1
|
Elshorbagy A, Lyons AR, Vallejo-Vaz AJ, Stevens CA, Dharmayat KI, Brandts J, Catapano AL, Freiberger T, Hovingh GK, Mata P, Raal FJ, Santos RD, Soran H, Watts GF, Abifadel M, Aguilar-Salinas CA, Alhabib KF, Alkhnifsawi M, Almahmeed W, Alonso R, Al-Rasadi K, Al-Sarraf A, Ashavaid TF, Banach M, Binder CJ, Bourbon M, Brunham LR, Chlebus K, Corral P, Cruz D, Davletov K, Descamps OS, Ezhov M, Gaita D, Groselj U, Harada-Shiba M, Holven KB, Kayikcioglu M, Khovidhunkit W, Lalic K, Latkovskis G, Laufs U, Liberopoulos E, Lima-Martinez MM, Lin J, Maher V, Marais AD, März W, Mirrakhimov E, Miserez AR, Mitchenko O, Nawawi H, Nordestgaard BG, Panayiotou AG, Paragh G, Petrulioniene Z, Pojskic B, Postadzhiyan A, Reda A, Reiner Ž, Reyes X, Sadiq F, Sadoh WE, Schunkert H, Shek AB, Stroes E, Su TC, Subramaniam T, Susekov AV, Tilney M, Tomlinson B, Truong TH, Tselepis AD, Tybjærg-Hansen A, Vázquez AC, Viigimaa M, Vohnout B, Wang L, Yamashita S, Arca M, Averna M, Schreier L, Pang J, Ebenbichler C, Dieplinger H, Innerhofer R, Winhofer-Stöckl Y, Greber-Platzer S, Krychtiuk K, Speidl W, Toplak H, Widhalm K, Stulnig T, Huber K, Höllerl F, Rega-Kaun G, Kleemann L, Mäser M, Scholl-Bürgi S, Säly C, Mayer FJ, Sperone A, Tanghe C, Gérard AC, Pojskic L, Sisic I, Durak Nalbantic A, Ejubovic M, Jannes CE, Pereira AC, Krieger JE, Petrov I, Goudev A, Nikolov F, Tisheva S, Yotov Y, Tzvetkov I, Baass A, Bergeron J, Bernard S, Brisson D, Brunham LR, Cermakova L, Couture P, Francis GA, Gaudet D, Hegele RA, Khoury E, Mancini GJ, McCrindle BW, Paquette M, Ruel I, Iatan I, Cuevas A, Wang X, Meng K, Song X, Yong Q, Jiang T, Liu Z, Duan Y, Hong J, Ye P, Chen Y, Qi J, Liu Z, Li Y, Zhang C, Peng J, Yang Y, Yu W, Wang Q, Yuan H, Cheng S, Jiang L, Chong M, Jiao J, Wu Y, Wen W, Xu L, Zhang R, Qu Y, He J, Fan X, Wang Z, Chow E, Pećin I, Perica D, Symeonides P, Vrablik M, Ceska R, Soska V, Tichy L, Adamkova V, Franekova J, Cifkova R, Kraml P, Vonaskova K, Cepova J, Dusejovska M, Pavlickova L, Blaha V, Rosolova H, Nussbaumerova B, Cibulka R, Vaverkova H, Cibickova L, Krejsova Z, Rehouskova K, Malina P, Budikova M, Palanova V, Solcova L, Lubasova A, Podzimkova H, Bujdak J, Vesely J, Jordanova M, Salek T, Urbanek R, Zemek S, Lacko J, Halamkova H, Machacova S, Mala S, Cubova E, Valoskova K, Burda L, Benn M, Bendary A, Daoud I, Emil S, Elbahry A, Rafla S, Sanad O, Kazamel G, Ashraf DM, Sobhy M, El-Hadidy A, Shafy MA, Kamal S, Bendary M, Talviste G, Christmann J, Dressel A, Fath F, Ferraro C, Frenzke L, Gopon A, Klein I, Pienkowska D, Sietmann T, Sonntag A, Adjan O, Bahrmann P, Baessler A, Barkowski R, Beckerdjian R, Berr C, Birkenfeld A, Böll G, Carstensen A, Demuth I, Finkernagel H, Gouni-Berthold I, Hahmann H, Hamerle M, Halder J, Heide M, Julius U, Kassner U, Katzmann JL, Kirschbaum A, Klose G, Könemann S, König C, König W, Krämer B, Kuprat G, Koschker AC, Krämer B, Kilic Ö, Laufs U, Lindenmeier G, Van de Loo I, Lorenz B, Lorenz E, Löhr B, McChord J, Maslarska M, Methe H, Merkel M, Moussaoui Z, Müller-Kozarez I, Olivier CB, Ong P, Otte B, Parhofer K, Partsch CJ, Paulus M, Pehlivanli S, Pflederer T, Pusl T, Richter V, Rosner S, Sanin V, Schäfer S, Schäfer C, Schatz U, Schirmer S, Schmidt C, Seeger W, Sisovic S, Spens A, Jablonski KS, Stadelmann A, Steinhagen-Thiessen E, Stürzebecher P, Tafelmeier M, Tillack D, Tselmin S, Tünnemann-Tarr A, Vogt A, Beckerath JV, Wilke A, Wolf U, Zemmrich C, Rizos CV, Skoumas I, Tziomalos K, Rallidis L, Kotsis V, Doumas M, Athyros V, Skalidis E, Kolovou G, Kolovou V, Garoufi A, Bilianou E, Koutagiar I, Kiouri E, Antza C, Zacharis E, Attilakos A, Sfikas G, Koumaras C, Anagnostis P, Anastasiou G, Liamis G, Koutsogianni AD, Petkou E, Milionis H, Koulouri A, Prodromiadou E, Karányi Z, Harangi M, Bajnok L, Audikovszky M, Márk L, Benczúr B, Reiber I, Nagy G, Nagy A, Reddy LL, Shah SAV, Ponde CK, Dalal JJ, Sawhney JP, Verma IC, Altaey M, Al-Jumaily K, Rasul D, Abdalsahib AF, Jabbar AA, Al-ageedi M, Abdalsahib AF, Al-ageedi M, Dhamin M, AlFil S, Khadhim F, Miahy S, Agar R, Catapano AL, Arca M, Averna M, Calandra S, Tarugi P, Casula M, Galimberti F, Olmastroni E, Sarzani R, Ferri C, Repetti E, Piro S, Suppressa P, Meregalli G, Borghi C, Muntoni S, Calabrò P, Cipollone F, Purrello F, Pujia A, Passaro A, Marcucci R, Pecchioli V, Pisciotta L, Mandraffino G, Pellegatta F, Mombelli G, Branchi A, Fiorenza AM, Pederiva C, Werba JP, Parati G, Carubbi F, Iughetti L, Fortunato G, Iannuzzi A, Iannuzzo G, Cefalù AB, Biasucci G, Zambon S, Pirro M, Sbrana F, Trenti C, D'Erasmo L, Federici M, Ben MD, Bartuli A, Giaccari A, Pipolo A, Citroni N, Guardamagna O, Lia S, Benso A, Biolo G, Maroni L, Lupi A, Bonanni L, Rinaldi E, Zenti MG, Matsuki K, Hori M, Ogura M, Masuda D, Kobayashi T, Nagahama K, Al-Jarallah M, Radovic M, Lunegova O, Bektasheva E, Abilova S, Erglis A, Gilis D, Nesterovics G, Saripo V, Meiere R, Skudrina G, Terauda E, Jambart S, Ayoub C, Ghaleb Y, Aliosaitiene U, Kutkiene S, Abdul Kadir SHS, Kasim NAM, Nor NSM, Abdul Hamid H, Abdul Razak S, Al-Khateeb A, Abd Muid S, Abdul Rahman T, Kasim SS, Radzi ABM, Ibrahim KS, Rosli MM, Razali R, Chua YA, Razman AZ, Nazli SA, Aziz N, Rosman A, Abdul Murad N, Jalaludin MA, Abdul Latif AZ, Azzopardi C, Mehta R, Martagon AJ, Ramirez GAG, Villa NEA, Vazquez AV, Elias-Lopez D, Retana GG, Rodriguez B, Macías JJC, Zazueta AR, Alvarado RM, Portano JDM, Lopez HA, Sauque-Reyna L, Herrera LGG, Mendia LES, Aguilar HG, Cooremans ER, Aparicio BP, Zubieta VM, Gonzalez PAC, Ferreira-Hermosillo A, Portilla NC, Dominguez GJ, Garcia AYR, Cazares HEA, Gonzalez JR, Valencia CVM, Padilla FG, Prado RM, Ibarra MODLR, Villicaña RDA, Rivera KJA, Carrera RA, Alvarez JA, Martinez JCA, Bustillo MDLRB, Vargas GC, Chacon RC, Andrade MHF, Ortega AF, Alcala HG, de Leon LEG, Guzman BG, Garcia JJG, Cuellar JCG, Cruz JRG, Garcia AH, Almada JRH, Herrera UJ, Sobrevilla FL, Rodriguez EM, Sibaja CM, Rodriguez ABM, Oyervides JCM, Vazquez DIP, Rodriguez EAR, Osorio MLR, Saucedo JR, Tamayo MT, Talavera LAV, Arroyo LEV, Carrillo EAZ, Stroes ES, Defesche J, Zuurbier L, Reeskamp L, Ibrahim S, Roeters van Lennep J, Wiegman A, Isara A, Obaseki DE, Al-Waili K, Al-Zadjali F, Al-Zakwani I, Al-Kindi M, Al-Mukhaini S, Al-Barwani H, Rana A, Shah LSU, Al-Nouri F, Starostecka E, Konopka A, Bielecka-Dabrowa A, Lewek J, Sosnowska B, Gąsior M, Dyrbuś K, Jóźwiak J, Pajkowski M, Romanowska-Kocejko M, Żarczyńska-Buchowiecka M, Chmara M, Wasąg B, Stróżyk A, Michalska-Grzonkowska A, Medeiros AM, Alves AC, Silva F, Lobarinhas G, Palma I, de Moura JP, Rico MT, Rato Q, Pais P, Correia S, Moldovan O, Virtuoso MJ, Araujo F, Salgado JM, Colaço I, Dumitrescu A, Lengher C, Mosteoru S, Meshkov A, Ershova A, Rozhkova T, Korneva V, Yu KT, Zafiraki V, Voevoda M, Gurevich V, Duplyakov D, Ragino Y, Chubykina U, Shaposhnik I, Alkaf F, Khudari A, Rwaili N, Al-Allaf F, Alghamdi M, Batais MA, Almigbal TH, Kinsara A, AlQudaimi AHA, Awan Z, Elamin OA, Altaradi H, Popovic L, Singh S, Rasulic I, Petakov A, Lalic NM, Lam C, Le TJ, Siang ELT, Dissanayake S, I-Shing JT, Shyong TE, Jin TCS, Ting SPL, Ming JHK, Drum CL, Nastar FA, Jia LW, Ya NKS, Jie MCW, Dalan R, Wei YQ, sian TY, Keong YK, Rong SK, Jin DSE, Ming IKJ, Chang TH, Peng FYK, Vasanwala RF, Raslova K, Balinth K, Buganova I, Fabryova L, Kadurova M, Klabnik A, Kozárová M, Sirotiakova J, Battelino T, Cevc M, Debeljak M, Torkar AD, Fras Z, Jug B, Cugalj BK, Kovac J, Mlinaric M, Sikonja J, Pilcher GJ, Blom DJ, Wolmarans KH, Brice BC, Muñiz-Grijalvo O, Díaz-Díaz JL, de Isla LP, Fuentes F, Badimon L, Martin F, Miserez EB, Shipton JL, Ganokroj P, Chattranukulchai P, Jiamjarasrungsi W, Thongtang N, Krittayaphong R, Vathesatogkit P, Sriphrapradang C, Phimphilai M, Leelawattana R, Anthanont P, Suraamornkul S, Deerochanawong C, Senthong V, Torpongpun A, Suteerayongprasert P, Pengpong N, Sathavarodom N, Sunanta U, Porntharukchareon T, Kiatpanabhikul P, Kaewkrasaesin C, Kongkit J, Umphonsathien M, Akbulut M, Alici G, Bayram F, Can LH, Celik A, Ceyhan C, Coskun FY, Demir M, Demircan S, Dogan V, Durakoglugil E, Dural İE, Gedikli O, Hacioglu A, Ildizli M, Kilic S, Kirilmaz B, Kutlu M, Oguz A, Ozdogan O, Onrat E, Ozer S, Sabuncu T, Sahin T, Sivri F, Sonmez A, Temizhan A, Topcu S, Tokgozoglu L, Tuncez A, Vural M, Yenercag M, Yesilbursa D, Yigit Z, Yildirim AB, Yildirir A, Yilmaz MB, Atallah B, Traina M, Sabbour H, Abdul Hay D, Luqman N, Elfatih A, Abdulrasheed A, Manla Y, Kwok S, DellOca N, Alieva RB, Fozilov KG, Hoshimov SU, Nizamov UI, Kan LE, Kim AR, Abdullaeva GJ, Abdullaev AA, Do DL, Nguyen MNT, Kim NT, Le TT, Le HA, Ray KK. Association of BMI, lipid-lowering medication, and age with prevalence of type 2 diabetes in adults with heterozygous familial hypercholesterolaemia: a worldwide cross-sectional study. Lancet Diabetes Endocrinol 2024; 12:811-823. [PMID: 39374602 DOI: 10.1016/s2213-8587(24)00221-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/05/2024] [Accepted: 07/15/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Statins are the cornerstone treatment for patients with heterozygous familial hypercholesterolaemia but research suggests it could increase the risk of type 2 diabetes in the general population. A low prevalence of type 2 diabetes was reported in some familial hypercholesterolaemia cohorts, raising the question of whether these patients are protected against type 2 diabetes. Obesity is a well known risk factor for the development of type 2 diabetes. We aimed to investigate the associations of known key determinants of type 2 diabetes with its prevalence in people with heterozygous familial hypercholesterolaemia. METHODS This worldwide cross-sectional study used individual-level data from the EAS FHSC registry and included adults older than 18 years with a clinical or genetic diagnosis of heterozygous familial hypercholesterolaemia who had data available on age, BMI, and diabetes status. Those with known or suspected homozygous familial hypercholesterolaemia and type 1 diabetes were excluded. The main outcome was prevalence of type 2 diabetes overall and by WHO region, and in relation to obesity (BMI ≥30·0 kg/m2) and lipid-lowering medication as predictors. The study population was divided into 12 risk categories based on age (tertiles), obesity, and receiving statins, and the risk of type 2 diabetes was investigated using logistic regression. FINDINGS Among 46 683 adults with individual-level data in the FHSC registry, 24 784 with heterozygous familial hypercholesterolaemia were included in the analysis from 44 countries. 19 818 (80%) had a genetically confirmed diagnosis of heterozygous familial hypercholesterolaemia. Type 2 diabetes prevalence in the total population was 5·7% (1415 of 24 784), with 4·1% (817 of 19 818) in the genetically diagnosed cohort. Higher prevalence of type 2 diabetes was observed in the Eastern Mediterranean (58 [29·9%] of 194), South-East Asia and Western Pacific (214 [12·0%] of 1785), and the Americas (166 [8·5%] of 1955) than in Europe (excluding the Netherlands; 527 [8·0%] of 6579). Advancing age, a higher BMI category (obesity and overweight), and use of lipid-lowering medication were associated with a higher risk of type 2 diabetes, independent of sex and LDL cholesterol. Among the 12 risk categories, the probability of developing type 2 diabetes was higher in people in the highest risk category (aged 55-98 years, with obesity, and receiving statins; OR 74·42 [95% CI 47·04-117·73]) than in those in the lowest risk category (aged 18-38 years, without obesity, and not receiving statins). Those who did not have obesity, even if they were in the upper age tertile and receiving statins, had lower risk of type 2 diabetes (OR 24·42 [15·57-38·31]). The corresponding results in the genetically diagnosed cohort were OR 65·04 (40·67-104·02) for those with obesity in the highest risk category and OR 20·07 (12·73-31·65) for those without obesity. INTERPRETATION Adults with heterozygous familial hypercholesterolaemia in most WHO regions have a higher type 2 diabetes prevalence than in Europe. Obesity markedly increases the risk of diabetes associated with age and use of statins in these patients. Our results suggest that heterozygous familial hypercholesterolaemia does not protect against type 2 diabetes, hence managing obesity is essential to reduce type 2 diabetes in this patient population. FUNDING Pfizer, Amgen, MSD, Sanofi-Aventis, Daiichi-Sankyo, and Regeneron.
