26900642 PMCID: PMC4880162 DOI: 10.1210/jc.2015-3891] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CONTEXT Apolipoprotein CIII (apoCIII), an inhibitor of lipoprotein lipase, plays an important role in triglyceride metabolism. However, the role of apoCIII in hypertriglyceridemia in lipodystrophy and the effects of leptin replacement on apoCIII levels are unknown. OBJECTIVE The objective of the study was to test the hypotheses that apoCIII is elevated in hypertriglyceridemic patients with lipodystrophy and that leptin replacement in these patients lowers circulating apoCIII. DESIGN, SETTING, STUDY PARTICIPANTS, INTERVENTION, AND OUTCOME MEASURES Using a post hoc cross-sectional case-control design, we compared serum apoCIII levels from patients with lipodystrophy not associated with HIV (n = 60) and age-, gender-, race-, and ethnicity-matched controls (n = 54) participating in ongoing studies at the National Institutes of Health. In a prospective, open-label, ongoing study, we studied the effects of 6–12 months of leptin replacement on apoCIII in lipodystrophy patients as an exploratory outcome. RESULTS ApoCIII was higher in lipodystrophy patients (geometric mean [25th and 75th percentiles]) (23.9 mg/dL [14.6, 40.3]) compared with controls (14.9 mg/dL [12.3, 17.7]) (P < .0001). ApoCIII and triglyceride levels were positively correlated in patients with lipodystrophy (R = 0.72, P < .0001) and healthy controls (R = 0.6, P < .0001). Leptin replacement (6–12 mo) did not significantly alter apoCIII (before leptin: 23.4 mg/dL [14.5, 40.1]; after leptin: 21.4 mg/dL [16.7, 28.3]; P = .34). CONCLUSIONS Leptin replacement in lipodystrophy did not alter serum apoCIII levels. Elevated apoCIII may play a role in the hypertriglyceridemia of lipodystrophy independent of leptin deficiency and replacement.
Collapse
Affiliation(s)
- Andrea Kassai
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Ranganath Muniyappa
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Amy E Levenson
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Mary F Walter
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Brent S Abel
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Michael Ring
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Simeon I Taylor
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Sudha B Biddinger
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Monica C Skarulis
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Phillip Gorden
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Rebecca J Brown
- Diabetes, Endocrinology and Obesity Branch (A.K., R.M., B.S.A., M.R., M.C.S., P.G., R.J.B.), Clinical Core Laboratory (M.F.W.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Division of Endocrinology (A.E.L., S.B.B.), Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115; and Division of Endocrinology, Diabetes, and Nutrition (S.I.T.), University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
57
|
Imbalanced insulin action in chronic over nutrition: Clinical harm, molecular mechanisms, and a way forward. Atherosclerosis 2016; 247:225-82. [PMID: 26967715 DOI: 10.1016/j.atherosclerosis.2016.02.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/31/2015] [Accepted: 02/02/2016] [Indexed: 02/08/2023]
Abstract
The growing worldwide prevalence of overnutrition and underexertion threatens the gains that we have made against atherosclerotic cardiovascular disease and other maladies. Chronic overnutrition causes the atherometabolic syndrome, which is a cluster of seemingly unrelated health problems characterized by increased abdominal girth and body-mass index, high fasting and postprandial concentrations of cholesterol- and triglyceride-rich apoB-lipoproteins (C-TRLs), low plasma HDL levels, impaired regulation of plasma glucose concentrations, hypertension, and a significant risk of developing overt type 2 diabetes mellitus (T2DM). In addition, individuals with this syndrome exhibit fatty liver, hypercoagulability, sympathetic overactivity, a gradually rising set-point for body adiposity, a substantially increased risk of atherosclerotic cardiovascular morbidity and mortality, and--crucially--hyperinsulinemia. Many lines of evidence indicate that each component of the atherometabolic syndrome arises, or is worsened by, pathway-selective insulin resistance and responsiveness (SEIRR). Individuals with SEIRR require compensatory hyperinsulinemia to control plasma glucose levels. The result is overdrive of those pathways that remain insulin-responsive, particularly ERK activation and hepatic de-novo lipogenesis (DNL), while carbohydrate regulation deteriorates. The effects are easily summarized: if hyperinsulinemia does something bad in a tissue or organ, that effect remains responsive in the atherometabolic syndrome and T2DM; and if hyperinsulinemia might do something good, that effect becomes resistant. It is a deadly imbalance in insulin action. From the standpoint of human health, it is the worst possible combination of effects. In this review, we discuss the origins of the atherometabolic syndrome in our historically unprecedented environment that only recently has become full of poorly satiating calories and incessant enticements to sit. Data are examined that indicate the magnitude of daily caloric imbalance that causes obesity. We also cover key aspects of healthy, balanced insulin action in liver, endothelium, brain, and elsewhere. Recent insights into the molecular basis and pathophysiologic harm from SEIRR in these organs are discussed. Importantly, a newly discovered oxide transport chain functions as the master regulator of the balance amongst different limbs of the insulin signaling cascade. This oxide transport chain--abbreviated 'NSAPP' after its five major proteins--fails to function properly during chronic overnutrition, resulting in this harmful pattern of SEIRR. We also review the origins of widespread, chronic overnutrition. Despite its apparent complexity, one factor stands out. A sophisticated junk food industry, aided by subsidies from willing governments, has devoted years of careful effort to promote overeating through the creation of a new class of food and drink that is low- or no-cost to the consumer, convenient, savory, calorically dense, yet weakly satiating. It is past time for the rest of us to overcome these foes of good health and solve this man-made epidemic.
