Diabetes Research and Therapy-Sci Forschen

Full Text

Short Commentary
Diet and Nutrition Reverse Type 3 Diabetes and Accelerated Aging Linked to Global Chronic Diseases

  Ian James Martins1,2,3*      Ana María Calderón de la Barca1   

1Centre of Excellence in Alzheimer’s Disease Research and Care School of Medical Sciences, Edith Cowan University, Australia
2School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Nedlands, Australia
3McCusker Alzheimer’s Research Foundation, Hollywood Medical Centre, Nedlands, 6009, Australia

*Corresponding author: Ian James Martins, School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia 6027, Australia, Tel: +61863042574; E-mail: i.martins@ecu.edu.au


Abstract

The acceleration in the rate of chronic diseases that involve insulin resistance has become of global concern. The rate of the most prevalent chronic disease such as cardiovascular disease is linked to the metabolic syndrome, non alcoholic fatty liver disease (NAFLD) and other chronic diseases that include obesity, diabetes and neurodegenerative diseases. The gene-environment interaction in Western countries indicates that with urbanization access to food and its content may lead to induction of epigenetic alterations and identify the gene Sirtuin 1 (Sirt 1) to be responsible for the increased risk for insulin resistance and NAFLD relevant to Type 1, Type 2 and Type 3 diabetes in these countries. Nutrigenomics is linked to neuron and liver telomere maintenance, cell division and tissue growth and has become important with essential nutrients that regulate Sirt 1 function important to prevent NAFLD in individuals with diabetes. Nutrigenomic diets, exercise, drugs and lifestyle changes regulate Type 3 diabetes with neuron Sirt 1 transcriptional responses associated with DNA modifications that regulate brain insulin resistance relevant to NAFLD and diabetes.

Keywords

Nutrigenomics; Obesity; Diabetes; Accelerated Aging; Endocrine; Chronic Diseases; Diet

Background

In Western countries and third world countries the global obesity epidemic has been reported to effect at least 10% of the global population with projected health care costs to obesity related medical expenses reported to be 344 billion dollars to the year 2018 and may account for 21% of health costs in the United States. The acceleration in the rate of chronic diseases that involve insulin resistance has also become of global concern. The rate of the most prevalent chronic disease such as cardiovascular disease [1-4] is linked to the metabolic syndrome and non alcoholic fatty liver disease (NAFLD) and environmental factors such as stress, anxiety and depression are important to consider with the global increase in other chronic diseases that include diabetes and neurodegenerative diseases.

In 2013 the world health organization (WHO 2013) indicated that the number of global deaths (63%) included cardiovascular disease (48%), cancer (21%) and chronic respiratory conditions (12%). Nutritional and anti-aging therapies may prevent accelerated aging involved in the global obesity epidemic that currently involves various endocrine and chronic diseases in Western populations. Obesity and diabetes are defined as endocrine disorders with the medical conditions such as hyperinsulinemia involved with hormonal imbalance with various inflammatory complications of organs such as the liver, brain, thyroid, parathyroid, adrenal gland and pancreas [5,6]. Stress, fatigue, anxiety and depression are closely linked to chronic diseases and the molecular mechanisms that involve the apelinergic pathway have become important to Type 3 diabetes and neuroendocrine disease with theinduction of insulin resistance that lead to obesity and diabetes [7-12]. Metabolic diseases such as cardiovascular disease and neurodegeneration are associated with accelerated aging process and involve alteration in blood lipids such as cholesterol and triglyceride with a decrease in high density lipoprotein (HDL) [13-16]. Anxiety disorders effect mental health and induce changes in the brain associated with plasma hormone dysregulation and alterations in the biological clock especially in tissues such as the liver, adipose tissue and brain.

The gene-environment interaction in Western countries indicates that with urbanization [17] and access to food and its content may lead to induction of epigenetic alterations that are associated with lipid and glucose dyshomeostasis with increased risk for insulin resistance relevant to Type 1, Type 2 and Type 3 diabetes and obesity in these countries. Diets that contain various organic pollutants with overnutrition lead to abnormal xenobiotic metabolism [18-22] with marked effects on DNA strand breakage with cell apoptosis. High calorie diets, exercise and lifestyle changes regulate transcriptional responses with DNA modifications that include DNA methylation, histone tails, chromatin and micro RNA (short ribonucleic acid gene regulators)that regulate DNA expression and promote chronic disease susceptibility (Figure1). The gene environment interaction now identify the nuclear receptor Sirtuin 1 (Sirt 1)that regulates appetite [23] to be involved in the induction of insulin resistance and Type 3 diabetes and involve alterations in nuclear receptors, micro RNA with chromatin remodelling [24] that are now closely associated with obesity, diabetes and neurodegenerative disease.