Collapse
|
2
|
Basdorf P, Kocher T, Baumeister SE, Pink C, Budde K, Petersmann A, Friedrich N, Völzke H, Nauck M, Holtfreter B. Periodontitis adversely affects lipoprotein subfractions - results from the cohort study SHIP-TREND: Periodontitis adversely affects lipoprotein subfractions. DIABETES & METABOLISM 2024; 50:101584. [PMID: 39396553 DOI: 10.1016/j.diabet.2024.101584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/19/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
AIM We aimed to investigate the medium-term associations of periodontitis and the number of missing teeth with serum lipoproteins and their plasma subfractions using follow-up data from the population-based Study of Health in Pomerania (SHIP-TREND). METHODS A total of 2,058 participants with 7-year follow-up data underwent periodontal examinations, serum lipid panel tests, and proton nuclear magnetic resonance (1H-NMR) spectroscopy of plasma lipoproteins and their subfractions. Generalized models with gamma distribution and loglink were used to analyze associations between periodontal variables and lipoproteins and their subfractions, adjusting for confounders using propensity score weighting. RESULTS Periodontal variables were consistently associated with elevated follow-up serum levels of triglycerides, total cholesterol, and low-density lipoprotein cholesterol levels. When plasma lipoprotein subfractions were evaluated, periodontal variables were associated with elevated levels of triglycerides and cholesterol-enriched apolipoprotein B-containing lipoprotein particles, particularly small dense low-density lipoprotein, very-low-density lipoprotein and intermediate density lipoprotein. In addition, altered high-density lipoprotein particle composition was observed, suggesting potential functional changes. CONCLUSION This study provides evidence for causal effects of periodontitis on conventional serum lipids and plasma lipoprotein subfractions. As the underlying biological mechanisms are not fully understood, further research is needed.
Collapse
Affiliation(s)
- Pascal Basdorf
- Department of Restorative Dentistry, Periodontology and Endodontology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Kocher
- Department of Restorative Dentistry, Periodontology and Endodontology, University Medicine Greifswald, Greifswald, Germany
| | | | - Christiane Pink
- Department of Restorative Dentistry, Periodontology and Endodontology, University Medicine Greifswald, Greifswald, Germany
| | - Kathrin Budde
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Astrid Petersmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany; Institute for Community Medicine, SHIP/Clinical-Epidemiological Research, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Birte Holtfreter
- Department of Restorative Dentistry, Periodontology and Endodontology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
3
|
Zubirán R, Cruz-Bautista I, Aguilar-Salinas CA. Interaction Between Primary Hyperlipidemias and Type 2 Diabetes: Therapeutic Implications. Diabetes Ther 2024; 15:1979-2000. [PMID: 39080218 PMCID: PMC11330433 DOI: 10.1007/s13300-024-01626-2] [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: 06/07/2024] [Accepted: 07/10/2024] [Indexed: 08/18/2024] Open
Abstract
There is a gap of knowledge about the clinical and pathophysiological implications resulting from the interaction between primary hyperlipidemias and type 2 diabetes (T2D). Most of the existing evidence comes from sub-analyses of cohorts; scant information derives from randomized clinical trials. The expected clinical implications of T2D in patients with primary hyperlipidemias is an escalation of their already high cardiovascular risk. There is a need to accurately identify patients with this dual burden and to adequately prescribe lipid-lowering therapies, with the current advancements in newer therapeutic options. This review provides an update on the interactions of primary hyperlipidemias, such as familial combined hyperlipidemia, familial hypercholesterolemia, multifactorial chylomicronemia, lipoprotein (a), and type 2 diabetes.
Collapse
Affiliation(s)
- Rafael Zubirán
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ivette Cruz-Bautista
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Carlos A Aguilar-Salinas
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico.
- Dirección de Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico.
| |
Collapse
|
4
|
Brunham LR. Familial hypercholesterolemia-Plus: is the metabolic syndrome changing the clinical picture of familial hypercholesterolemia? Curr Opin Lipidol 2024; 35:219-221. [PMID: 38640084 DOI: 10.1097/mol.0000000000000938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review article was to describe recent advances in our knowledge about how diabetes and metabolic syndrome are changing the face of familial hypercholesterolemia. RECENT FINDINGS Heterozygous familial hypercholesterolemia, most commonly caused by disruption to LDL receptor function, leads to lifelong elevation of LDL cholesterol and increased risk of atherosclerotic cardiovascular disease. Familial hypercholesterolemia was originally described as a form of 'pure' hypercholesterolemia, in the sense that levels of LDL were uniquely affected. Studies of familial hypercholesterolemia among individuals of predominantly Western European descent conformed to the perception that individuals with familial hypercholesterolemia tended to be lean and otherwise metabolically healthy. More recently, as we have studied familial hypercholesterolemia in more diverse global populations, we have learned that in some regions, rates of diabetes and obesity among familial hypercholesterolemia patients are very high, mirroring the global increases in the prevalence of metabolic disease. SUMMARY When diabetes and metabolic disease coexist, they amplify the cardiovascular risk in familial hypercholesterolemia, and may require more aggressive treatment.
Collapse
Affiliation(s)
- Liam R Brunham
- Centre for Heart Lung Innovation
- Department of Medicine
- Division of Cardiology
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
5
|
Lu F, Li E, Yang X. Proprotein convertase subtilisin/kexin type 9 deficiency in extrahepatic tissues: emerging considerations. Front Pharmacol 2024; 15:1413123. [PMID: 39139638 PMCID: PMC11319175 DOI: 10.3389/fphar.2024.1413123] [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: 04/06/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is primarily secreted by hepatocytes. PCSK9 is critical in liver low-density lipoprotein receptors (LDLRs) metabolism. In addition to its hepatocellular presence, PCSK9 has also been detected in cardiac, cerebral, islet, renal, adipose, and other tissues. Once perceived primarily as a "harmful factor," PCSK9 has been a focal point for the targeted inhibition of both systemic circulation and localized tissues to treat diseases. However, PCSK9 also contributes to the maintenance of normal physiological functions in numerous extrahepatic tissues, encompassing both LDLR-dependent and -independent pathways. Consequently, PCSK9 deficiency may harm extrahepatic tissues in close association with several pathophysiological processes, such as lipid accumulation, mitochondrial impairment, insulin resistance, and abnormal neural differentiation. This review encapsulates the beneficial effects of PCSK9 on the physiological processes and potential disorders arising from PCSK9 deficiency in extrahepatic tissues. This review also provides a comprehensive analysis of the disparities between experimental and clinical research findings regarding the potential harm associated with PCSK9 deficiency. The aim is to improve the current understanding of the diverse effects of PCSK9 inhibition.
Collapse
Affiliation(s)
- Fengyuan Lu
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - En Li
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Yang
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
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
|
7
|
Panagiotou S, Tan KW, Nguyen PM, Müller A, Oqua AI, Tomas A, Wendt A, Eliasson L, Tengholm A, Solimena M, Idevall-Hagren O. OSBP-mediated PI(4)P-cholesterol exchange at endoplasmic reticulum-secretory granule contact sites controls insulin secretion. Cell Rep 2024; 43:113992. [PMID: 38536815 DOI: 10.1016/j.celrep.2024.113992] [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: 10/04/2023] [Revised: 02/07/2024] [Accepted: 03/07/2024] [Indexed: 04/28/2024] Open
Abstract
Insulin is packaged into secretory granules that depart the Golgi and undergo a maturation process that involves changes in the protein and lipid composition of the granules. Here, we show that insulin secretory granules form physical contacts with the endoplasmic reticulum and that the lipid exchange protein oxysterol-binding protein (OSBP) is recruited to these sites in a Ca2+-dependent manner. OSBP binding to insulin granules is positively regulated by phosphatidylinositol-4 (PI4)-kinases and negatively regulated by the PI4 phosphate (PI(4)P) phosphatase Sac2. Loss of Sac2 results in excess accumulation of cholesterol on insulin granules that is normalized when OSBP expression is reduced, and both acute inhibition and small interfering RNA (siRNA)-mediated knockdown of OSBP suppress glucose-stimulated insulin secretion without affecting insulin production or intracellular Ca2+ signaling. In conclusion, we show that lipid exchange at endoplasmic reticulum (ER)-granule contact sites is involved in the exocytic process and propose that these contacts act as reaction centers with multimodal functions during insulin granule maturation.
Collapse
Affiliation(s)
| | - Kia Wee Tan
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Phuoc My Nguyen
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Andreas Müller
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Affiong Ika Oqua
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Anna Wendt
- Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Diabetes Center (LUDC), Lund, Sweden
| | - Lena Eliasson
- Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Diabetes Center (LUDC), Lund, Sweden
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Michele Solimena
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | |
Collapse
|
8
|
Sheng R, Li Y, Wu Y, Liu C, Wang W, Han X, Li Y, Lei L, Jiang X, Zhang Y, Zhang Y, Li S, Hong B, Liu C, Xu Y, Si S. A pan-PPAR agonist E17241 ameliorates hyperglycemia and diabetic dyslipidemia in KKAy mice via up-regulating ABCA1 in islet, liver, and white adipose tissue. Biomed Pharmacother 2024; 172:116220. [PMID: 38308968 DOI: 10.1016/j.biopha.2024.116220] [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: 09/13/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024] Open
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) is a common chronic metabolic disease. Peroxisome proliferator-activated receptors (PPARs) play crucial roles in regulating glucolipid metabolism. Previous studies showed that E17241 could ameliorate atherosclerosis and lower fasting blood glucose levels in ApoE-/- mice. In this work, we investigated the role of E17241 in glycolipid metabolism in diabetic KKAy mice. APPROACH AND RESULTS We confirmed that E17241 is a powerful pan-PPAR agonist with a potent agonistic activity on PPARγ, a high activity on PPARα, and a moderate activity on PPARδ. E17241 also significantly increased the protein expression of ATP-binding cassette transporter 1 (ABCA1), a crucial downstream target gene for PPARs. E17241 clearly lowered plasma glucose levels, improved OGTT and ITT, decreased islet cholesterol content, improved β-cell function, and promoted insulin secretion in KKAy mice. Moreover, E17241 could significantly lower plasma total cholesterol and triglyceride levels, reduce liver lipid deposition, and improve the adipocyte hypertrophy and the inflammatory response in epididymal white adipose tissue. Further mechanistic studies indicated that E17241 boosts cholesterol efflux and insulin secretion in an ABCA1 dependent manner. RNA-seq and qRT-PCR analysis demonstrated that E17241 induced different expression of PPAR target genes in liver and adipose tissue differently from the PPARγ agonist rosiglitazone. In addition, E17241 treatment was also demonstrated to have an exhilarating cardiorenal benefits. CONCLUSIONS Our results demonstrate that E17241 regulates glucolipid metabolism in KKAy diabetic mice while having cardiorenal benefits without inducing weight gain. It is a promising drug candidate for the treatment of T2DM.