Collapse
|
58
|
Abstract
PURPOSE OF REVIEW Gene-based therapies are designed to modulate gene expression in specific tissues by introducing into cells transgenes, antisense oligonucleotides, RNA interference, microRNAs, or a variety of other oligonucleotide-based compounds and their delivery systems. Several types of gene-based therapies are already available or in clinical development to treat severe lipid-related disorders and associated risk. The review briefly presents the current status and future challenges of these therapies in clinical lipidology, focusing on most advanced and promising agents or mechanisms. RECENT FINDINGS Gene-based agents address several unmet medical needs in lipidology such as homozygous familial hypercholesterolemia, familial or multifactorial chylomicronemia, severe hypertriglyceridemia, elevated lipoprotein (a), familial partial lipodystrophy, nonalcoholic fatty liver disease, and hypoalphalipoproteinemia. Most advanced antisense oligonucleotide drugs target apolipoprotein C-III, apolipoprotein (a), angiopoietin-like 3, and diacylglycerol o-acyltransferase-2. Long-term efficacy and safety data are now available for two gene-based agents, mipomersen, approved in the USA for homozygous familial hypercholesterolemia and Glybera, AAV1-LPLS447X gene therapy, conditionally approved in Europe for lipoprotein lipase deficiency. SUMMARY Although positive to date, the overall benefit-risk ratio of gene-based therapies is yet to be documented long term across additional patients and conditions. The next generation of such therapies might improve their therapeutic index.
Collapse
Affiliation(s)
- Daniel Gaudet
- Lipidology Unit, Community genomic medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21, Chicoutimi, Quebec, Canada
| | | |
Collapse
|
59
|
Low n-6/n-3 PUFA Ratio Improves Lipid Metabolism, Inflammation, Oxidative Stress and Endothelial Function in Rats Using Plant Oils as n-3 Fatty Acid Source. Lipids 2015; 51:49-59. [DOI: 10.1007/s11745-015-4091-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 10/19/2015] [Indexed: 01/10/2023]
|
60
|
Orho-Melander M. Genetics of coronary heart disease: towards causal mechanisms, novel drug targets and more personalized prevention. J Intern Med 2015; 278:433-46. [PMID: 26477595 DOI: 10.1111/joim.12407] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Coronary heart disease (CHD) is an archetypical multifactorial disorder that is influenced by genetic susceptibility as well as both modifiable and nonmodifiable risk factors, and their interactions. Advances during recent years in the field of multifactorial genetics, in particular genomewide association studies (GWASs) and their meta-analyses, have provided the statistical power to identify and replicate genetic variants in more than 50 risk loci for CHD and in several hundreds of loci for cardiometabolic risk factors for CHD such as blood lipids and lipoproteins. Although for a great majority of these loci both the causal variants and mechanisms remain unknown, progress in identifying the causal variants and underlying mechanisms has already been made for several genetic loci. Furthermore, identification of rare loss-of-function variants in genes such as PCSK9, NPC1L1, APOC3 and APOA5, which cause a markedly decreased risk of CHD and no adverse side effects, illustrates the power of translating genetic findings into novel mechanistic information and provides some optimism for the future of developing novel drugs, given the many genes associated with CHD in GWASs. Finally, Mendelian randomization can be used to reveal or exclude causal relationships between heritable biomarkers and CHD, and such approaches have already provided evidence of causal relationships between CHD and LDL cholesterol, triglycerides/remnant particles and lipoprotein(a), and indicated a lack of causality for HDL cholesterol, C-reactive protein and lipoprotein-associated phospholipase A2. Together, these genetic findings are beginning to lead to promising new drug targets and novel interventional strategies and thus have great potential to improve prevention, prediction and therapy of CHD.
Collapse
Affiliation(s)
- M Orho-Melander
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| |
Collapse
|
61
|
Stanhope KL. Sugar consumption, metabolic disease and obesity: The state of the controversy. Crit Rev Clin Lab Sci 2015; 53:52-67. [PMID: 26376619 DOI: 10.3109/10408363.2015.1084990] [Citation(s) in RCA: 431] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The impact of sugar consumption on health continues to be a controversial topic. The objective of this review is to discuss the evidence and lack of evidence that allows the controversy to continue, and why resolution of the controversy is important. There are plausible mechanisms and research evidence that supports the suggestion that consumption of excess sugar promotes the development of cardiovascular disease (CVD) and type 2 diabetes (T2DM) both directly and indirectly. The direct pathway involves the unregulated hepatic uptake and metabolism of fructose, leading to liver lipid accumulation, dyslipidemia, decreased insulin sensitivity and increased uric acid levels. The epidemiological data suggest that these direct effects of fructose are pertinent to the consumption of the fructose-containing sugars, sucrose and high fructose corn syrup (HFCS), which are the predominant added sugars. Consumption of added sugar is associated with development and/or prevalence of fatty liver, dyslipidemia, insulin resistance, hyperuricemia, CVD and T2DM, often independent of body weight gain or total energy intake. There are diet intervention studies in which human subjects exhibited increased circulating lipids and decreased insulin sensitivity when consuming high sugar compared with control diets. Most recently, our group has reported that supplementing the ad libitum diets of young adults with beverages containing 0%, 10%, 17.5% or 25% of daily energy requirement (Ereq) as HFCS increased lipid/lipoprotein risk factors for CVD and uric acid in a dose-response manner. However, un-confounded studies conducted in healthy humans under a controlled, energy-balanced diet protocol that enables determination of the effects of sugar with diets that do not allow for body weight gain are lacking. Furthermore, recent reports conclude that there are no adverse effects of consuming beverages containing up to 30% Ereq sucrose or HFCS, and the conclusions from several meta-analyses suggest that fructose has no specific adverse effects relative to any other carbohydrate. Consumption of excess sugar may also promote the development of CVD and T2DM indirectly by causing increased body weight and fat gain, but this is also a topic of controversy. Mechanistically, it is plausible that fructose consumption causes increased energy intake and reduced energy expenditure due to its failure to stimulate leptin production. Functional magnetic resonance imaging (fMRI) of the brain demonstrates that the brain responds differently to fructose or fructose-containing sugars compared with glucose or aspartame. Some epidemiological studies show that sugar consumption is associated with body weight gain, and there are intervention studies in which consumption of ad libitum high-sugar diets promoted increased body weight gain compared with consumption of ad libitum low- sugar diets. However, there are no studies in which energy intake and weight gain were compared in subjects consuming high or low sugar, blinded, ad libitum diets formulated to ensure both groups consumed a comparable macronutrient distribution and the same amounts of fiber. There is also little data to determine whether the form in which added sugar is consumed, as beverage or as solid food, affects its potential to promote weight gain. It will be very challenging to obtain the funding to conduct the clinical diet studies needed to address these evidence gaps, especially at the levels of added sugar that are commonly consumed. Yet, filling these evidence gaps may be necessary for supporting the policy changes that will help to turn the food environment into one that does not promote the development of obesity and metabolic disease.