Sirtuin 1 contributes to the Post-Transcriptional Dysregulation in Type 1, Type 2 and Type 3 Diabetes

Nutritional research has concentrated on the identification of nutrient sensing diets that provide regulation of histone deacetylases (HDAC) that are involved in epigenetic control of gene expression that controls metabolic and tissue glucose and cholesterol homeostasis [7,14,17]. Sirt 1 is a NAD+ dependent protein deacetylase and is involved in the deacetylation of the nuclear receptors (Figure 1) with its critical involvement in insulin resistance [25]. The anti-aging protein Sirt 1 and cell senescence has been closely linked to telomere biology and global DNA repair which provides mechanistic explanations for Sirt 1 functions in the protection of DNA damage, and thus genomic stability [26,27] with relevance to Type 1, Type 2 and Type 3 diabetes. Anti-aging strategies that target telomere shortening and brain glucose dysfunction in obesity and diabetes are of particular interest to biological aging since telomere shortening has been associated as an early risk for dementia and Type 3 diabetes.

In calorie restriction regulation of the Sirt 1 gene involved in lifespan and aging has now become essential therapy for glucose and cholesterol maintenance with reversal of chronic diseases such as obesity, diabetes and neurodegenerative diseases in global communities [24]. Considerable interest in chronic diseases of the central nervous system that include neurodegenerative diseases such as Parkinson’s disease (PD) and Alzheimer’s disease (AD) may provide a useful model for the prevention and management of various chronic diseases. Risk factors such as diet and lifestyle indicate that Sirt 1 dysregulation is important to liver and metabolic health [24] and central to Type 3 diabetes since Sirt 1 is involved in neuron loss [28] associated with insulin resistance and neurodegenerative disease.

In Figure 2 a summary is shown of the relevance of Sirt 1 in the genetic regulation of diabetes. HLA class I, II genes and genes of the HLA region of chromosome 6 have also been shown to be involved greatly with the risk for Type 1 diabetes (HLA-DR, DQ and DP). In caucasian populations contribution by DQA1and DQB1 haplotypes, linkages to DRB1*03 and DRB1*04 haplotypes have been associated with Type 1 diabetes. Other ethnic groups such as African, Americans, Japanese and Chinese specific haplotypes have also been associated with Type 1 diabetes [29]. An interest in global chronic diseases has identified Sirt 1 dysregulation to involve Type 1 diabetes with nutritional regulation of Sirt 1 important to assist with glucose homeostasis in these Type 1individuals. Sirt 1 regulation of the MODY gene via transcription factors hepatocyte nuclear factor 1(HNF-1) and HNF-4 or HNF1/HNF4 complex [30-32] has been shown with evidence of Sirt1/HNF-4genetic regulation of liver and pancreas in Type 1 diabetes.

In Type 2 diabetes more than 150 genetic loci are associated with the development of diabetes and 50 candidate genes have shown to play a major part in the development of the disease and include genes such as peroxisome proliferator-activated receptors, ATP-binding cassette transporter sub-family C member 8, KATP mutations and Calpain 10 [29]. These genes are involved in pancreatic β cell function, insulin action and glucose metabolism in metabolic conditions (Figure 2). In Type 2 diabetes the relevance of stress to defective apelinergic pathways that involve the pancreas, liver, kidney and brain have been identified [7] with severity of diabetes associated with poor insulin actions and glucose regulation. Sirt 1 plays an important role in the regulation of the apelinergic pathway relevant to Type 2 diabetes with connections to brain insulin resistance (stroke, dementia, AD) and Type 3 diabetes [7]. Individuals with Type 1 and Type 2 diabetes involve early and accelerated organ diseases of the brain when associated with Type 3 diabetes and the liver with Sirt 1 repression closely involved with insulin resistance in these individuals.

A single gene effect associated with Sirt 1 repression versus multiple diabetic genes effect may indicate either the interaction of the diabetic individual is sensitive to gene-environments events that involve diet and lifestyle changes. Cellular miRNA have become important since they may be regulated by diet, drugs, xenobiotics, stress and anxiety (environment) that may be relevant to the stress sensitive Sirt 1 [7]. Nutritional intervention is now critical early in life to maintain normal Sirt 1/ fibroblast growth factor 1 regulation of circadian rhythm that maintain brain-liver pathways and glucose homeostasis [32,33]. Sirt 1 regulation targets various transcription factors peroxisome proliferator-activated receptor-gamma co-activator (PGC-1 alpha), p53 tumour suppressor protein, pregnane x receptor (PXR) to adapt gene expression to metabolic activity, insulin resistance and inflammation [24]. Effects of feeding on Sirt 1and p53 interactions are involved in nuclear-mitochondria interactions, mutations, cell death (apoptosis) or permanent cellular senescence [24] with relevance to Sirt 1 deacetylation of p53 that determine DNA repair critical for maintenance of cell glucose homeostasis [34,35].

Figure 1: High calorie diets, stress, exercise and lifestyle changes regulate transcriptional responses with DNA modifications that regulate neuron micro RNA and DNA expression and promote chronic diseases associated with obesity, diabetes (Type 1,2,3) and neurodegenerative disease. The gene environment interaction indicates that the stress sensitive anti-aging gene Sirtuin 1 is downregulated with effects on drug therapy linked to the induction of Type 3 diabetes.

Figure 2: The genetic regulation of Type 1 and Type 2 diabetes is associated with the gene expression of Sirt 1 in global populations. A single gene effect associated with the repression of the anti-aging gene Sirt 1 versus multiple diabetic genes may indicate the interaction of the diabetic individual to gene-environments events such as unhealthy diets and stress that involves Sirt 1 dysregulation of glucose and cholesterol in the brain and peripheral tissues.