Collapse
Affiliation(s)
- Ren Sheng
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Yining Li
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Yexiang Wu
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Chang Liu
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Weizhi Wang
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Xiaowan Han
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, CAMS & PUMC, Beijing 100050, China
| | - Yinghong Li
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Lijuan Lei
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Xinhai Jiang
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Yuyan Zhang
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Yuhao Zhang
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Shunwang Li
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Bin Hong
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China
| | - Chao Liu
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China.
| | - Yanni Xu
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China.
| | - Shuyi Si
- NHC Key Laboratory of Biotechnology for Microbial Drugs, National Center for Screening Novel Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Tiantan Xili 1#, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, CAMS & PUMC, Beijing 100050, China.
| |
Collapse
|
9
|
Tajadod S, Shekari S, Khalatbari Mohseni G, Abbasi K, Torki SA, Salimi Z, Keshavarz Mohammadian M, Shapouri M, Jarrahi SAM, Sobhani Far F, Shafaei H, Doaei S, YazdiMoghaddam H, Gholamalizadeh M. Association between type 2 diabetes and different types of dietary fats: A case-control study. Clin Nutr ESPEN 2023; 58:67-72. [PMID: 38057038 DOI: 10.1016/j.clnesp.2023.08.034] [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: 12/28/2022] [Revised: 08/14/2023] [Accepted: 08/30/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND The effect of dietary fats on type 2 diabetes (T2D) is not clear. This study aimed to determine the association between T2D and dietary fatty acids among Iranian adults. METHODS This case-control study was performed on 4241 participants aged 35-70, including 1804 people with T2D and pre-diabetes as the case group and 2437 people without diabetes as the control group. Dietary intake was assessed using a food frequency questionnaire (FFQ). RESULTS The cases had higher age (48.36 ± 8.62 vs. 54.53 ± 7.75 y, P < 0.001), weight (73.7713.41 vs. 76.18 ± 13.49 kg, P = 0.001), body mass index (BMI) (28.02 ± 4.70 vs. 24 ± 4.74 kg/m2, P = 0.001), right systolic blood pressure (RSBP) (113.33 ± 16.7 vs. 121.61 ± 17.24 mmHg, P = 0.001), right diastolic blood pressure (RDBP) (71.41 ± 10.53 vs. 75.33 ± 9.92 mmHg, P = 0.001), fasting blood sugar (FBS) (96.87 ± 19.39 vs. 169.95 ± 69.28 mg/dl, P = 0.001), blood urine nitrogen (BUN) (13.65 ± 3.74 vs. 14.26 ± 4.03 mg/dl, P = 0.001), triglyceride (TG) (141.61 ± 99.37 vs. 175.96 ± 114.74 mg/dl, P = 0.001), alkaline phosphatase (ALP) (218.24 ± 66.35 vs. 246.97 ± 72.65 IU/L, P = 0.001), low-density lipoprotein cholesterol (LDL) (111.68 ± 33.02 vs. 101.97 ± 36.54 mg/dl, P = 0.001), serum glutamic-pyruvic transaminase (SGPT) (21.88 ± 15.15 vs. 23.55 ± 15.96 IU/L, P = 0.001), gamma-glutamyl transferase (GGT) (24.66 ± 20.42 vs. 30.72 ± 30.43 IU/L P = 0.001), and cholesterol (192.45 ± 39.1190 vs. 187.12 ± 46.19 mg/dl P = 0.001) compared to the control group. T2D was negatively associated with dietary intake of PUFAs (OR = 0.93, CI95%:0.84-1.03, P = 0.01) and positively associated with dietary cholesterol (OR: 1.01, CI95%:1.001-1.01, P = 0.02). CONCLUSION In summary, cholesterol was positively and PUFAs were negatively associated with diabetes. If the results of the present study on the effect of fat intake on diabetes are proven, future dietary recommendations for people at risk of diabetes may be corrected by providing diets rich in polyunsaturated fatty acids and low in cholesterol.
Collapse
Affiliation(s)
- Shirin Tajadod
- Department of Nutrition, School Of Public Health, International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Soheila Shekari
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Golsa Khalatbari Mohseni
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Khadijeh Abbasi
- Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saheb Abbas Torki
- Department of Nutrition, Faculty of Nutrition Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Salimi
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Mahsa Shapouri
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Hanieh Shafaei
- Nursing and Midwifery School, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeid Doaei
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamideh YazdiMoghaddam
- Iranian Research Center on Healthy Aging, Operating Room Department, Faculty of Paramedics, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| | - Maryam Gholamalizadeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Laakso M, Fernandes Silva L. Statins and risk of type 2 diabetes: mechanism and clinical implications. Front Endocrinol (Lausanne) 2023; 14:1239335. [PMID: 37795366 PMCID: PMC10546337 DOI: 10.3389/fendo.2023.1239335] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/29/2023] [Indexed: 10/06/2023] Open
Abstract
Statins are widely used to prevent cardiovascular disease events. Cardiovascular diseases and type 2 diabetes are tightly connected since type 2 diabetes is a major risk factor for cardiovascular diseases. Additionally, cardiovascular diseases often precede the development of type 2 diabetes. These two diseases have common genetic and environmental antecedents. Statins are effective in the lowering of cardiovascular disease events. However, they have also important side effects, including an increased risk of type 2 diabetes. The first study reporting an association of statin treatment with the risk of type 2 diabetes was the WOSCOPS trial (West of Scotland Coronary Prevention Study) in 2001. Other primary and secondary cardiovascular disease prevention studies as well as population-based studies have confirmed original findings. The purpose of our review is to examine and summarize the most important findings of these studies as well as to describe the mechanisms how statins increase the risk of type 2 diabetes.
Collapse
Affiliation(s)
- Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Kuopio University Hospital, Kuopio, Finland
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
11
|
Graham A. Modulation of the Cellular microRNA Landscape: Contribution to the Protective Effects of High-Density Lipoproteins (HDL). BIOLOGY 2023; 12:1232. [PMID: 37759631 PMCID: PMC10526091 DOI: 10.3390/biology12091232] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
High-density lipoproteins (HDL) play an established role in protecting against cellular dysfunction in a variety of different disease contexts; however, harnessing this therapeutic potential has proved challenging due to the heterogeneous and relative instability of this lipoprotein and its variable cargo molecules. The purpose of this study is to examine the contribution of microRNA (miRNA; miR) sequences, either delivered directly or modulated endogenously, to these protective functions. This narrative review introduces the complex cargo carried by HDL, the protective functions associated with this lipoprotein, and the factors governing biogenesis, export and the uptake of microRNA. The possible mechanisms by which HDL can modulate the cellular miRNA landscape are considered, and the impact of key sequences modified by HDL is explored in diseases such as inflammation and immunity, wound healing, angiogenesis, dyslipidaemia, atherosclerosis and coronary heart disease, potentially offering new routes for therapeutic intervention.
Collapse
Affiliation(s)
- Annette Graham
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Cowcaddens Road, Glasgow G4 0BA, UK
| |
Collapse
|
12
|
Saruarov Y, Nuskabayeva G, Gencer MZ, Sadykova K, Zhunissova M, Tatykayeva U, Iskandirova E, Sarsenova G, Durmanova A, Gaipov A, Atageldiyeva K, Sarría-Santamera A. Associations of Clusters of Cardiovascular Risk Factors with Insulin Resistance and Β-Cell Functioning in a Working-Age Diabetic-Free Population in Kazakhstan. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3918. [PMID: 36900929 PMCID: PMC10001384 DOI: 10.3390/ijerph20053918] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
Cardiovascular risk factors aggregate in determined individuals. Patients with Type 2 diabetes mellitus (T2DM) have higher cardiovascular This study aimed to investigate insulinresistance (IR) and β-cell function using the homeostasis model assessment (HOMA) indexes in a general Kazakh population and determine the effect he effect that cardiovascular factors may have on those indexes. We conducted a cross-sectional study among employees of the Khoja Akhmet Yassawi International Kazakh-Turkish University (Turkistan, Kazakhstan) aged between 27 and 69 years. Sociodemographic variables, anthropometric measurements (body mass, height, waist circumference, hip circumference), and blood pressure were obtained. Fasting blood samples were collected to measure insulin, glucose, total cholesterol (TC), triglycerides (TG), and high- (HDL) andlow-density lipoprotein (LDL) levels. Oral glucose tolerance tests were performed. Hierarchical and K-means cluster analyses were obtained. The final sample was composed of 427 participants. Spearmen correlation analysis showed that cardiovascular parameters were statistically associated with HOMA-β (p < 0.001) and not with HOMA IR. Participants were aggregated into the three clusters where the cluster with a higher age and cardiovascular risk revealed deficient β-cell functioning, but not IR (p < 0.000 and p = 0.982). Common and easy to obtain biochemical and anthropometric measurements capturing relevant cardiovascular risk factors have been demonstrated to be associated with significant deficiency in insulin secretion. Although further longitudinal studies of the incidence of T2DM are needed, this study highlights that cardiovascular profiling has a significant role not just for risk stratification of patients for cardiovascular prevention but also for targeted vigilant glucose monitoring.
Collapse
Affiliation(s)
- Yerbolat Saruarov
- Department of Special Clinical Disciplines, Faculty of Medicine, Khoja Akhmet Yassawi International Kazakh-Turkish University, Turkistan 161200, Kazakhstan
| | - Gulnaz Nuskabayeva
- Department of Special Clinical Disciplines, Faculty of Medicine, Khoja Akhmet Yassawi International Kazakh-Turkish University, Turkistan 161200, Kazakhstan
| | - Mehmet Ziya Gencer
- Department of Special Clinical Disciplines, Faculty of Medicine, Khoja Akhmet Yassawi International Kazakh-Turkish University, Turkistan 161200, Kazakhstan
| | - Karlygash Sadykova
- Department of Special Clinical Disciplines, Faculty of Medicine, Khoja Akhmet Yassawi International Kazakh-Turkish University, Turkistan 161200, Kazakhstan
| | - Mira Zhunissova
- Department of Special Clinical Disciplines, Faculty of Medicine, Khoja Akhmet Yassawi International Kazakh-Turkish University, Turkistan 161200, Kazakhstan
| | - Ugilzhan Tatykayeva
- Department of Human Pathology and Physiology, Faculty of Dentistry, Khoja Akhmet Yassawi International Kazakh-Turkish University, Turkistan 161200, Kazakhstan
| | - Elmira Iskandirova
- Department of Therapy, Shymkent Medical Institute, Khoja Akhmet Yassawi International Kazakh-Turkish University, Shymkent 160019, Kazakhstan
| | - Gulmira Sarsenova
- Department of Therapy, Shymkent Medical Institute, Khoja Akhmet Yassawi International Kazakh-Turkish University, Shymkent 160019, Kazakhstan
| | - Aigul Durmanova
- Academic Department of Internal Medicine, University Medical Center, Astana 020000, Kazakhstan
| | - Abduzhappar Gaipov
- Academic Department of Internal Medicine, University Medical Center, Astana 020000, Kazakhstan
- Department of Medicine, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan
| | - Kuralay Atageldiyeva
- Academic Department of Internal Medicine, University Medical Center, Astana 020000, Kazakhstan
- Department of Medicine, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan
| | | |
Collapse
|
13
|
Galli A, Arunagiri A, Dule N, Castagna M, Marciani P, Perego C. Cholesterol Redistribution in Pancreatic β-Cells: A Flexible Path to Regulate Insulin Secretion. Biomolecules 2023; 13:224. [PMID: 36830593 PMCID: PMC9953638 DOI: 10.3390/biom13020224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Pancreatic β-cells, by secreting insulin, play a key role in the control of glucose homeostasis, and their dysfunction is the basis of diabetes development. The metabolic milieu created by high blood glucose and lipids is known to play a role in this process. In the last decades, cholesterol has attracted significant attention, not only because it critically controls β-cell function but also because it is the target of lipid-lowering therapies proposed for preventing the cardiovascular complications in diabetes. Despite the remarkable progress, understanding the molecular mechanisms responsible for cholesterol-mediated β-cell function remains an open and attractive area of investigation. Studies indicate that β-cells not only regulate the total cholesterol level but also its redistribution within organelles, a process mediated by vesicular and non-vesicular transport. The aim of this review is to summarize the most current view of how cholesterol homeostasis is maintained in pancreatic β-cells and to provide new insights on the mechanisms by which cholesterol is dynamically distributed among organelles to preserve their functionality. While cholesterol may affect virtually any activity of the β-cell, the intent of this review is to focus on early steps of insulin synthesis and secretion, an area still largely unexplored.
Collapse
Affiliation(s)
- Alessandra Galli
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università degli Studi di Milano, 20134 Milan, Italy
| | - Anoop Arunagiri
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MA 48106, USA
| | - Nevia Dule
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università degli Studi di Milano, 20134 Milan, Italy
| | - Michela Castagna
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università degli Studi di Milano, 20134 Milan, Italy
| | - Paola Marciani
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università degli Studi di Milano, 20134 Milan, Italy
| | - Carla Perego
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), Università degli Studi di Milano, 20134 Milan, Italy
| |
Collapse
|
14
|
Yang G, Schooling CM. Investigating sex-specific associations of lipid traits with type 2 diabetes, glycemic traits and sex hormones using Mendelian randomization. Cardiovasc Diabetol 2023; 22:3. [PMID: 36624450 PMCID: PMC9830908 DOI: 10.1186/s12933-022-01714-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 12/01/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Low-density lipoprotein (LDL)-cholesterol is positively associated with cardiovascular disease (CVD) and inversely associated with type 2 diabetes, which could detract from lipid modification. Here, we examined whether lipid traits potentially relevant to CVD aetiology, i.e. apolipoprotein B (apoB), triglycerides (TG) and lipoprotein(a) [Lp(a)] exhibited the same associations. We investigated sex-specifically, including the role of sex hormones, because sex disparities exist in lipid profile and type 2 diabetes. We also replicated where possible. METHODS We used Mendelian randomization (MR) to examine sex-specific associations of apoB, TG and Lp(a) with type 2 diabetes, HbA1c, fasting insulin, fasting glucose, testosterone and estradiol in the largest relevant sex-specific genome-wide association studies (GWAS) in people of European ancestry and replicated where possible. We also assessed sex-specific associations of liability to type 2 diabetes with apoB, TG and Lp(a). RESULTS Genetically predicted apoB and Lp(a) had little association with type 2 diabetes or glycemic traits in women or men. Genetically predicted higher TG was associated with higher type 2 diabetes risk [odds ratio (OR) 1.44 per standard deviation (SD), 95% confidence interval (CI) 1.26 to 1.65], HbA1c and fasting insulin specifically in women. Higher TG was associated with lower testosterone in women and higher testosterone in men, but with lower estradiol in men and women. Genetic liability to type 2 diabetes was associated with higher TG in women, and possibly with lower apoB in men. CONCLUSIONS Lipid traits potentially relevant to CVD aetiology do not exhibit contrasting associations with CVD and type 2 diabetes. However, higher TG is associated with higher type 2 diabetes risk and glycemic traits, which in turn further increases TG specifically in women, possibly driven by sex hormones.
Collapse
Affiliation(s)
- Guoyi Yang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - C Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Graduate School of Public Health and Health Policy, City University of New York, New York, USA.
| |
Collapse
|
15
|
Nezhadebrahimi A, Sepehri H, Jahanshahi M, Marjani M, Marjani A. The effect of simvastatin on gene expression of low-density lipoprotein receptor, sterol regulatory element-binding proteins, stearoyl-CoA desaturase 1 mRNA in rat hepatic tissues. Arch Physiol Biochem 2022; 128:1383-1390. [PMID: 32643419 DOI: 10.1080/13813455.2020.1772829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The study aimed to assess the effect of simvastatin on gene expression of LDLR, SREBPs, and SCD1 in rat hepatic tissues fed with high-fat diets (HFD) and its association with some biochemical parameters. Thirty-two male Wister albino rats were divided into four equal groups (three test and one control groups). The biochemical parameters were determined by using spectrophotometer techniques and the Elisa method. Low-density lipoprotein receptor, sterol regulatory element-binding proteins, stearoyl-CoA desaturase1, Beta-actin were analysed by real-time quantitative polymerase chain reaction (RT-PCR) method. At the end of study, the livers of the rats were separated and changes of hepatic tissue were determined. LDLR, SREBP2, and SCD1 expression increased significantly when compared G1 versus G4 and G2 versus G4. The expression of LDLR, SREBP2, and SCD1 also increased significantly when compared G2 versus G3, G1versus G3 and G1 versus G3 and G2 versus G3. The serum level of cholesterol, triglyceride, glucose, LDL, and HDL increased significantly when compared G1 versus G3. LDL showed significantly decreased when compared G1 versus G2. Cholesterol, glucose and HDL and triglyceride levels were increased significantly when compared G1 versus G4 and G2. Treatment of rats with HFD and simvastatin 20 mg/kg, triglyceride and LDL were almost the same as a control group and LDLR expression increased 98% in liver tissue. Gene expressions may be up-regulated in liver tissue and they showed different effects on biochemical parameters.