Collapse
Affiliation(s)
- Kimber L Stanhope
- a Department of Molecular Biosciences , School of Veterinary Medicine and.,b Department of Nutrition , University of California , Davis , CA , USA
| |
Collapse
|
62
|
Kivelä AM, Huusko J, Ylä-Herttuala S. Prospect and progress of gene therapy in treating atherosclerosis. Expert Opin Biol Ther 2015; 15:1699-712. [PMID: 26328616 DOI: 10.1517/14712598.2015.1084282] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Despite considerable improvements in therapies, atherosclerotic cardiovascular diseases remain the leading cause of death worldwide. Therefore, in addition to current treatment options, new therapeutic approaches are still needed. AREAS COVERED In this review, novel gene and RNA interference-based therapy approaches and promising target genes for treating atherosclerosis are addressed. In addition, relevant animal models for the demonstration of the efficacy of different gene therapy applications, and current progress toward more efficient, targeted and safer gene transfer vectors are reviewed. EXPERT OPINION Atherosclerosis represents a complex multifactorial disease that is dependent on the interplay between lipoprotein metabolism, cellular reactions and inflammation. Recent advances and novel targets, especially in the field of RNA interference-based therapies, are very promising. However, it should be noted that the modulation of a particular gene is not as clearly associated with a complex polygenic disease as it is in the case of monogenic diseases. A deeper understanding of molecular mechanisms of atherosclerosis, further progress in vector development and the demonstration of treatment efficacy in relevant animal models will be required before gene therapy of atherosclerosis meets its clinical reality.
Collapse
Affiliation(s)
- Annukka M Kivelä
- a 1 University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine , Kuopio, Finland +358 403 552 075 ;
| | - Jenni Huusko
- a 1 University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine , Kuopio, Finland +358 403 552 075 ;
| | - Seppo Ylä-Herttuala
- a 1 University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Department of Biotechnology and Molecular Medicine , Kuopio, Finland +358 403 552 075 ; .,b 2 Science Service Center , Kuopio, Finland.,c 3 Kuopio University Hospital, Gene Therapy Unit , Kuopio, Finland
| |
Collapse
|
63
|
Bell TA, Graham MJ, Baker BF, Crooke RM. Therapeutic inhibition of apoC-III for the treatment of hypertriglyceridemia. ACTA ACUST UNITED AC 2015. [DOI: 10.2217/clp.15.7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
64
|
New insights into the pathophysiology of dyslipidemia in type 2 diabetes. Atherosclerosis 2015; 239:483-95. [PMID: 25706066 DOI: 10.1016/j.atherosclerosis.2015.01.039] [Citation(s) in RCA: 278] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 01/28/2015] [Accepted: 01/30/2015] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality for patients with type 2 diabetes, despite recent significant advances in management strategies to lessen CVD risk factors. A major cause is the atherogenic dyslipidemia, which consists of elevated plasma concentrations of both fasting and postprandial triglyceride-rich lipoproteins (TRLs), small dense low-density lipoprotein (LDL) and low high-density lipoprotein (HDL) cholesterol. The different components of diabetic dyslipidemia are not isolated abnormalities but closely linked to each other metabolically. The underlying disturbances are hepatic overproduction and delayed clearance of TRLs. Recent results have unequivocally shown that triglyceride-rich lipoproteins and their remnants are atherogenic. To develop novel strategies for the prevention and treatment of dyslipidaemia, it is essential to understand the pathophysiology of dyslipoproteinaemia in humans. Here, we review recent advances in our understanding of the pathophysiology of diabetic dyslipidemia.