Collapse
Affiliation(s)
- Abbas Nezhadebrahimi
- Department of Biochemistry and Biophysics, Student Research Center, Metabolic Disorders Research Center, Gorgan Faculty of Medicine, Golestan University Medical Sciences, Gorgan, Iran
- Department of Physiology, Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Hamid Sepehri
- Department of Physiology, Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Marjani
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus, Turkey
| | - Abdoljalal Marjani
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University Medical Sciences, Gorgan, Iran
| |
Collapse
|
16
|
Yang L, Fye MA, Yang B, Tang Z, Zhang Y, Haigh S, Covington BA, Bracey K, Taraska JW, Kaverina I, Qu S, Chen W. Genome-wide CRISPR screen identified a role for commander complex mediated ITGB1 recycling in basal insulin secretion. Mol Metab 2022; 63:101541. [PMID: 35835371 PMCID: PMC9304790 DOI: 10.1016/j.molmet.2022.101541] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES Pancreatic beta cells secrete insulin postprandially and during fasting to maintain glucose homeostasis. Although glucose-stimulated insulin secretion (GSIS) has been extensively studied, much less is known about basal insulin secretion. Here, we performed a genome-wide CRISPR/Cas9 knockout screen to identify novel regulators of insulin secretion. METHODS To identify genes that cell autonomously regulate insulin secretion, we engineered a Cas9-expressing MIN6 subclone that permits irreversible fluorescence labeling of exocytic insulin granules. Using a fluorescence-activated cell sorting assay of exocytosis in low glucose and high glucose conditions in individual cells, we performed a genome-wide CRISPR/Cas9 knockout screen. RESULTS We identified several members of the COMMD family, a conserved family of proteins with central roles in intracellular membrane trafficking, as positive regulators of basal insulin secretion, but not GSIS. Mechanistically, we show that the Commander complex promotes insulin granules docking in basal state. This is mediated, at least in part, by its function in ITGB1 recycling. Defective ITGB1 recycling reduces its membrane distribution, the number of focal adhesions and cortical ELKS-containing complexes. CONCLUSIONS We demonstrated a previously unknown function of the Commander complex in basal insulin secretion. We showed that by ITGB1 recycling, Commander complex increases cortical adhesions, which enhances the assembly of the ELKS-containing complexes. The resulting increase in the number of insulin granules near the plasma membrane strengthens basal insulin secretion.
Collapse
Affiliation(s)
- Liu Yang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Margret A Fye
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Bingyuan Yang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Zihan Tang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Yue Zhang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sander Haigh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Brittney A Covington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kai Bracey
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Justin W Taraska
- Biochemistry and Biophysics Center, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Irina Kaverina
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
17
|
Pencina KM, Pencina MJ, Dufresne L, Holmes M, Thanassoulis G, Sniderman AD. An adverse lipoprotein phenotype-hypertriglyceridaemic hyperapolipoprotein B-and the long-term risk of type 2 diabetes: a prospective, longitudinal, observational cohort study. THE LANCET. HEALTHY LONGEVITY 2022; 3:e339-e346. [PMID: 36098309 DOI: 10.1016/s2666-7568(22)00079-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND This study examines the risk of new-onset diabetes in patients with hypertriglyceridaemic hyperapolipoprotein B (high triglycerides, high apolipoprotein B [apoB], low LDL cholesterol to apoB ratio, and low HDL cholesterol). The aim was to establish whether this lipoprotein phenotype identified a substantial group at high risk of developing diabetes over the next 20 years. METHODS In this prospective, longitudinal, observational cohort study, we used data from the Framingham Offspring cohort (recruited in Framingham, MA, USA). Participants were aged 40-69 years and free of diabetes and cardiovascular disease at a baseline examination done between April, 1987, and November, 1991, and were followed up until March, 2014. Cox proportional hazards regression with hierarchical adjustment for age and sex, waist circumference, and fasting blood glucose were used to model the relationship between each lipid marker and incident diabetes, as well as the relationship between hypertriglyceridaemic hyperapoB (defined as values greater than sample medians of triglycerides and apoB, and less than medians of HDL cholesterol and LDL cholesterol to apoB ratio) and incident diabetes. FINDINGS Of 3446 individuals aged 40-69 years who completed baseline examination, 2515 participants were eligible and included in all analyses. During median 21·1 years (IQR 11·1-23·1) of follow-up, 402 (16·0%) individuals developed diabetes. Age (p=0·032), waist circumference (p<0·0001), fasting blood glucose (p<0·0001), and natural logarithm-transformed triglycerides (p<0·0001) were associated with new-onset diabetes, as were apoB (p=0·0016), LDL cholesterol to apoB ratio (p=0·0018), and HDL cholesterol (p=0·0016) when added to this model. The age and sex-adjusted incidence of diabetes in the hypertriglyceridaemic hyperapoB group was 32·4% (95% CI 27·8-37·7) versus 5·5% (3·5-8·6) in the optimal lipid phenotype group and 15·5% (13·5-17·7) in the mixed lipid phenotype group. The fully adjusted hazard ratio, including glucose and waist circumference, for individuals with hypertriglyceridaemic hyperapoB was 3·30 (95% CI 2·06-5·30; p=0·0008) and for mixed lipid phenotype was 2·17 (1·38-3·40; p<0·0001) compared with those with the optimal lipid phenotype. INTERPRETATION Our findings suggest that individuals with hypertriglyceridaemic hyperapoB are at high risk of new-onset diabetes and might benefit from intensive measures to prevent diabetes. The association between this phenotype and incident diabetes is consistent with a pro-diabetic effect due to increased clearance of apoB particles from plasma, which could injure pancreatic islet cells. This mechanism might explain the increased risk of diabetes with statin therapy. FUNDING Doggone Foundation.
Collapse
Affiliation(s)
- Karol M Pencina
- Section on Men's Health, Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J Pencina
- Duke University School of Medicine, Biostatistics and Bioinformatics, Duke Clinical Research Institute, Durham, NC, USA
| | - Line Dufresne
- Mike and Valeria Rosenbloom Centre for Cardiovascular Prevention, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Michael Holmes
- MRC Population Health Research Unit at the University of Oxford, Oxford, UK; Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - George Thanassoulis
- Mike and Valeria Rosenbloom Centre for Cardiovascular Prevention, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Allan D Sniderman
- Mike and Valeria Rosenbloom Centre for Cardiovascular Prevention, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
18
|
Ghaffari T, Moradi N, Chamani E, Ebadi Z, Fadaei R, Alizadeh-Fanalou S, Yarahmadi S, Fallah S. Captopril and Spironolactone Can Attenuate Diabetic Nephropathy in Wistar Rats by Targeting ABCA1 and microRNA-33. Curr Pharm Des 2022; 28:1367-1372. [DOI: 10.2174/1381612828666220401143249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/24/2022] [Indexed: 11/22/2022]
Abstract
Background:
Nephropathy diabetes is one of the important causes of death and a more prevalent cause of end-stage renal disease.
Objective:
The present study investigated the effect of applying spironolactone and captopril and their combination on some renal performance indices and cholesterol-efflux-related gene expression in nephropathy diabetic rats.
Methods:
Intraperitoneal injection of streptozotocin was used to induce diabetes in rats. FBS, creatinine, and BUN were assayed using the calorimetry technique; also, urine microalbumin was assayed by ELISA. Hepatic gene expressions of ABCA1, ABCG1, and miR-33 were evaluated by the real-time PCR method.
Results:
FBS levels in the captopril-treated group were significantly decreased compared with the untreated diabetic group. BUN levels of treated groups with captopril and a combination of captopril + spironolactone were significantly increased. GFR of both treated diabetic groups with captopril and spironolactone was significantly lower than an untreated diabetic group. ABCA1 gene expression in hepatic cells of the combination of spironolactone + captopril treated group was significantly increased compared to other treated and untreated diabetic groups. The hepatic expression of the ABCG1 gene in the treated and untreated diabetic groups was significantly lower than in the control group. Treatment of the diabetic group with only combination therapy decreased the hepatic gene expression of miR-33 significantly.
Conclusion:
Obtained results suggest that S+C combination therapy can improve nephropathy and diabetes disorders by targeting the ABCA1 and miR-33 gene expression. It is suggested miR-33 and ABCA1 genes evaluation could be a new therapeutic strategy for nephropathy diabetes remediation.
Collapse
Affiliation(s)
- Tina Ghaffari
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| | - Nariman Moradi
- Department of Clinical Biochemistry, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Elham Chamani
- Department of Clinical Biochemistry, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Zahra Ebadi
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| | - Reza Fadaei
- Sleep Disorders Research Center, Research Institute for Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shahin Alizadeh-Fanalou
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| | - Sahar Yarahmadi
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| | - Soudabeh Fallah
- Department of Biochemistry and Nutrition, School of Medicine Iran University of Medical Sciences
| |
Collapse
|
19
|
Marouf BH, Iqbal Z, Mohamad JB, Bashir B, Schofield J, Syed A, Kilpatrick ES, Stefanutti C, Soran H. Efficacy and Safety of PCSK9 Monoclonal Antibodies in Patients With Diabetes. Clin Ther 2022; 44:331-348. [PMID: 35246337 DOI: 10.1016/j.clinthera.2021.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/28/2021] [Accepted: 12/09/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are novel drugs that have proven efficacy in improving cardiovascular outcomes. Roles for the PCSK9 molecule in metabolic pathways beyond LDL receptor processing and cholesterol homeostasis are well established. PCSK9 genetic variants associated with lower LDL-C levels correlate with a higher incidence of type 2 diabetes (T2DM), calling into question the appropriateness of these drugs in patients with T2DM and those at high risk of developing diabetes, and whether cardiovascular benefit seen with PCSK9 inhibitors might be offset by resultant dysglycemia. The purpose of this review was to examine the role of PCSK9 protein in glucose homeostasis, the impact of PCSK9 inhibition in relation to glucose homeostasis, and whether some of the cardiovascular benefit seen with PCSK9 inhibitors and statins might be offset by resultant dysglycemia. METHODS Comprehensive literature searches of electronic databases of PubMed, EMBASE, and OVID were conducted by using the search terms hyperlipidaemia, PCSK9, diabetes, and glucose as well as other relevant papers of interest collected by the authors. The retrieved papers were reviewed and shortlisted most relevant ones. FINDINGS Genetically determined lower circulating LDL-C and PCSK9 concentrations may have an incremental effect in increasing T2DM incidence, but any perceived harm is outweighed by the reduced risk of atherosclerotic cardiovascular disease achieved through lower lifetime exposure to LDL-C. PCSK9 monoclonal antibodies are effective and safe in patients with T2DM and those at high risk of developing it. The number-needed-to-treat to prevent one atherosclerotic cardiovascular disease event in the FOURIER (Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk) study in the subgroup with diabetes is significantly lower than for those without. Therefore, T2DM or being at high risk to develop it should not be a reason to avoid these agents. The safety of PCSK9 inhibition in relation to glucose homeostasis may depend on the method of inhibition and whether it occurs in circulation or the cells. Data from experimental studies and randomized controlled trials suggest no detrimental effect of PCSK9 monoclonal antibodies on glucose homeostasis. More data and large randomized controlled studies are needed to assess the impact of other methods of PCSK9 inhibition on glucose homeostasis. IMPLICATIONS PCSK9monoclonal antibodies markedly reduce LDL-C and consistently reduce cardiovascular mortality in patients with and without diabetes. Current evidence does not suggest an adverse effect of PCSK9 monoclonal antibodies on glycemic parameters.
Collapse
Affiliation(s)
- Bushra Hassan Marouf
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Sulaimani, Sulaimani, Federal Region of Kurdistan, Iraq
| | - Zohaib Iqbal
- Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Centre for Diabetes, Endocrinology and Metabolism, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Jamal Basheer Mohamad
- Department of Internal Medicine, College of Medicine, University of Duhok, Duhok, Federal Region of Kurdistan, Iraq
| | - Bilal Bashir
- Centre for Diabetes, Endocrinology and Metabolism, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Jonathan Schofield
- Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Centre for Diabetes, Endocrinology and Metabolism, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Akheel Syed
- Department of Diabetes, Endocrinology and Obesity Medicine, Salford Royal NHS Foundation and University Teaching Trust, Salford, United Kingdom
| | - Eric S Kilpatrick
- Department of Clinical Biochemistry, Manchester University NHS Foundation Trust, Manchester, and Hull York Medical School, Hull, United Kingdom
| | - Claudia Stefanutti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Handrean Soran
- Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Centre for Diabetes, Endocrinology and Metabolism, Manchester University NHS Foundation Trust, Manchester, United Kingdom.
| |
Collapse
|
20
|
Xepapadaki E, Nikdima I, Sagiadinou EC, Zvintzou E, Kypreos KE. HDL and type 2 diabetes: the chicken or the egg? Diabetologia 2021; 64:1917-1926. [PMID: 34255113 DOI: 10.1007/s00125-021-05509-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022]
Abstract
HDL is a complex macromolecular cluster of various components, such as apolipoproteins, enzymes and lipids. Quality evidence from clinical and epidemiological studies led to the principle that HDL-cholesterol (HDL-C) levels are inversely correlated with the risk of CHD. Nevertheless, the failure of many cholesteryl ester transfer protein inhibitors to protect against CVD casts doubts on this principle and highlights the fact that HDL functionality, as dictated by its proteome and lipidome, also plays an important role in protecting against metabolic disorders. Recent data indicate that HDL-C levels and HDL particle functionality are correlated with the pathogenesis and prognosis of type 2 diabetes mellitus, a major risk factor for CVD. Hyperglycaemia leads to reduced HDL-C levels and deteriorated HDL functionality, via various alterations in HDL particles' proteome and lipidome. In turn, reduced HDL-C levels and impaired HDL functionality impact the performance of key organs related to glucose homeostasis, such as pancreas and skeletal muscles. Interestingly, different structural alterations in HDL correlate with distinct metabolic abnormalities, as indicated by recent data evaluating the role of apolipoprotein A1 and lecithin-cholesterol acyltransferase deficiency in glucose homeostasis. While it is becoming evident that not all HDL disturbances are causatively associated with the development and progression of type 2 diabetes, a bidirectional correlation between these two conditions exists, leading to a perpetual self-feeding cycle.