Collapse
|
65
|
Amyloid-Forming Properties of Human Apolipoproteins: Sequence Analyses and Structural Insights. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 855:175-211. [PMID: 26149931 DOI: 10.1007/978-3-319-17344-3_8] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Apolipoproteins are protein constituents of lipoproteins that transport cholesterol and fat in circulation and are central to cardiovascular health and disease. Soluble apolipoproteins can transiently dissociate from the lipoprotein surface in a labile free form that can misfold, potentially leading to amyloid disease. Misfolding of apoA-I, apoA-II, and serum amyloid A (SAA) causes systemic amyloidoses, apoE4 is a critical risk factor in Alzheimer's disease, and apolipoprotein misfolding is also implicated in cardiovascular disease. To explain why apolipoproteins are over-represented in amyloidoses, it was proposed that the amphipathic α-helices, which form the lipid surface-binding motif in this protein family, have high amyloid-forming propensity. Here, we use 12 sequence-based bioinformatics approaches to assess amyloid-forming potential of human apolipoproteins and to identify segments that are likely to initiate β-aggregation. Mapping such segments on the available atomic structures of apolipoproteins helps explain why some of them readily form amyloid while others do not. Our analysis shows that nearly all amyloidogenic segments: (i) are largely hydrophobic, (ii) are located in the lipid-binding amphipathic α-helices in the native structures of soluble apolipoproteins, (iii) are predicted in both native α-helices and β-sheets in the insoluble apoB, and (iv) are predicted to form parallel in-register β-sheet in amyloid. Most of these predictions have been verified experimentally for apoC-II, apoA-I, apoA-II and SAA. Surprisingly, the rank order of the amino acid sequence propensity to form amyloid (apoB>apoA-II>apoC-II≥apoA-I, apoC-III, SAA, apoC-I>apoA-IV, apoA-V, apoE) does not correlate with the proteins' involvement in amyloidosis. Rather, it correlates directly with the strength of the protein-lipid association, which increases with increasing protein hydrophobicity. Therefore, the lipid surface-binding function and the amyloid-forming propensity are both rooted in apolipoproteins' hydrophobicity, suggesting that functional constraints make it difficult to completely eliminate pathogenic apolipoprotein misfolding. We propose that apolipoproteins have evolved protective mechanisms against misfolding, such as the sequestration of the amyloidogenic segments via the native protein-lipid and protein-protein interactions involving amphipathic α-helices and, in case of apoB, β-sheets.
Collapse
|
66
|
Siri-Tarino PW, Chiu S, Bergeron N, Krauss RM. Saturated Fats Versus Polyunsaturated Fats Versus Carbohydrates for Cardiovascular Disease Prevention and Treatment. Annu Rev Nutr 2015; 35:517-43. [PMID: 26185980 PMCID: PMC4744652 DOI: 10.1146/annurev-nutr-071714-034449] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The effects of saturated fatty acids (SFAs) on cardiovascular disease (CVD) risk are modulated by the nutrients that replace them and their food matrices. Replacement of SFAs with polyunsaturated fatty acids has been associated with reduced CVD risk, although there is heterogeneity in both fatty acid categories. In contrast, replacement of SFAs with carbohydrates, particularly sugar, has been associated with no improvement or even a worsening of CVD risk, at least in part through effects on atherogenic dyslipidemia, a cluster of traits including small, dense low-density lipoprotein particles. The effects of dietary SFAs on insulin sensitivity, inflammation, vascular function, and thrombosis are less clear. There is growing evidence that SFAs in the context of dairy foods, particularly fermented dairy products, have neutral or inverse associations with CVD. Overall dietary patterns emphasizing vegetables, fish, nuts, and whole versus processed grains form the basis of heart-healthy eating and should supersede a focus on macronutrient composition.
Collapse
Affiliation(s)
- Patty W. Siri-Tarino
- Atherosclerosis Research Program, Children’s Hospital Oakland Research Institute, Oakland, California 94609
| | - Sally Chiu
- Atherosclerosis Research Program, Children’s Hospital Oakland Research Institute, Oakland, California 94609
| | - Nathalie Bergeron
- Atherosclerosis Research Program, Children’s Hospital Oakland Research Institute, Oakland, California 94609
- College of Pharmacy, Touro University California, Vallejo, California 94594
| | - Ronald M. Krauss
- Atherosclerosis Research Program, Children’s Hospital Oakland Research Institute, Oakland, California 94609
| |
Collapse
|
67
|
Gaudet D, Brisson D, Tremblay K, Alexander VJ, Singleton W, Hughes SG, Geary RS, Baker BF, Graham MJ, Crooke RM, Witztum JL. Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med 2014; 371:2200-6. [PMID: 25470695 DOI: 10.1056/nejmoa1400284] [Citation(s) in RCA: 368] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The familial chylomicronemia syndrome is a genetic disorder characterized by severe hypertriglyceridemia and recurrent pancreatitis due to a deficiency in lipoprotein lipase (LPL). Currently, there are no effective therapies except for extreme restriction in the consumption of dietary fat. Apolipoprotein C-III (APOC3) is known to inhibit LPL, although there is also evidence that APOC3 increases the level of plasma triglycerides through an LPL-independent mechanism. We administered an inhibitor of APOC3 messenger RNA (mRNA), called ISIS 304801, to treat three patients with the familial chylomicronemia syndrome and triglyceride levels ranging from 1406 to 2083 mg per deciliter (15.9 to 23.5 mmol per liter). After 13 weeks of study-drug administration, plasma APOC3 levels were reduced by 71 to 90% and triglyceride levels by 56 to 86%. During the study, all patients had a triglyceride level of less than 500 mg per deciliter (5.7 mmol per liter) with treatment. These data support the role of APOC3 as a key regulator of LPL-independent pathways of triglyceride metabolism.