Collapse
Affiliation(s)
- Eva Xepapadaki
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Rio Achaias, Greece
| | - Ioanna Nikdima
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Rio Achaias, Greece
| | - Eleftheria C Sagiadinou
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Rio Achaias, Greece
| | - Evangelia Zvintzou
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Rio Achaias, Greece
| | - Kyriakos E Kypreos
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Rio Achaias, Greece.
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| |
Collapse
|
21
|
Peyot ML, Roubtsova A, Lussier R, Chamberland A, Essalmani R, Murthy Madiraju SR, Seidah NG, Prentki M, Prat A. Substantial PCSK9 inactivation in β-cells does not modify glucose homeostasis or insulin secretion in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158968. [PMID: 33992809 DOI: 10.1016/j.bbalip.2021.158968] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 01/06/2023]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an important role in cholesterol homeostasis by promoting the degradation of the LDL receptor (LDLR). PCSK9 loss-of-function mutations are associated with increased fasting plasma glucose levels and slightly elevated risk of type 2-diabetes. Considering the known detrimental effects of cholesterol accumulation in β-cell, and the widespread use of PCSK9 inhibitors to treat hypercholesterolemia, it is important to gain insight into the role of pancreatic PCSK9 in glucose homeostasis and β-cell function. We generated the first β-cell-specific KO of PCSK9 (βKO). PCSK9 mRNA and protein expression were reduced by 48% and 78% in βKO islets, respectively, indicating that β-cells constitute a major site of PCSK9 expression. In islets, loss of β-cell PCSK9 resulted in unchanged LDLR protein levels, but reduced LDLR mRNA, indicating that cholesterol internalization is enhanced and that β-cell PCSK9 promotes LDLR degradation. In contrast, whole body PCSK9 KO mice exhibited 2-fold higher LDLR protein levels in islets and a stable expression of cholesterogenic genes. Whole body KO and βKO mice presented normal glucose tolerance, insulin release in response to glucose load and insulin sensitivity. Ex vivo glucose-stimulated insulin secretion in presence or absence of fatty acids was similar in WT and KO islets. Like KO mice, individuals carrying loss-of-function PCSK9 variants may be protected from cholesterol-induced toxicity due to reduced circulating cholesterol levels. Using both whole body KO or βKO models, our data demonstrate that PCSK9 deletion in mouse does not have any toxic effect on β-cell function and glucose homeostasis.
Collapse
Affiliation(s)
- Marie-Line Peyot
- Department of Nutrition, Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Anna Roubtsova
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada
| | - Roxane Lussier
- Department of Nutrition, Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Ann Chamberland
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada
| | - Rachid Essalmani
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada
| | - S R Murthy Madiraju
- Department of Nutrition, Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada
| | - Marc Prentki
- Department of Nutrition, Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal (IRCM), Montreal, Canada.
| |
Collapse
|
22
|
Li R, Sun X, Li P, Li W, Zhao L, Zhu L, Zhu S. GLP-1-Induced AMPK Activation Inhibits PARP-1 and Promotes LXR-Mediated ABCA1 Expression to Protect Pancreatic β-Cells Against Cholesterol-Induced Toxicity Through Cholesterol Efflux. Front Cell Dev Biol 2021; 9:646113. [PMID: 34307343 PMCID: PMC8292745 DOI: 10.3389/fcell.2021.646113] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 06/09/2021] [Indexed: 12/11/2022] Open
Abstract
T2DM (Type 2 diabetes) is a complex, chronic disease characterized as insulin resistance and islet β-cell dysfunction. Bariatric surgeries such as Roux-en-Y gastric bypass (RYGB) surgery and laparoscopic sleeve gastrectomy (LSG) have become part of a critical treatment regimen in the treatment of obesity and T2DM. Moreover, GLP-1 increase following bariatric surgery has been regarded as a significant event in bariatric surgery-induced remission of T2DM. In this study, a high concentration cholesterol-induced lipotoxicity was observed in INS-1 cells, including inhibited cell viability and insulin secretion. Enhanced cell apoptosis and inhibited cholesterol efflux from INS-1 cells; meanwhile, ABCA1 protein level was decreased by cholesterol stimulation. Cholesterol-induced toxicity and ABCA1 downregulation were attenuated by GLP-1 agonist EX-4. GLP-1 induced AMPK phosphorylation during the protection against cholesterol-induced toxicity. Under cholesterol stimulation, GLP-1-induced AMPK activation inhibited PARP-1 activity, therefore attenuating cholesterol-induced toxicity in INS-1 cells. In INS-1 cells, PARP-1 directly interacted with LXR, leading to the poly(ADP-ribosyl)ation of LXRα and downregulation of LXR-mediated ABCA1 expression. In the STZ-induced T2DM model in rats, RYGB surgery or EX-4 treatment improved the glucose metabolism and lipid metabolism in rats through GLP-1 inhibition of PARP-1 activity. In conclusion, GLP-1 inhibits PARP-1 to protect islet β cell function against cholesterol-induced toxicity in vitro and in vivo through enhancing cholesterol efflux. GLP-1-induced AMPK and LXR-mediated ABCA1 expression are involved in GLP-1 protective effects.
Collapse
Affiliation(s)
- Rao Li
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xulong Sun
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengzhou Li
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Weizheng Li
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lei Zhao
- Department of General Surgery, First Affiliated Hospital of University of South China, Hengyang, China
| | - Liyong Zhu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shaihong Zhu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
MicroRNA Sequences Modulated by Beta Cell Lipid Metabolism: Implications for Type 2 Diabetes Mellitus. BIOLOGY 2021; 10:biology10060534. [PMID: 34203703 PMCID: PMC8232095 DOI: 10.3390/biology10060534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/23/2022]
Abstract
Alterations in lipid metabolism within beta cells and islets contributes to dysfunction and apoptosis of beta cells, leading to loss of insulin secretion and the onset of type 2 diabetes. Over the last decade, there has been an explosion of interest in understanding the landscape of gene expression which influences beta cell function, including the importance of small non-coding microRNA sequences in this context. This review sought to identify the microRNA sequences regulated by metabolic challenges in beta cells and islets, their targets, highlight their function and assess their possible relevance as biomarkers of disease progression in diabetic individuals. Predictive analysis was used to explore networks of genes targeted by these microRNA sequences, which may offer new therapeutic strategies to protect beta cell function and delay the onset of type 2 diabetes.
Collapse
|
24
|
Richardson TG, Wang Q, Sanderson E, Mahajan A, McCarthy MI, Frayling TM, Ala-Korpela M, Sniderman A, Smith GD, Holmes MV. Effects of apolipoprotein B on lifespan and risks of major diseases including type 2 diabetes: a mendelian randomisation analysis using outcomes in first-degree relatives. THE LANCET. HEALTHY LONGEVITY 2021; 2:e317-e326. [PMID: 34729547 PMCID: PMC7611924 DOI: 10.1016/s2666-7568(21)00086-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Apolipoprotein B (apoB) is emerging as the crucial lipoprotein trait for the role of lipoprotein lipids in the aetiology of coronary heart disease. In this study, we evaluated the effects of genetically predicted apoB on outcomes in first-degree relatives. METHODS Data on lipoprotein lipids and disease outcomes in first-degree relatives were obtained from the UK Biobank study. We did a univariable mendelian randomisation analysis using a weighted genetic instrument for apoB. For outcomes with which apoB was associated at a false discovery rate (FDR) of less than 5%, multivariable mendelian randomisation analyses were done, including genetic instruments for LDL cholesterol and triglycerides. Associations between apoB and self-reported outcomes in first-degree relatives were characterised for 12 diseases (including heart disease, stroke, and hypertension) and parental vital status together with age at death. Estimates were inferred causal effects per 1 SD elevated lipoprotein trait (for apoB, 1 SD=0·24 g/L). Replication of estimates for lifespan and type 2 diabetes was done using conventional two-sample mendelian randomisation with summary estimates from genome-wide association study consortia. FINDINGS In univariable mendelian randomisation, genetically elevated apoB in participants was identified to lead to a shorter lifespan in parents (fathers: 0·89 years of life lost per 1 SD higher apoB in offspring, 95% CI 0·63-1·16, FDR-adjusted p=4·0 × 10-10; mothers: 0·48 years of life lost per 1 SD higher apoB in offspring, 0·25-0·71, FDR-adjusted p=1·7 × 10-4). The effects were strengthened to around 2 years of life lost in multivariable mendelian randomisation and were replicated in conventional two-sample mendelian randomisation (odds ratio [OR] of surviving to the 90th centile of lifespan: 0·38 per 1 SD higher apoB in offspring, 95% CI 0·22-0·65). Genetically elevated apoB caused higher risks of heart disease in all first-degree relatives and a higher risk of stroke in mothers. Findings in first-degree relatives were replicated in two-sample multivariable mendelian randomisation, which identified apoB to increase (OR 2·32 per 1 SD higher apoB, 95% CI 1·49-3·61) and LDL cholesterol to decrease (0·34 per 1 SD higher LDL cholesterol, 0·21-0·54) the risk of type 2 diabetes. INTERPRETATION Higher apoB shortens lifespan, increases risks of heart disease and stroke, and in multivariable analyses that account for LDL cholesterol, increases risk of diabetes. FUNDING British Heart Foundation, UK Medical Research Council, and UK Research and Innovation.
Collapse
Affiliation(s)
| | - Qin Wang
- Medical Research Council Integrative Epidemiology Unit; University of Bristol, Bristol, UK; Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health; University of Oxford, Oxford, UK; Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | | | | | | | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Mika Ala-Korpela
- Population Health Sciences, Bristol Medical School; Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland; Center for Life Course Health Research, University of Oulu, Oulu, Finland; NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Allan Sniderman
- Department of Medicine, McGill University, Montreal, QC, Canada
| | | | - Michael V Holmes
- Medical Research Council Integrative Epidemiology Unit; University of Bristol, Bristol, UK; Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health; Medical Research Council Population Health Research Unit
| |
Collapse
|
25
|
Huerta Guevara AP, McGowan SJ, Kazantzis M, Stallons TR, Sano T, Mulder NL, Jurdzinski A, van Dijk TH, Eggen BJL, Jonker JW, Niedernhofer LJ, Kruit JK. Increased insulin sensitivity and diminished pancreatic beta-cell function in DNA repair deficient Ercc1 d/- mice. Metabolism 2021; 117:154711. [PMID: 33493548 PMCID: PMC8625516 DOI: 10.1016/j.metabol.2021.154711] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/03/2021] [Accepted: 01/20/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Type 2 diabetes (T2DM) is an age-associated disease characterized by hyperglycemia due to insulin resistance and decreased beta-cell function. DNA damage accumulation has been associated with T2DM, but whether DNA damage plays a role in the pathogenesis of the disease is unclear. Here, we used mice deficient for the DNA excision-repair gene Ercc1 to study the impact of persistent endogenous DNA damage accumulation on energy metabolism, glucose homeostasis and beta-cell function. METHODS ERCC1-XPF is an endonuclease required for multiple DNA repair pathways and reduced expression of ERCC1-XPF causes accelerated accumulation of unrepaired endogenous DNA damage and accelerated aging in humans and mice. In this study, energy metabolism, glucose metabolism, beta-cell function and insulin sensitivity were studied in Ercc1d/- mice, which model a human progeroid syndrome. RESULTS Ercc1d/- mice displayed suppression of the somatotropic axis and altered energy metabolism. Insulin sensitivity was increased, whereas, plasma insulin levels were decreased in Ercc1d/- mice. Fasting induced hypoglycemia in Ercc1d/- mice, which was the result of increased glucose disposal. Ercc1d/- mice exhibit a significantly reduced beta-cell area, even compared to control mice of similar weight. Glucose-stimulated insulin secretion in vivo was decreased in Ercc1d/- mice. Islets isolated from Ercc1d/- mice showed increased DNA damage markers, decreased glucose-stimulated insulin secretion and increased susceptibility to apoptosis. CONCLUSION Spontaneous DNA damage accumulation triggers an adaptive response resulting in improved insulin sensitivity. Loss of DNA repair, however, does negatively impacts beta-cell survival and function in Ercc1d/- mice.
Collapse
Affiliation(s)
- Ana P Huerta Guevara
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Sara J McGowan
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA; Department of Metabolism and Aging, Scripps Research Institute, Jupiter, FL 33458, USA
| | | | | | - Tokio Sano
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, FL 33458, USA
| | - Niels L Mulder
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Angelika Jurdzinski
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Theo H van Dijk
- Laboratory Medicine, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Johan W Jonker
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church St., Minneapolis, MN 55455, USA; Department of Metabolism and Aging, Scripps Research Institute, Jupiter, FL 33458, USA
| | - Janine K Kruit
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands.
| |
Collapse
|
26
|
Protection against Glucolipotoxicity by High Density Lipoprotein in Human PANC-1 Hybrid 1.1B4 Pancreatic Beta Cells: The Role of microRNA. BIOLOGY 2021; 10:biology10030218. [PMID: 33805674 PMCID: PMC8000094 DOI: 10.3390/biology10030218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023]
Abstract
High-density lipoproteins provide protection against the damaging effects of glucolipotoxicity in beta cells, a factor which sustains insulin secretion and staves off onset of type 2 diabetes mellitus. This study examines epigenetic changes in small non-coding microRNA sequences induced by high density lipoproteins in a human hybrid beta cell model, and tests the impact of delivery of a single sequence in protecting against glucolipotoxicity. Human PANC-1.1B4 cells were used to establish Bmax and Kd for [3H]cholesterol efflux to high density lipoprotein, and minimum concentrations required to protect cell viability and reduce apoptosis to 30mM glucose and 0.25 mM palmitic acid. Microchip array identified the microRNA signature associated with high density lipoprotein treatment, and one sequence, hsa-miR-21-5p, modulated via delivery of a mimic and inhibitor. The results confirm that low concentrations of high-density lipoprotein can protect against glucolipotoxicity, and report the global microRNA profile associated with this lipoprotein; delivery of miR-21-5p mimic altered gene targets, similar to high density lipoprotein, but could not provide sufficient protection against glucolipotoxicity. We conclude that the complex profile of microRNA changes due to HDL treatment may be difficult to replicate using a single microRNA, findings which may inform current drug strategies focused on this approach.
Collapse
|
27
|
Statin Treatment-Induced Development of Type 2 Diabetes: From Clinical Evidence to Mechanistic Insights. Int J Mol Sci 2020; 21:ijms21134725. [PMID: 32630698 PMCID: PMC7369709 DOI: 10.3390/ijms21134725] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Statins are the gold-standard treatment for the prevention of primary and secondary cardiovascular disease, which is the leading cause of mortality worldwide. Despite the safety and relative tolerability of statins, observational studies, clinical trials and meta-analyses indicate an increased risk of developing new-onset type 2 diabetes mellitus (T2DM) after long-term statin treatment. It has been shown that statins can impair insulin sensitivity and secretion by pancreatic β-cells and increase insulin resistance in peripheral tissues. The mechanisms involved in these processes include, among others, impaired Ca2+ signaling in pancreatic β-cells, down-regulation of GLUT-4 in adipocytes and compromised insulin signaling. In addition, it has also been described that statins’ impact on epigenetics may also contribute to statin-induced T2DM via differential expression of microRNAs. This review focuses on the evidence and mechanisms by which statin therapy is associated with the development of T2DM. This review describes the multifactorial combination of effects that most likely contributes to the diabetogenic effects of statins. Clinically, these findings should encourage clinicians to consider diabetes monitoring in patients receiving statin therapy in order to ensure early diagnosis and appropriate management.
Collapse
|
28
|
Yalcinkaya M, Kerksiek A, Gebert K, Annema W, Sibler R, Radosavljevic S, Lütjohann D, Rohrer L, von Eckardstein A. HDL inhibits endoplasmic reticulum stress-induced apoptosis of pancreatic β-cells in vitro by activation of Smoothened. J Lipid Res 2020; 61:492-504. [PMID: 31907205 DOI: 10.1194/jlr.ra119000509] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/18/2019] [Indexed: 01/20/2023] Open
Abstract
Loss of pancreatic β-cell mass and function as a result of sustained ER stress is a core step in the pathogenesis of diabetes mellitus type 2. The complex control of β-cells and insulin production involves hedgehog (Hh) signaling pathways as well as cholesterol-mediated effects. In fact, data from studies in humans and animal models suggest that HDL protects against the development of diabetes through inhibition of ER stress and β-cell apoptosis. We investigated the mechanism by which HDL inhibits ER stress and apoptosis induced by thapsigargin, a sarco/ER Ca2+-ATPase inhibitor, in β-cells of a rat insulinoma cell line, INS1e. We further explored effects on the Hh signaling receptor Smoothened (SMO) with pharmacologic agonists and inhibitors. Interference with sterol synthesis or efflux enhanced β-cell apoptosis and abrogated the anti-apoptotic activity of HDL. During ER stress, HDL facilitated the efflux of specific oxysterols, including 24-hydroxycholesterol (OHC). Supplementation of reconstituted HDL with 24-OHC enhanced and, in cells lacking ABCG1 or the 24-OHC synthesizing enzyme CYP46A1, restored the protective activity of HDL. Inhibition of SMO countered the beneficial effects of HDL and also LDL, and SMO agonists decreased β-cell apoptosis in the absence of ABCG1 or CYP46A1. The translocation of the SMO-activated transcription factor glioma-associated oncogene GLI-1 was inhibited by ER stress but restored by both HDL and 24-OHC. In conclusion, the protective effect of HDL to counter ER stress and β-cell death involves the transport, generation, and mobilization of oxysterols for activation of the Hh signaling receptor SMO.
Collapse
Affiliation(s)
- Mustafa Yalcinkaya
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Katrin Gebert
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Wijtske Annema
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Rahel Sibler
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Silvija Radosavljevic
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
29
|
Ahamad J, Toufeeq I, Khan MA, Ameen MSM, Anwer ET, Uthirapathy S, Mir SR, Ahmad J. Oleuropein: A natural antioxidant molecule in the treatment of metabolic syndrome. Phytother Res 2019; 33:3112-3128. [DOI: 10.1002/ptr.6511] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/25/2019] [Accepted: 08/23/2019] [Indexed: 02/03/2023]
Affiliation(s)
- Javed Ahamad
- Faculty of PharmacyTishk International University Erbil Iraq
| | - Ibrahim Toufeeq
- Faculty of PharmacyTishk International University Erbil Iraq
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and ResearchJamia Hamdard New Delhi India
| | | | - Esra T. Anwer
- Faculty of PharmacyTishk International University Erbil Iraq
| | | | - Showkat R. Mir
- Department of Pharmacognosy, School of Pharmaceutical Education and Research (Formerly Faculty of Pharmacy)Jamia Hamdard New Delhi India
| | - Javed Ahmad
- Department of Pharmaceutics, College of PharmacyNajran University Najran Kingdom of Saudi Arabia
| |
Collapse
|
30
|
Fatahi S, Kord-Varkaneh H, Talaei S, Mardali F, Rahmani J, Ghaedi E, Tan SC, Shidfar F. Impact of phytosterol supplementation on plasma lipoprotein(a) and free fatty acid (FFA) concentrations: A systematic review and meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis 2019; 29:1168-1175. [PMID: 31582198 DOI: 10.1016/j.numecd.2019.07.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIM Although some earlier studies have indicated the effect of phytosterol (PS) supplementation on serum lipoprotein(a) (Lp(a)) and free fatty acid (FFA) concentration, findings are still conflicting. We aimed to assess the impact of PS supplementation on serum Lp(a) and FFA concentration through a systematic review and meta-analysis of available RCTs. METHODS AND RESULTS We performed a systematic search of all available RCTs conducted up to 21 February 2019 in the following databases: PubMed, Scopus, and Cochrane. The choice of fixed- or random-effect model for analysis was determined according to the I2 statistic. Effect sizes were expressed as weighted mean difference (WMD) and 95% confidence interval (CI). Pooling of 12 effect sizes from seven articles revealed a significant reduction of Lp(a) levels following PS supplementation (MD: -0.025 mg/dl, 95% CI: -0.045, -0.004, p = 0.017) without significant heterogeneity among the studies (I2 = 0.0%, p = 0.599). Also, PS supplementation significantly lowered FFA (MD: -0.138 mg/dl, 95% CI: -0.195, -0.081, p = 0.000) without significant heterogeneity among the studies (I2 = 0.0%, p = 0.911). The results for meta-regression and sensitivity analysis were not significant. CONCLUSION The meta-analysis suggests that oral PS supplementation could cause a significant reduction in serum Lp(a) and FFA.
Collapse
Affiliation(s)
- Somaye Fatahi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran; Student Research Committee, Faculty of Public Health Branch, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Kord-Varkaneh
- Student Research Committee, Department Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sam Talaei
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzane Mardali
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Jamal Rahmani
- Student Research Committee, Department Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Ghaedi
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shing C Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Affiliation(s)
- Maaike Kockx
- ANZAC Research Institute, Concord Repatriation General Hospital and University of Sydney, Sydney, Australia
| | - Leonard Kritharides
- ANZAC Research Institute, Concord Repatriation General Hospital and University of Sydney, Sydney, Australia.,Department of Cardiology, Concord Repatriation General Hospital and University of Sydney, Sydney, Australia
| |
Collapse
|
32
|
Perego C, Da Dalt L, Pirillo A, Galli A, Catapano AL, Norata GD. Cholesterol metabolism, pancreatic β-cell function and diabetes. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2149-2156. [DOI: 10.1016/j.bbadis.2019.04.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/11/2022]
|
33
|
Beigrezaei S, Ghiasvand R, Feizi A, Iraj B. Relationship between Dietary Patterns and Incidence of Type 2 Diabetes. Int J Prev Med 2019; 10:122. [PMID: 31367285 PMCID: PMC6639850 DOI: 10.4103/ijpvm.ijpvm_206_17] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 09/17/2017] [Indexed: 12/22/2022] Open
Abstract
Introduction Increasing rate of type 2 diabetes (T2D) prevalence during the recent years has caused concern about significant risks for the public health. Dietary patterns have recently attracted great attention in the evaluation of the relationship between diet and health. In the present study, we investigated the relationship between the major identified dietary patterns and T2D. Methods In this matched case-control study, 315 individuals (125 newly diagnosed cases and 190 controls); 18-60 years of age were selected. A valid semiquantitative food frequency questionnaire was used to collect dietary intakes of individuals. Anthropometric characteristics and blood pressure were measured with standard instructions and body mass index and waist to hip ratio were calculated. Factor analysis was used to identify major dietary patterns. The relationship between major food patterns and T2D was assessed by logistic regression analysis. Results Two dietary patterns were identified: healthy and Western dietary patterns. The second tertile of the healthy dietary pattern had significantly association with decreased risk of T2D in the crude model (Odds ratios [OR]: 0.51, 95% Confidence interval [CI]: 0.29-0.9; P for trend = 0.018), Model II (OR: 0.5, 95% CI: 0.27-0.9; P for trend = 0.019), and Model III (OR: 0.56, 95% CI: 0.23-1.4 P for trend = 0.048). The inverse association of the second tertile of Western dietary pattern score with the T2D was significant in crude (OR: 9.25, 95% CI: 4.95-17.4; P for trend <0.001) and multivariable-adjusted model (OR: 16.65, 95% CI: 2.99-92.84; P for trend <0001). Conclusions Our study found an inverse relationship between adherence of healthy pattern and direct association with Western dietary pattern and the risk of T2D.
Collapse
Affiliation(s)
- Sara Beigrezaei
- Department of Community Nutrition, School of Nutrition and Food Sciences, Isfahan, Iran
| | - Reza Ghiasvand
- Department of Community Nutrition, School of Nutrition and Food Sciences, Isfahan, Iran
| | - Awat Feizi
- Department of Biostatistics and Epidemiology, School of Health, Isfahan, Iran
| | - Bijan Iraj
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
34
|
Yang J, Lv Y, Zhao Z, Li W, Xiang S, Zhou L, Gao A, Yan B, Ou L, Ling H, Xiao X, Liu J. A microRNA‑24‑to‑secretagogin regulatory pathway mediates cholesterol‑induced inhibition of insulin secretion. Int J Mol Med 2019; 44:608-616. [PMID: 31173188 PMCID: PMC6605698 DOI: 10.3892/ijmm.2019.4224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 05/20/2019] [Indexed: 12/16/2022] Open
Abstract
Hypercholesterolemia is a key factor leading to β‑cell dysfunction, but its underlying mechanisms remain unclear. Secretagogin (Scgn), a Ca2+ sensor protein that is expressed at high levels in the islets, has been shown to play a key role in regulating insulin secretion through effects on the soluble N‑ethylmaleimide‑sensitive factor attachment receptor protein complexes. However, further studies are required to determine whether Scgn plays a role in hypercholesterolemia‑associated β‑cell dysfunction. The present study investigated the involvement of a microRNA‑24 (miR‑24)‑to‑Scgn regulatory pathway in cholesterol‑induced β‑cell dysfunction. In the present study, MIN6 cells were treated with increasing concentrations of cholesterol and then, the cellular functions and changes in the miR‑24‑to‑Scgn signal pathway were observed. Excessive uptake of cholesterol in MIN6 cells increased the expression of miR‑24, resulting in a reduction in Sp1 expression by directly targeting its 3' untranslated region. As a transcriptional activator of Scgn, downregulation of Sp1 decreased Scgn levels and subsequently decreased the phosphorylation of focal adhesion kinase and paxillin, which is regulated by Scgn. Therefore, the focal adhesions in insulin granules were impaired and insulin exocytosis was reduced. The present study concluded that a miR‑24‑to‑Scgn pathway participates in the mechanism regulating cholesterol accumulation‑induced β‑cell dysfunction.
Collapse
Affiliation(s)
- Jing Yang
- Department of Endocrinology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yuncheng Lv
- Laboratory of Clinical Anatomy and Reproductive Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhibo Zhao
- Department of Endocrinology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wu Li
- Department of Endocrinology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| | - Sunmin Xiang
- Department of Endocrinology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lingzhi Zhou
- Department of Paediatrics, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| | - Anbo Gao
- Laboratory of Clinical Anatomy and Reproductive Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Bin Yan
- Department of Endocrinology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lingling Ou
- Department of Endocrinology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong Ling
- Emergency Surgery, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xinhua Xiao
- Department of Endocrinology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jianghua Liu
- Department of Endocrinology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
35
|
Ma Z, Ketelhuth DFJ, Wirström T, Ohki T, Forteza MJ, Wang H, Grill V, Wollheim CB, Björklund A. Increased uptake of oxLDL does not exert lipotoxic effects in insulin-secreting cells. J Mol Endocrinol 2019; 62:159-168. [PMID: 30917339 DOI: 10.1530/jme-18-0146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/27/2019] [Indexed: 01/13/2023]
Abstract
Modified lipoproteins can negatively affect beta cell function and survival. However, the mechanisms behind interactions of modified lipoproteins with beta cells - and in particular, relationships to increased uptake - are only partly clarified. By over-expressing the scavenger receptor CD36 (Tet-on), we increased the uptake of fluorescent low-density modified lipoprotein (oxLDL) into insulin-secreting INS-1 cells. The magnitude of uptake followed the degree of CD36 over-expression. CD36 over-expression increased concomitant efflux of 3H-cholesterol in proportion to the cellular contents of 3H-cholesterol. Exposure to concentrations of oxLDL from 20 to 100 µg/mL dose-dependently increased toxicity (evaluated by MTT) as well as apoptosis. However, the increased uptake of oxLDL due to CD36 over-expression did not exert additive effects on oxLDL toxicity - neither on viability, nor on glucose-induced insulin release and cellular content. Reciprocally, blocking CD36 receptors by Sulfo-N-Succinimidyl Oleate decreased the uptake of oxLDL but did not diminish the toxicity. Pancreatic islets of CD36-/- mice displayed reduced uptake of 3H-cholesterol-labeled oxLDL vs wild type but similar toxicity to oxLDL. OxLDL was found to increase the expression of CD36 in islets and INS-1 cells. In summary, given the experimental conditions, our results indicate that (1) increased uptake of oxLDL is not responsible for toxicity of oxLDL, (2) increased efflux of the cholesterol moiety of oxLDL counterbalances, at least in part, increased uptake and (3) oxLDL participates in the regulation of CD36 in pancreatic islets and in INS-1 cells.