Collapse
Affiliation(s)
- Daniel Gaudet
- From the ECOGENE-21 Clinical Research Center, Chicoutimi Hospital, Chicoutimi, and the Department of Medicine, Université de Montréal, Montreal - both in Canada (D.G., D.B., K.T.); and Isis Pharmaceuticals, Carlsbad (V.J.A., W.S., S.G.H., R.S.G., B.F.B., M.J.G., R.M.C.), and the Department of Medicine, Division of Endocrinology-Metabolism, University California, San Diego, School of Medicine, La Jolla (J.L.W.) - both in California
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Abstract
INTRODUCTION Hypercholesterolaemia is a significant risk factor for cardiovascular disease (CVD), a major cause of morbidity and mortality. Up to now, the appropriate management has been aggressive hypolipidaemic therapy, particularly with statins, aiming at certain low-density lipoprotein cholesterol (LDL-C) levels for each patient population. This strategy has reduced CVD-related morbidity and mortality. However, many cardiovascular events still occur, probably as a consequence of lipid disorders other than high LDL-C concentration or other risk factors. Because statins do not eliminate the residual CVD risk, there seems to be place for novel lipid modifying drugs with different mechanisms of action. AREAS COVERED This review is an update since 2010 regarding lipid-modifying drugs in development and their potent role in clinical practice. It focuses on cholesterol ester transfer protein inhibitors, mainly anacetrapib and evacetrapib, microsomal triglyceride transfer protein inhibitors, antisense oligonucleotides, pre-protein convertase subtilisin kexin-9 inhibitors and high-density lipoprotein mimetics. EXPERT OPINION Several novel lipid-modifying drugs may be beneficial for certain patient populations. However, ongoing and future studies with clinical outcomes will clarify their actual role in clinical practice.
Collapse
Affiliation(s)
- Matilda Florentin
- Medical School, University of Ioannina, Department of Internal Medicine , Ioannina , Greece
| | | | | | | |
Collapse
|
69
|
Ding X, Yang Z, Han Y, Yu H. Fatty acid oxidation changes and the correlation with oxidative stress in different preeclampsia-like mouse models. PLoS One 2014; 9:e109554. [PMID: 25302499 PMCID: PMC4193787 DOI: 10.1371/journal.pone.0109554] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/03/2014] [Indexed: 01/18/2023] Open
Abstract
Background Long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) expression is decreased in placenta of some cases of preeclampsia (PE) which may result in free fatty acid (FFA) increased. High FFA level will induce oxidative stress, so abnormal long-chain fatty acid-oxidation may participate in the pathogenesis of PE through oxidative stress pathway. Methods PE-like groups were ApoC3 transgenic mice with abnormal fatty acid metabolism, classical PE-like models with injection of Nw-nitro-L-arginine-methyl ester (L-NA) or lipopolysaccharide (LPS) and the antiphospholipid syndrome (APS) mouse model with β2GPI injection (ApoC3+NS, ApoC3+L-NA, L-NA, LPS and β2GPI groups). The control group was wild-type mice with normal saline injection. Except for β2GPI mice, the other mice were subdivided into pre-implantation (Pre) and mid-pregnancy (Mid) subgroups by injection time. Results All PE-like groups showed hypertension and proteinuria except ApoC3+NS mice only showed hypertension. Serum FFA levels increased significantly except in LPS group compared to controls (P<0.05). LCHAD mRNA and protein expression in the liver and placenta was significantly higher for ApoC3+NS, ApoC3+L-NA and β2GPI mice and lower for L-NA mice than controls (P<0.05) but did not differ between LPS mice and controls. P47phox mRNA and protein expression in the liver significantly increased in all PE-like groups except LPS group, while P47phox expression in the placenta only significantly increased in L-NA and β2GPI groups. Conclusions Abnormal long-chain fatty acid-oxidation may play a different role in different PE-like models and in some cases participate in the pathogenesis of PE through oxidative stress pathway.
Collapse
Affiliation(s)
- Xiaoyan Ding
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, PR China
| | - Zi Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, PR China
- * E-mail:
| | - Yiwei Han
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, PR China
| | - Huan Yu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, PR China
| |
Collapse
|
70
|
Jørgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjærg-Hansen A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med 2014; 371:32-41. [PMID: 24941082 DOI: 10.1056/nejmoa1308027] [Citation(s) in RCA: 708] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND High plasma levels of nonfasting triglycerides are associated with an increased risk of ischemic cardiovascular disease. Whether lifelong low levels of nonfasting triglycerides owing to mutations in the gene encoding apolipoprotein C3 (APOC3) are associated with a reduced risk of ischemic cardiovascular disease in the general population is unknown. METHODS Using data from 75,725 participants in two general-population studies, we first tested whether low levels of nonfasting triglycerides were associated with reduced risks of ischemic vascular disease and ischemic heart disease. Second, we tested whether loss-of-function mutations in APOC3, which were associated with reduced levels of nonfasting triglycerides, were also associated with reduced risks of ischemic vascular disease and ischemic heart disease. During follow-up, ischemic vascular disease developed in 10,797 participants, and ischemic heart disease developed in 7557 of these 10,797 participants. RESULTS Participants with nonfasting triglyceride levels of less than 1.00 mmol per liter (90 mg per deciliter) had a significantly lower incidence of cardiovascular disease than those with levels of 4.00 mmol per liter (350 mg per deciliter) or more (hazard ratio for ischemic vascular disease, 0.43; 95% confidence interval [CI], 0.35 to 0.54; hazard ratio for ischemic heart disease, 0.40; 95% CI, 0.31 to 0.52). Heterozygosity for loss-of-function mutations in APOC3, as compared with no APOC3 mutations, was associated with a mean reduction in nonfasting triglyceride levels of 44% (P<0.001). The cumulative incidences of ischemic vascular disease and ischemic heart disease were reduced in heterozygotes as compared with noncarriers of APOC3 mutations (P=0.009 and P=0.05, respectively), with corresponding risk reductions of 41% (hazard ratio, 0.59; 95% CI, 0.41 to 0.86; P=0.007) and 36% (hazard ratio, 0.64; 95% CI, 0.41 to 0.99; P=0.04). CONCLUSIONS Loss-of-function mutations in APOC3 were associated with low levels of triglycerides and a reduced risk of ischemic cardiovascular disease. (Funded by the European Union and others.).
Collapse
Affiliation(s)
- Anders Berg Jørgensen
- From Copenhagen University Hospital and Faculty of Health and Medical Sciences, University of Copenhagen (A.B.J., R.F.-S., B.G.N., A.T.-H.), the Department of Clinical Biochemistry, Rigshospitalet (A.B.J., R.F.-S., A.T.-H.), the Department of Clinical Biochemistry (B.G.N.) and the Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital, and the Copenhagen City Heart Study, Frederiksberg Hospital (B.G.N., A.T.-H.) - all in Copenhagen
| | | | | | | |
Collapse
|
71
|
Krauss RM. All Low-Density Lipoprotein Particles Are Not Created Equal. Arterioscler Thromb Vasc Biol 2014; 34:959-61. [DOI: 10.1161/atvbaha.114.303458] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ronald M. Krauss
- From the Children’s Hospital Oakland Research Institute, Oakland, CA
| |
Collapse
|
72
|
Yao Z, Zhang L, Ji G. Efficacy of polyphenolic ingredients of Chinese herbs in treating dyslipidemia of metabolic syndromes. JOURNAL OF INTEGRATIVE MEDICINE 2014; 12:135-146. [PMID: 24861834 DOI: 10.1016/s2095-4964(14)60023-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
There is an increasing interest and popularity of Chinese herbal medicine worldwide, which is accompanied by increasing concerns about its effectiveness and potential toxicity. Several ingredients, such as polyphenolic compounds berberine, flavonoids, and curcumin, have been studied extensively by using various animal models. Effectiveness of treatment and amelioration of metabolic syndromes, including insulin resistance and dyslipidemia, has been demonstrated. This review summarizes the major checkpoints and contributing factors in regulation of exogenous and endogenous lipid metabolism, with particular emphasis centered on triglyceride-rich and cholesterol-rich lipoproteins. Available experimental evidence demonstrating the lipid-lowering effect of berberine, flavonoids and curcumin in cell culture and animal models is compiled, and the strengths and shortcomings of experimental designs in these studies are discussed.
Collapse
Affiliation(s)
- Zemin Yao
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of System Biology, University of Ottawa, Ottawa K1H 8M5, Canada; E-mail:
| | - Li Zhang
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
73
|
Zheng C. Updates on apolipoprotein CIII: fulfilling promise as a therapeutic target for hypertriglyceridemia and cardiovascular disease. Curr Opin Lipidol 2014; 25:35-9. [PMID: 24345989 DOI: 10.1097/mol.0000000000000040] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The lipid hypothesis of atherosclerosis is mainly predicated on the function of apolipoprotein (apo)B lipoproteins, which promote atherosclerosis, and apoA lipoproteins, which prevent it. However, accumulating evidence suggests causal roles of other apolipoproteins, abundant surface components of apoB and apoA lipoprotein, in promoting atherosclerosis and other metabolic diseases. This article reviews recent literature on one such apolipoprotein: apoCIII. RECENT FINDINGS Population studies have consistently demonstrated that plasma apoCIII strongly predicts cardiovascular disease. ApoCIII's atherogenicity was traditionally attributed to hypertriglyceridemia because of its inhibition on the lipolysis of triglyceride-rich lipoproteins. Recent evidence expands this function and reveals apoCIII's key role in hepatic assembly and secretion of triglyceride-rich lipoproteins. In addition to these indirect atherogenic functions mediated through dyslipidemia, recent research discovers that apoCIII directly provoke proinflammatory responses in vascular cells, including monocytes and endothelial cells. These direct atherogenic effects are dependent on apoCIII. ApoCIII is also involved in pancreatic beta-cell biology and contributes to type I diabetes. SUMMARY Recent data further strengthen the theory that apoCIII exerts strong atherogenic functions through both indirect and direct mechanisms. Encouraging results from early stage clinical trials demonstrate that modulating apoCIII per se is a novel and potent therapeutic approach to managing dyslipidemia and cardiovascular disease risk.
Collapse
Affiliation(s)
- Chunyu Zheng
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
74
|
Hibert P, Prunier-Mirebeau D, Beseme O, Chwastyniak M, Tamareille S, Pinet F, Prunier F. Modifications in rat plasma proteome after remote ischemic preconditioning (RIPC) stimulus: identification by a SELDI-TOF-MS approach. PLoS One 2014; 9:e85669. [PMID: 24454915 PMCID: PMC3890329 DOI: 10.1371/journal.pone.0085669] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/06/2013] [Indexed: 11/18/2022] Open
Abstract
Remote ischemic preconditioning’s (RIPC) ability to render the myocardium resistant to subsequent prolonged ischemia is now clearly established in different species, including humans. Strong evidence suggests that circulating humoral mediators play a key role in signal transduction, but their identities still need to be established. Our study sought to identify potential circulating RIPC mediators using a proteomic approach. Rats were exposed to 10-min limb ischemia followed by 5- (RIPC 5′) or 10-min (RIPC 10′) reperfusion prior to blood sampling. The control group only underwent blood sampling. Plasma samples were isolated for proteomic analysis using surface-enhanced laser desorption and ionization - time of flight - mass spectrometry (SELDI-TOF-MS). A total of seven proteins, including haptoglobin and transthyretin, were detected as up- or down-regulated in response to RIPC. These proteins had previously been identified as associated with organ protection, anti-inflammation, and various cellular and molecular responses to ischemia. In conclusion, this study indicates that RIPC results in significant modulations of plasma proteome.