Collapse
Affiliation(s)
- Z Ma
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - D F J Ketelhuth
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - T Wirström
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - T Ohki
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - M J Forteza
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - H Wang
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - V Grill
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Institute of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - C B Wollheim
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - A Björklund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
Zhang X, van den Munckhof ICL, Rutten JHW, Netea MG, Groen AK, Zwinderman AH. Association of hemoglobin A1C with circulating metabolites in Dutch with European, African Surinamese and Ghanaian background. Nutr Diabetes 2019; 9:15. [PMID: 31040268 PMCID: PMC6491479 DOI: 10.1038/s41387-019-0082-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/28/2019] [Accepted: 04/11/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The prevalence of type 2 diabetes mellitus (T2DM) varies significantly across ethnic groups. A better understanding of the mechanisms underlying the variation in different ethnic groups may help to elucidate the pathophysiology of T2DM. The present work aims to generate a hypothesis regarding "why do subjects with African background have excess burden of T2DM?". METHODS In the current study, we performed metabolite profiling of plasma samples derived from 773 subjects of three ethnic groups (Dutch with European, Ghanaian and African Surinamese background). We performed Bayesian lognormal regression analyses to assess associations between HbA1c and circulating metabolites. RESULTS Here we show that subjects with African Surinamese and Ghanaian background had similar associations of HbA1c with circulating amino acids and triglyceride-rich lipoproteins as subjects with European background. In contrast, subjects with Ghanaian and African Surinamese background had different associations of HbA1c with acetoacetate, small LDL particle and small HDL particle concentrations, compared to the subjects with European background. CONCLUSIONS On the basis of the observations, we hypothesize that the excess burden of T2DM in subjects with African background may be due to impaired cholesterol efflux capacity or abnormal cholesterol uptake.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | - Joost H W Rutten
- Department of Internal Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud university medical Center, Nijmegen, The Netherlands
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Albert K Groen
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Aeilko H Zwinderman
- Clinical Epidemiology, Biostatistics, and Bioinformatics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Kim DW, Kim DH, Park JH, Choi M, Kim S, Kim H, Seul DE, Park SG, Jung JH, Han K, Park YG. Association between statin treatment and new-onset diabetes mellitus: a population based case-control study. Diabetol Metab Syndr 2019; 11:30. [PMID: 31044020 PMCID: PMC6477721 DOI: 10.1186/s13098-019-0427-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 04/16/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Several studies suggest that statin may increase the risk of new-onset diabetes mellitus (NODM). This study aimed to evaluate the association between the duration and recent use of statin, and the risk of NODM, based on population-based data sets. METHODS We used the South Korean National Health Insurance Service National Sample Cohort database for this nationwide case-control study. Of the 1 million participants, 6417 participants with NODM in 2012-2013 and 32,085 controls without diabetes (1:5 propensity score matched with age, sex, index year, and year of diabetes diagnosis) were included. In these patients, we examined the statin prescription record for 3 years preceding the outcome. We used conditional logistic regression to calculate the odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS After adjustment of covariates, there were no significant differences in the risk of NODM when analyzed according to cumulative use days. The risk of NODM was increased only in the short-term and recent user group (OR 1.48, 95% CI 1.21 to 1.82) whose cumulative prescription days are less than 6 months and whose last prescription is within 6 months of diagnosis. CONCLUSIONS The risk of NODM was not associated with an increase in the cumulative duration of statin use or with non-recent use. Only recent short-term use of statin was associated with an increased risk of NODM. Diabetes screening are warranted during initial statin therapy.
Collapse
Affiliation(s)
- Dong-Won Kim
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Republic of Korea
| | - Do-Hoon Kim
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Republic of Korea
| | - Joo-Hyun Park
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Republic of Korea
| | - Moonyoung Choi
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Republic of Korea
| | - Shinhye Kim
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Republic of Korea
| | - Hyonchong Kim
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Republic of Korea
| | - Da-eun Seul
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Republic of Korea
| | - Soo-Gyeong Park
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Republic of Korea
| | - Jin-Hyung Jung
- Department of Biostatistics, Catholic University College of Medicine, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Biostatistics, Catholic University College of Medicine, Seoul, Republic of Korea
| | - Yong-Gyu Park
- Department of Biostatistics, Catholic University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
38
|
Mahmoud AI, Galdos FX, Dinan KA, Jedrychowski MP, Davis JC, Vujic A, Rachmin I, Shigley C, Pancoast JR, Lee S, Hollister-Lock J, MacGillivray CM, Gygi SP, Melton DA, Weir GC, Lee RT. Apolipoprotein E is a pancreatic extracellular factor that maintains mature β-cell gene expression. PLoS One 2018; 13:e0204595. [PMID: 30303984 PMCID: PMC6179231 DOI: 10.1371/journal.pone.0204595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023] Open
Abstract
The in vivo microenvironment of tissues provides myriad unique signals to cells. Thus, following isolation, many cell types change in culture, often preserving some but not all of their in vivo characteristics in culture. At least some of the in vivo microenvironment may be mimicked by providing specific cues to cultured cells. Here, we show that after isolation and during maintenance in culture, adherent rat islets reduce expression of key β-cell transcription factors necessary for β-cell function and that soluble pancreatic decellularized matrix (DCM) can enhance β-cell gene expression. Following chromatographic fractionation of pancreatic DCM, we performed proteomics to identify soluble factors that can maintain β-cell stability and function. We identified Apolipoprotein E (ApoE) as an extracellular protein that significantly increased the expression of key β-cell genes. The ApoE effect on beta cells was mediated at least in part through the JAK/STAT signaling pathway. Together, these results reveal a role for ApoE as an extracellular factor that can maintain the mature β-cell gene expression profile.
Collapse
Affiliation(s)
- Ahmed I. Mahmoud
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Francisco X. Galdos
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Katherine A. Dinan
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Mark P. Jedrychowski
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States of America
| | - Jeffrey C. Davis
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Ana Vujic
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Inbal Rachmin
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Christian Shigley
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - James R. Pancoast
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Samuel Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Jennifer Hollister-Lock
- Islet Cell and Regenerative Biology Section, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, United States of America
| | - Catherine M. MacGillivray
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States of America
| | - Douglas A. Melton
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
| | - Gordon C. Weir
- Islet Cell and Regenerative Biology Section, Joslin Diabetes Center, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, United States of America
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
39
|
Yu Q, Wang F, Meng X, Gong Y, Wang Y, Xu C, Wang S. Short‑term use of atorvastatin affects glucose homeostasis and suppresses the expression of LDL receptors in the pancreas of mice. Mol Med Rep 2018; 18:2780-2788. [PMID: 30015940 PMCID: PMC6102652 DOI: 10.3892/mmr.2018.9239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/03/2018] [Indexed: 01/30/2023] Open
Abstract
Low-density lipoprotein receptors (LDLRs) may serve a role in the diabetogenic effect of statins; however, the effects of statins on LDLR expression and its regulation in the pancreas and islets have yet to be determined. To exclude the long-term effects of treatment with atorvastatin, which allows mice to adapt, male C57BL/j and apolipoprotein E-deficient mice were acutely treated with oral atorvastatin for 6 weeks, and glucose homeostasis and LDLR expression in the pancreas and islets were examined. In the present study, it was observed that the short-term use of atorvastatin affected insulin sensitivity in normal mice and glucose tolerance in hyperlipidemic mice. Furthermore, it was identified that 6 weeks of treatment with atorvastatin suppressed LDLR expression in the pancreas and pancreatic islets in C57BL/j mice, and an increase in proprotein convertase subtilisin/kexin type 9 expression was additionally observed in the pancreas. However, 6 weeks of treatment with atorvastatin did not affect LDLR expression in the pancreas of hyperlipidemic mice. It may be concluded that the short-term use of atorvastatin disturbs glucose homeostasis and suppresses LDLR expression in the pancreas and pancreatic islets in C57BL/j mice, suggesting that the role of LDLR in the diabetogenic effect of statins requires further investigation.
Collapse
Affiliation(s)
- Qi Yu
- Institute of Material Medical, School of Pharmacy, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Fang Wang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Xiaodong Meng
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Yiren Gong
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Yanli Wang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Cangbao Xu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Siwang Wang
- Institute of Material Medical, School of Pharmacy, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
40
|
Zanoni P, Velagapudi S, Yalcinkaya M, Rohrer L, von Eckardstein A. Endocytosis of lipoproteins. Atherosclerosis 2018; 275:273-295. [PMID: 29980055 DOI: 10.1016/j.atherosclerosis.2018.06.881] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/04/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
During their metabolism, all lipoproteins undergo endocytosis, either to be degraded intracellularly, for example in hepatocytes or macrophages, or to be re-secreted, for example in the course of transcytosis by endothelial cells. Moreover, there are several examples of internalized lipoproteins sequestered intracellularly, possibly to exert intracellular functions, for example the cytolysis of trypanosoma. Endocytosis and the subsequent intracellular itinerary of lipoproteins hence are key areas for understanding the regulation of plasma lipid levels as well as the biological functions of lipoproteins. Indeed, the identification of the low-density lipoprotein (LDL)-receptor and the unraveling of its transcriptional regulation led to the elucidation of familial hypercholesterolemia as well as to the development of statins, the most successful therapeutics for lowering of cholesterol levels and risk of atherosclerotic cardiovascular diseases. Novel limiting factors of intracellular trafficking of LDL and the LDL receptor continue to be discovered and to provide drug targets such as PCSK9. Surprisingly, the receptors mediating endocytosis of high-density lipoproteins or lipoprotein(a) are still a matter of controversy or even new discovery. Finally, the receptors and mechanisms, which mediate the uptake of lipoproteins into non-degrading intracellular itineraries for re-secretion (transcytosis, retroendocytosis), storage, or execution of intracellular functions, are largely unknown.
Collapse
Affiliation(s)
- Paolo Zanoni
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Srividya Velagapudi
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Mustafa Yalcinkaya
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Lucia Rohrer
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
41
|
Baghdasarian S, Lin HP, Pickering RT, Mott MM, Singer MR, Bradlee ML, Moore LL. Dietary Cholesterol Intake Is Not Associated with Risk of Type 2 Diabetes in the Framingham Offspring Study. Nutrients 2018; 10:E665. [PMID: 29794966 PMCID: PMC6024792 DOI: 10.3390/nu10060665] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/15/2018] [Accepted: 05/18/2018] [Indexed: 02/01/2023] Open
Abstract
Identification of diet and lifestyle risk factors for prevention of type 2 diabetes mellitus (T2DM) is of great importance. The specific role of dietary cholesterol (DC) in T2DM risk is unclear. This study uses data from 2192 Framingham Offspring Study subjects to estimate the effects of DC alone and in combination with markers of a healthy diet and other lifestyle factors on fasting glucose and risk of T2DM or impaired fasting glucose (IFG) over 20 years of follow-up. Dietary data were derived from two sets of three-day food records. Statistical methods included mixed linear regression and Cox proportional hazard's modeling to adjust for confounding. There were no statistically significant differences in glucose levels over 20 years of follow-up across DC intake categories (.
Collapse
Affiliation(s)
- Siyouneh Baghdasarian
- Department of Medicine Section of Preventative Medicine and Epidemiology, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Hsuan-Ping Lin
- Department of Medicine Section of Preventative Medicine and Epidemiology, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Richard T Pickering
- Department of Medicine Section of Preventative Medicine and Epidemiology, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Melanie M Mott
- Department of Medicine Section of Preventative Medicine and Epidemiology, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Martha R Singer
- Department of Medicine Section of Preventative Medicine and Epidemiology, Boston University School of Medicine, Boston, MA 02118, USA.
| | - M Loring Bradlee
- Department of Medicine Section of Preventative Medicine and Epidemiology, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Lynn L Moore
- Department of Medicine Section of Preventative Medicine and Epidemiology, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Recent studies have demonstrated a higher risk of incident diabetes associated with statin use, causing concern among patients and clinicians. In this review, we will assess the evidence and proposed mechanisms behind statin therapy and its association with incident diabetes. We will then review the current recommendations for statin use in light of this association and suggest next steps for clinicians managing these patients and researchers exploring this phenomenon. RECENT FINDINGS The annual risk of developing new-onset diabetes with statin treatment is approximately 0.1%. In comparison, the absolute risk reduction of major coronary events with statin use is approximately 0.42% annually. Statins are associated with the development of incident diabetes, particularly among those with predisposing risk factors for diabetes. However, the benefit of statin use among these patients in preventing major coronary events strongly favors statin use despite its risk of incident diabetes.
Collapse
|
43
|
Tricò D, Trifirò S, Mengozzi A, Morgantini C, Baldi S, Mari A, Natali A. Reducing Cholesterol and Fat Intake Improves Glucose Tolerance by Enhancing β Cell Function in Nondiabetic Subjects. J Clin Endocrinol Metab 2018; 103:622-631. [PMID: 29095990 DOI: 10.1210/jc.2017-02089] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/23/2017] [Indexed: 12/22/2022]
Abstract
CONTEXT A diet low in cholesterol and fat is commonly recommended to prevent metabolic and cardiovascular diseases; however, its effect on glucose tolerance is largely unknown. OBJECTIVE We examined whether and by which mechanisms a chronic reduction of cholesterol and fat intake affects glucose tolerance in nondiabetic individuals, independently of weight changes. DESIGN AND PARTICIPANTS In this crossover, randomized clinical trial, 30 healthy subjects, including 15 with family history of type 2 diabetes (T2D) (T2D offspring), underwent a 75-g oral glucose tolerance test (OGTT) after two 14-day isocaloric high-cholesterol, high-fat (HChF) or low-cholesterol, and low-fat (LChF) diets. MAIN OUTCOME MEASURES We evaluated changes in glucose tolerance, β cell function, insulin clearance, and insulin sensitivity by modeling plasma glucose, insulin, and C-peptide levels during the OGTT. RESULTS The shift from the HChF to the LChF diet was neutral on body weight but increased glucose tolerance (mean glucose -5%, P = 0.01) and three components of β cell function: glucose sensitivity (+17%, P = 0.01), insulin secretion at fasting glucose (+20%, P = 0.02), and potentiation (+19%, P = 0.03). The LChF diet improved insulin sensitivity (+7%, P = 0.048) only in T2D offspring, who tended to be more susceptible to the positive effect of the diet on glucose tolerance. CONCLUSIONS A chronic and isocaloric decrease in dietary cholesterol and fat intake improves glucose tolerance by diffusely ameliorating β cell function in nondiabetic subjects. Individuals genetically predisposed to develop T2D tend to be more susceptible to the positive effect of this dietary intervention on glucose tolerance and insulin sensitivity.