Collapse
Affiliation(s)
- Pierre Hibert
- L’UNAM Université, Angers, France
- Université d’Angers, Laboratoire Cardioprotection Remodelage et Thrombose, Angers, France
| | - Delphine Prunier-Mirebeau
- L’UNAM Université, Angers, France
- Université d’Angers, INSERM U771, CNRS UMR 6214, CHU Angers, Département de Biochimie et Génétique, Angers, France
| | - Olivia Beseme
- INSERM, U744, Lille, France
- Institut Pasteur de Lille, Lille, France
- Université Lille Nord de France, IFR142, Lille, France
| | - Maggy Chwastyniak
- INSERM, U744, Lille, France
- Institut Pasteur de Lille, Lille, France
- Université Lille Nord de France, IFR142, Lille, France
| | - Sophie Tamareille
- L’UNAM Université, Angers, France
- Université d’Angers, Laboratoire Cardioprotection Remodelage et Thrombose, Angers, France
| | - Florence Pinet
- INSERM, U744, Lille, France
- Institut Pasteur de Lille, Lille, France
- Université Lille Nord de France, IFR142, Lille, France
- Centre Hospitalier régional et Universitaire de Lille, Lille, France
| | - Fabrice Prunier
- L’UNAM Université, Angers, France
- Université d’Angers, Laboratoire Cardioprotection Remodelage et Thrombose, Angers, France
- CHU Angers, Service de Cardiologie, Angers, France
- * E-mail:
| |
Collapse
|
75
|
Abstract
Cardiovascular disease represents the most common cause of death in patients with nonalcoholic fatty liver disease (NAFLD). Patients with NAFLD exhibit an atherogenic dyslipidemia that is characterized by an increased plasma concentration of triglycerides, reduced concentration of high-density lipoprotein (HDL) cholesterol, and low-density lipoprotein (LDL) particles that are smaller and more dense than normal. The pathogenesis of NAFLD-associated atherogenic dyslipidemia is multifaceted, but many aspects are attributable to manifestations of insulin resistance. Here the authors review the structure, function, and metabolism of lipoproteins, which are macromolecular particles of lipids and proteins that transport otherwise insoluble triglyceride and cholesterol molecules within the plasma. They provide a current explanation of the metabolic perturbations that are observed in the setting of insulin resistance. An improved understanding of the pathophysiology of atherogenic dyslipidemia would be expected to guide therapies aimed at reducing morbidity and mortality in patients with NAFLD.
Collapse
Affiliation(s)
- Edward Fisher
- Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, New York
| | - David Cohen
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
76
|
Borén J, Taskinen MR, Olofsson SO, Levin M. Ectopic lipid storage and insulin resistance: a harmful relationship. J Intern Med 2013; 274:25-40. [PMID: 23551521 DOI: 10.1111/joim.12071] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Obesity increases the risk of metabolic diseases, including insulin resistance and type 2 diabetes, as well as cardiovascular disease. In addition to lipid accumulation in adipose tissue, obesity is associated with increased lipid storage in ectopic tissues, such as skeletal muscle and liver. Furthermore, lipid accumulation in the heart may result in cardiac dysfunction and heart failure. It has recently been demonstrated that intracellular lipid accumulation in ectopic tissues leads to pathological responses and impaired insulin signalling. Here, we will review the current understanding of how lipid storage and lipid droplet physiology affect the risk of developing metabolic diseases.
Collapse
Affiliation(s)
- J Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | | | | | | |
Collapse
|
77
|
Yao Z, Zhou H, Figeys D, Wang Y, Sundaram M. Microsome-associated lumenal lipid droplets in the regulation of lipoprotein secretion. Curr Opin Lipidol 2013; 24:160-70. [PMID: 23123764 DOI: 10.1097/mol.0b013e32835aebe7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Liver is the major organ in mammals that possesses the capacity to release triglyceride within VLDL. VLDL assembly requires apolipoprotein (apo) B-100 with the assistance of microsomal triglyceride transfer protein (MTP), which facilitates the mobilization of triglyceride into the microsomal lumen. Recent experimental evidence has suggested that the lumenal triglyceride associated with endoplasmic reticulum (ER)/Golgi may represent an entity serving as precursors for large VLDL1. RECENT FINDINGS Under lipid-rich conditions, discrete triglyceride-rich lipidic bodies, termed lumenal lipid droplets, are accumulated in association with ER/Golgi microsomes. Formation of the microsome-associated lumenal lipid droplets (MALD) is dependent on the activity of MTP, and the resulting apoB-free lipidic body is associated with a variety of proteins including apolipoproteins that are components of VLDL. Formation and utilization of MALD during the assembly and secretion of VLDL1 have a profound influence on hepatic cell physiology, such as ER stress responses. SUMMARY This review summarizes current understanding of hepatic triglyceride homeostasis in general, and highlights the functional significance of triglyceride compartmentalization between cytosol and microsomes in particular. Understanding of MALD metabolism may shed new light on the prevention and treatment of liver diseases associated with abnormally elevated intracellular triglycerides.
Collapse
Affiliation(s)
- Zemin Yao
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada.
| | | | | | | | | |
Collapse
|
78
|
Graham MJ, Lee RG, Bell TA, Fu W, Mullick AE, Alexander VJ, Singleton W, Viney N, Geary R, Su J, Baker BF, Burkey J, Crooke ST, Crooke RM. Antisense oligonucleotide inhibition of apolipoprotein C-III reduces plasma triglycerides in rodents, nonhuman primates, and humans. Circ Res 2013; 112:1479-90. [PMID: 23542898 DOI: 10.1161/circresaha.111.300367] [Citation(s) in RCA: 289] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE Elevated plasma triglyceride levels have been recognized as a risk factor for the development of coronary heart disease. Apolipoprotein C-III (apoC-III) represents both an independent risk factor and a key regulatory factor of plasma triglyceride concentrations. Furthermore, elevated apoC-III levels have been associated with metabolic syndrome and type 2 diabetes mellitus. To date, no selective apoC-III therapeutic agent has been evaluated in the clinic. OBJECTIVE To test the hypothesis that selective inhibition of apoC-III with antisense drugs in preclinical models and in healthy volunteers would reduce plasma apoC-III and triglyceride levels. METHODS AND RESULTS Rodent- and human-specific second-generation antisense oligonucleotides were identified and evaluated in preclinical models, including rats, mice, human apoC-III transgenic mice, and nonhuman primates. We demonstrated the selective reduction of both apoC-III and triglyceride in all preclinical pharmacological evaluations. We also showed that inhibition of apoC-III was well tolerated and not associated with increased liver triglyceride deposition or hepatotoxicity. A double-blind, placebo-controlled, phase I clinical study was performed in healthy subjects. Administration of the human apoC-III antisense drug resulted in dose-dependent reductions in plasma apoC-III, concomitant lowering of triglyceride levels, and produced no clinically meaningful signals in the safety evaluations. CONCLUSIONS Antisense inhibition of apoC-III in preclinical models and in a phase I clinical trial with healthy subjects produced potent, selective reductions in plasma apoC-III and triglyceride, 2 known risk factors for cardiovascular disease. This compelling pharmacological profile supports further clinical investigations in hypertriglyceridemic subjects.
Collapse
|
79
|
Fan N, Lai L. Genetically modified pig models for human diseases. J Genet Genomics 2013; 40:67-73. [PMID: 23439405 DOI: 10.1016/j.jgg.2012.07.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 12/10/2012] [Accepted: 12/30/2012] [Indexed: 02/08/2023]
Abstract
Genetically modified animal models are important for understanding the pathogenesis of human disease and developing therapeutic strategies. Although genetically modified mice have been widely used to model human diseases, some of these mouse models do not replicate important disease symptoms or pathology. Pigs are more similar to humans than mice in anatomy, physiology, and genome. Thus, pigs are considered to be better animal models to mimic some human diseases. This review describes genetically modified pigs that have been used to model various diseases including neurological, cardiovascular, and diabetic disorders. We also discuss the development in gene modification technology that can facilitate the generation of transgenic pig models for human diseases.
Collapse
Affiliation(s)
- Nana Fan
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | | |
Collapse
|
80
|
Pathobiology of obstructive sleep apnea-related dyslipidemia: focus on the liver. ISRN CARDIOLOGY 2013; 2013:687069. [PMID: 23346414 PMCID: PMC3549373 DOI: 10.1155/2013/687069] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/18/2012] [Indexed: 11/26/2022]
Abstract
Obstructive sleep apnea and dyslipidemia are common medical disorders that independently increase vascular morbidity and mortality. Current animal and human data show that, indeed, obstructive sleep apnea may mediate pathological alterations in cholesterol and triglyceride metabolism. The mechanisms involved are increased lipolysis, decreased lipoprotein clearance, and enhanced lipid output from the liver. Human evidence shows that the treatment of obstructive sleep apnea with continuous positive airway pressure leads to an improvement of postprandial hyperlipidemia. However, more studies are needed, to clarify the pathophysiology of the interrelationship between obstructive sleep apnea and dyslipidemia and whether treatment of obstructive sleep apnea will lead to an improvement in the lipid profile and, more importantly, reduce hyperlipidemia-related vascular outcomes.
Collapse
|
81
|
Sparks JD, Sparks CE, Adeli K. Selective hepatic insulin resistance, VLDL overproduction, and hypertriglyceridemia. Arterioscler Thromb Vasc Biol 2012; 32:2104-12. [PMID: 22796579 DOI: 10.1161/atvbaha.111.241463] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Insulin plays a central role in regulating energy metabolism, including hepatic transport of very low-density lipoprotein (VLDL)-associated triglyceride. Hepatic hypersecretion of VLDL and consequent hypertriglyceridemia leads to lower circulating high-density lipoprotein levels and generation of small dense low-density lipoproteins characteristic of the dyslipidemia commonly observed in metabolic syndrome and type 2 diabetes mellitus. Physiological fluctuations of insulin modulate VLDL secretion, and insulin inhibition of VLDL secretion upon feeding may be the first pathway to become resistant in obesity that leads to VLDL hypersecretion. This review summarizes the role of insulin-related signaling pathways that determine hepatic VLDL production. Disruption in signaling pathways that reduce generation of the second messenger phosphatidylinositide (3,4,5) triphosphate downstream of activated phosphatidylinositide 3-kinase underlies the development of VLDL hypersecretion. As insulin resistance progresses, a number of pathways are altered that further augment VLDL hypersecretion, including hepatic inflammatory pathways. Insulin plays a complex role in regulating glucose metabolism, and it is not surprising that the role of insulin in VLDL and lipid metabolism will prove equally complex.
Collapse
Affiliation(s)
- Janet D Sparks
- University of Rochester Medical Center, Department of Pathology and Laboratory Medicine, Rochester, NY, USA
| | | | | |
Collapse
|