Collapse
Affiliation(s)
- Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Silvia Trifirò
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Cecilia Morgantini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simona Baldi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
44
|
Ross S, Gerstein H, Paré G. The Genetic Link Between Diabetes and Atherosclerosis. Can J Cardiol 2018; 34:565-574. [PMID: 29731020 DOI: 10.1016/j.cjca.2018.01.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 02/06/2023] Open
Abstract
Epidemiological studies have indicated that the risk of atherothrombotic coronary artery disease (CAD) is higher in patients with diabetes, but these results have not been consistently observed across clinical trials. To address this apparent discrepancy, we can apply the results of genome-wide association studies (GWAS) to provide a better understanding of the shared genetic architecture of diabetes and atherothrombotic CAD. For instance, a large GWAS has identified 16 novel loci that are associated with both diabetes and atherothrombotic CAD. These genetic variants may also be used to assess potential causal relationships reported in observational studies and clinical trials through Mendelian randomization analyses. For example, several Mendelian randomization analyses have shown that diabetes is associated with CAD independent of other risk factors (odds ratio [OR]: 1.63, 95% confidence interval [CI]: 1.23-2.07; P = 0.002). Furthermore, Mendelian randomization analyses can provide more insight into the perceived risk of diabetes among patients without diabetes receiving statin therapy. Here, genetically lower activity of HMG-CoA reductase (HMGCR) was associated with a modest increase in diabetes (OR per allele: 1.02, 95% CI: 1.00-1.05). These results highlight the biological mechanisms that link diabetes with the use of statins. In addition, this work illustrates the great potential value of genetic studies to clarify the mechanistic relationships among atherosclerotic vascular disease, dysglycemia, and diabetes. More research is needed to delineate and subsequently better understand the genetic links between diabetes and atherosclerosis.
Collapse
Affiliation(s)
- Stephanie Ross
- Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - Hertzel Gerstein
- Department of Medicine and Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, Ontario, Canada; Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Canada.
| |
Collapse
|
45
|
Low-Carbohydrate, High-Protein, High-Fat Diets Rich in Livestock, Poultry and Their Products Predict Impending Risk of Type 2 Diabetes in Chinese Individuals that Exceed Their Calculated Caloric Requirement. Nutrients 2018; 10:nu10010077. [PMID: 29329254 PMCID: PMC5793305 DOI: 10.3390/nu10010077] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/18/2017] [Accepted: 01/08/2018] [Indexed: 01/04/2023] Open
Abstract
The evidence on the association between long-term low-carbohydrate, high-fat and high-protein diets and type 2 diabetes (T2D) is controversial. Until now, data is limited for Chinese populations, especially in considering the influence of extra energy intake. In this paper, we aimed to investigate the association of low-carbohydrate, high-fat and high-protein diets with type 2 diabetes (T2D) risk in populations consuming extra calories and those with normal caloric intake, We also determined whether the association is mediated by insulin resistance (IR) or β-cell dysfunction. A total of 3644 subjects in the Harbin People’s Health Study (Cohort 1, 2008–2012) and 7111 subjects in the Harbin Cohort Study on Diet, Nutrition and Chronic Non-Communicable Diseases (Cohort 2, 2010–2015) were analyzed, with a median follow-up of 4.2 and 5.3 years, respectively. Multivariate relative risks (RRs) and their 95% confidence intervals (95% CIs) were calculated to estimate the association between low-carbohydrate, high-fat and high-protein diet and T2D in logistic regression models. The multivariate RRs (95% CIs) were 1.00, 2.24 (1.07, 4.72) and 2.29 (1.07, 4.88) (Ptrend = 0.04), and 1.00, 1.45 (0.91, 2.31) and 1.64 (1.03, 2.61) (Ptrend = 0.04) across tertiles of low-carbohydrate, high-fat and high-protein diet scores in the population consuming extra calories in Cohort 1 and Cohort 2, respectively. The association was no longer significant after adjustment for livestock and its products, or poultry and its products. The mediation analysis discovered that this association in the population consuming extra calories was insulin resistance mediated, in both Cohort 1 and Cohort 2. However, the association was not significant among participants overall and participants with normal caloric intake. Our results indicated that long-term low-carbohydrate, high-fat and high-protein diets were associated with increased T2D risk among the population consuming extra calories, which may be caused by higher intake of animal-origin fat and protein as well as lower intake of vegetables, fruit and fiber. Additionally, the association was mediated by IR. In the population consuming extra calories, reducing the intake of livestock, poultry and their products and increasing the intake of vegetables, fruit and fiber might protect this population from developing T2D.
Collapse
|
46
|
Yao Y, Xu Y, Wang W, Zhang J, Li Q. Glucagon-like peptide-1 improves β-cell dysfunction by suppressing the miR-27a-induced downregulation of ATP-binding cassette transporter A1. Biomed Pharmacother 2017; 96:497-502. [DOI: 10.1016/j.biopha.2017.10.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/21/2017] [Accepted: 10/09/2017] [Indexed: 02/08/2023] Open
|
47
|
Femlak M, Gluba-Brzózka A, Ciałkowska-Rysz A, Rysz J. The role and function of HDL in patients with diabetes mellitus and the related cardiovascular risk. Lipids Health Dis 2017; 16:207. [PMID: 29084567 PMCID: PMC5663054 DOI: 10.1186/s12944-017-0594-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/16/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a major public health problem which prevalence is constantly raising, particularly in low- and middle-income countries. Both diabetes mellitus types (DMT1 and DMT2) are associated with high risk of developing chronic complications, such as retinopathy, nephropathy, neuropathy, endothelial dysfunction, and atherosclerosis. METHODS This is a review of available articles concerning HDL subfractions profile in diabetes mellitus and the related cardiovascular risk. In this review, HDL dysfunction in diabetes, the impact of HDL alterations on the risk diabetes development as well as the association between disturbed HDL particle in DM and cardiovascular risk is discussed. RESULTS Changes in the amount of circulation lipids, including triglycerides and LDL cholesterol as well as the HDL are frequent also in the course of DMT1 and DMT2. In normal state HDL exerts various antiatherogenic properties, including reverse cholesterol transport, antioxidative and anti-inflammatory capacities. However, it has been suggested that in pathological state HDL becomes "dysfunctional" which means that relative composition of lipids and proteins in HDL, as well as enzymatic activities associated to HDL, such as paraoxonase 1 (PON1) and lipoprotein-associated phospholipase 11 (Lp-PLA2) are altered. HDL properties are compromised in patients with diabetes mellitus (DM), due to oxidative modification and glycation of the HDL protein as well as the transformation of the HDL proteome into a proinflammatory protein. Numerous studies confirm that the ability of HDL to suppress inflammatory signals is significantly reduced in this group of patients. However, the exact underlying mechanisms remains to be unravelled in vivo. CONCLUSIONS The understanding of pathological mechanisms underlying HDL dysfunction may enable the development of therapies targeted at specific subpopulations and focusing at the diminishing of cardiovascular risk.
Collapse
Affiliation(s)
- Marek Femlak
- 105 Military Hospital with Outpatient Clinic in Żary, Domańskiego 2, 68-200, Żary, Poland
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, WAM Teaching Hospital of Lodz, Żeromskiego 113, Łódź, 90-549, Poland.
| | | | - Jacek Rysz
- Department of Nephrology Hypertension and Family Medicine, Medical University of Lodz, Żeromskiego 113, Łódź, 90-549, Poland
| |
Collapse
|
48
|
Climent E, Pérez-Calahorra S, Marco-Benedí V, Plana N, Sánchez R, Ros E, Ascaso JF, Puzo J, Almagro F, Lahoz C, Civeira F, Pedro-Botet J. Effect of LDL cholesterol, statins and presence of mutations on the prevalence of type 2 diabetes in heterozygous familial hypercholesterolemia. Sci Rep 2017; 7:5596. [PMID: 28717233 PMCID: PMC5514105 DOI: 10.1038/s41598-017-06101-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/07/2017] [Indexed: 11/09/2022] Open
Abstract
Patients with heterozygous familial hypercholesterolemia (HeFH) have been reported to be less vulnerable to type 2 diabetes mellitus (T2DM), although the mechanism is unknown. The aims of the present study were to assess the effects of low density lipoprotein (LDL) cholesterol concentration and the presence of FH-causing mutations on T2DM prevalence in HeFH. Data were collected from the Dyslipidemia Registry of the Spanish Arteriosclerosis Society. Inclusion criteria were definite or probable HeFH in patients aged ≥18 years. T2DM prevalence in HeFH patients was compared with data of the general population. 1732 patients were included. The prevalence of T2DM was lower in patients with HeFH compared with the general population (5.94% vs 9.44%; OR: 0.606, 95% CI 0.486-0.755, p < 0.001). Risk factors for developing T2DM were male sex, age, body mass index, hypertension, baseline triglyceride levels and years on statin therapy. The prevalence of T2DM in HeFH patients was 40% lower than that observed in the general population. Gene mutations and LDL cholesterol concentrations were not risk factors associated with the prevalence of T2DM in patients with HeFH. The prevalence of T2DM in patients with HeFH was 40% lower than in the general population matched for age and sex.
Collapse
Affiliation(s)
- Elisenda Climent
- Lipid and Vascular Unit, Department of Endocrinology and Nutrition, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sofía Pérez-Calahorra
- Lipid Unit. Hospital Universitario Miguel Servet, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Victoria Marco-Benedí
- Lipid Unit. Hospital Universitario Miguel Servet, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Nuria Plana
- Unitat de Medicina Vascular i Metabolisme, Hospital Universitari Sant Joan, Institut d´Investigació Sanitaria Pere Virgili (IISPV), Reus, Tarragona, Spain
| | - Rosa Sánchez
- Lipid Unit, Servicio de Endocrinología y Nutrición, Hospital Universitario Insular de Gran Canarias, Instituto Universitario de Investigaciones Biomédicas y Sanitarias de la Universidad de Las Palmas de Gran Canarias, Las Palmas, Spain
| | - Emilio Ros
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d'Investigacions Biomèdiques August Pi Sunyer, Hospital Clínic, Barcelona and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Juan F Ascaso
- Servicio de Endocrinología y Nutrición, Hospital Clínico Universitario, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Universitat de Valencia, Valencia, Spain
| | - Jose Puzo
- Lipid Unit. Hospital San Jorge, Huesca, Spain
| | | | - Carlos Lahoz
- Atherosclerosis Unit, Internal Medicine Department, Hospital Carlos III, Madrid, Spain
| | - Fernando Civeira
- Lipid Unit. Hospital Universitario Miguel Servet, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Juan Pedro-Botet
- Lipid and Vascular Unit, Department of Endocrinology and Nutrition, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
49
|
Yu Q, Chen Y, Xu CB. Statins and New-Onset Diabetes Mellitus: LDL Receptor May Provide a Key Link. Front Pharmacol 2017; 8:372. [PMID: 28659805 PMCID: PMC5468445 DOI: 10.3389/fphar.2017.00372] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 05/30/2017] [Indexed: 12/31/2022] Open
Abstract
Numerous studies have noted that populations treated with statins have increased risk for new-onset diabetes mellitus; however, the underlying molecular mechanisms are not fully understood. Interestingly, familial hypercholesterolemia (FH) patients with mutations in the low-density lipoprotein receptor (LDLR) gene are protected against diabetes mellitus (DM), despite these patients being subjected to long-term statin therapy. Since the common pathway between FH and statin therapy is LDLR-mediated cellular cholesterol uptake, the arising question is whether the LDLR plays an important role in the diabetogenic effect of statins. Indeed, given that statins can regulate the LDLR expression in liver and peripheral tissue, there is a possible mechanism that the increased LDLR causes cellular cholesterol accumulation and dysfunction in pancreatic islets, explaining why statins fail to increase the risk of DM in FH patients. In this paper, with regarded to recent literatures, we highlight the role of LDLR in the pathophysiology of cholesterol-induced pancreatic islets dysfunction, which may provide the key link between statins treatment and the increased risk of new-onset diabetes mellitus.
Collapse
Affiliation(s)
- Qi Yu
- Institute of Basic and Translational Medicine, Xi'an Medical UniversityXi'an, China.,Shaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an, China.,Institute of Material Medical, School of Pharmacy, The Fourth Military Medical UniversityXi'an, China
| | - Ying Chen
- Institute of Basic and Translational Medicine, Xi'an Medical UniversityXi'an, China.,Department of Information and Communication Engineering, Xi'an Jiaotong UniversityXi'an, China
| | - Cang-Bao Xu
- Institute of Basic and Translational Medicine, Xi'an Medical UniversityXi'an, China.,Shaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an, China
| |
Collapse
|
50
|
Olsson AG, Angelin B, Assmann G, Binder CJ, Björkhem I, Cedazo-Minguez A, Cohen J, von Eckardstein A, Farinaro E, Müller-Wieland D, Parhofer KG, Parini P, Rosenson RS, Starup-Linde J, Tikkanen MJ, Yvan-Charvet L. Can LDL cholesterol be too low? Possible risks of extremely low levels. J Intern Med 2017; 281:534-553. [PMID: 28295777 DOI: 10.1111/joim.12614] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Following the continuous accumulation of evidence supporting the beneficial role of reducing low-density lipoprotein cholesterol (LDL-C) levels in the treatment and prevention of atherosclerotic cardiovascular disease and its complications, therapeutic possibilities now exist to lower LDL-C to very low levels, similar to or even lower than those seen in newborns and nonhuman species. In addition to the important task of evaluating potential side effects of such treatments, the question arises whether extremely low LDL-C levels per se may provoke adverse effects in humans. In this review, we summarize information from studies of human cellular and organ physiology, phenotypic characterization of rare genetic diseases of lipid metabolism, and experience from clinical trials. Specifically, we emphasize the importance of the robustness of the regulatory systems that maintain balanced fluxes and levels of cholesterol at both cellular and organismal levels. Even at extremely low LDL-C levels, critical capacities of steroid hormone and bile acid production are preserved, and the presence of a cholesterol blood-brain barrier protects cells in the central nervous system. Apparent relationships sometimes reported between less pronounced low LDL-C levels and disease states such as cancer, depression, infectious disease and others can generally be explained as secondary phenomena. Drug-related side effects including an increased propensity for development of type 2 diabetes occur during statin treatment, whilst further evaluation of more potent LDL-lowering treatments such as PCSK9 inhibitors is needed. Experience from the recently reported and ongoing large event-driven trials are of great interest, and further evaluation including careful analysis of cognitive functions will be important.
Collapse
Affiliation(s)
- A G Olsson
- Department of Medicine and Health, Linköping University, Linköping, Sweden
| | - B Angelin
- Metabolism Unit, Department of Endocrinology, Metabolism and Diabetes, and KI/AZ Integrated CardioMetabolic Center, Department of Medicine, Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - G Assmann
- University of Münster, Münster, Germany
| | - C J Binder
- Medical University of Vienna & Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - I Björkhem
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - A Cedazo-Minguez
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet Huddinge, Stockholm, Sweden
| | - J Cohen
- UT Southwestern Medical Center, Dallas, TX, USA
| | | | | | - D Müller-Wieland
- Klinik II und Poliklinik für Innere Medizin der Universität zu Köln, Köln, Germany
| | - K G Parhofer
- Ludwig-Maximilians-University of Munich, Munich, Germany
| | - P Parini
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | | | | |
Collapse
|