Effects of p53 on gene regulators include micro RNA (miRNAs) [36,37] and their role in the induction of obesity [38] and diabetes [39-41] indicates altered expression of multiple miRNAs in metabolic tissues [42] Furthermore miRNAs such as miR-34a [43] and miR-122, miR-132 [44,45] that directly inhibit Sirt 1 are associated with poor activation of hepatic genes involved in glucose and lipid metabolism [46] with an increase in acetylated p53 involved with cell apoptosis and NAFLD [47,48]. The p53 effects on miR-34a transactivation involve Sirt1 expression associated with insulin resistance and the development of metabolic disease [49,50].

Nutrigenomics diets maintain Drug and Insulin Therapy with the Prevention of Type 3 Diabetes

Interest in the nature of food intake has increased since the nature of fat consumption (low or high) may require further evaluation and may contain xenobiotics with marked effects on neuronal apoptosis and neuroendocrine disease [17].The effects from environments in developing countries (urban inhabitants) contain xenobiotics (soil, water, air) that may contribute to Type 3 diabetes. High fibre diets [51] that contain fruit and vegetables have become important for the treatment of NAFLD with the reduction in the absorption of lipophilic xenobitics with the prevention of Type 3 diabetes. Activation of hepatic nuclear receptors such as the Sirt1/pregnane X receptor (PXR) by calorie restriction (low glycemic index/fatty acid consumption) are important to the science of nutrigenomics [17] with relevance to activation of hepatic xenobiotic metabolism connected to the prevention of neuronal apoptosis.

The consumption of fruits such as apples that contain pyruvic acid (450 mg/apple) an antioxidant and an activator of Sirt1 [52-54] is essential for diabetes treatment. Other brain nutrients to prevent Type 3 diabetes to maintain brain glucose homeostasis include phosphatidyl inositol and leucine both important for the maintenance of Sirt 1 related neuron function. Exercise and heavy work activities may rapidly deplete brain pyruvic acid, leucine and phosphatidyl inositol with the acceleration of brain insulin resistance [55]. Excessive ingestion of vegetables not more than (1-2 gm/day) may cause suprachiasmatic disorders with accumulation of brain phytosterols with aging [51]. The nature of phytosterols that regulate liver cholesterol metabolism are critical to prevent and reverse NAFLD with HDL cholesterol metabolism closely linked to phytosterol ingestion [51]. Specific polyphenols found in vegetables and fruits need careful evaluation since high doses may cause increased oxidative stress with toxicity to the liver and induction of NAFLD and chronic disease [13]. Nutritional science that involves the maintenance of Sirt 1 regulation of DNA repair require effective function of neuronal telomeres and ingestion of therapeutic foods is essential to prevent Type 3 diabetes [29]. Nutrigenomic diets have become important to prevent Sirt 1 inhibition with diets rich in palmitic acid and alcohol discouraged and both are inhibitors of Sirt 1 [29]. Butyric acid is a histone deacetylase inhibitor used in the treatment of diet induced insulin resistance [56,57] with doses required to reduce amyloid beta protein aggregation in AD but butyric acid may completely inhibit the Sirt 1 deacetylase activity essential for cell telomere growth and mitochondria survival [24].

Sirt 1 is essential for maintenance of insulin therapy in diabetes and aging [58-60]. In diabetes interest in the hepatic drug transport has accelerated with toxic effects of drugs on insulin therapy. More than 600 drugs (eg: Asprin, Warfarin, Furosemide, Atrorvastatin, Clopidogrel, Levothyroxin Acetaminophen, Cholecalciferol, Simvastatin) have been listed that corrupt insulin therapy with drug-drug interactions involved in diabetes [61]. Interests in nutrigenomic diets and drug transport indicate the importance of Sirt 1 in brain to liver drug transport pathways via the blood brain barrier [62-66] and require hepatic Sirt 1 regulation of glucose, drug, cholesterol and bile acid metabolism [24]. Diabetes and hepatic and brain amyloid beta metabolism [29] are connected to drug metabolism (Figure 3) and defective hepatic drug metabolism may be the primary defect in diabetes with acceleration of neuronal apoptosis by toxic brain amyloid beta metabolism secondary to Type 3 complications associated with Type 1 or Type 2 diabetic disease [29].

Figure 3: Unhealthy diets such as high fat diets downregulate hepatic drug metabolism with the corruption of insulin therapy in diabetes and aging. Amyloid beta metabolism in the brain and liver are connected to drug metabolism and defective hepatic drug metabolism may be the primary defect in Type 3 diabetes with acceleration of neuronal apoptosis by abnormal brain amyloid beta metabolism associated with Type 1 or Type 2 diabetes. Abbreviations: P-gp1, P glycoprotein, PXR, pregnane X receptor, CAR, constitutive androstane receptor, ABCA1, ATP binding cassette transporter 1, BBB, blood brain barrier, LDLr, low density lipoprotein receptor, LRP, LDL receptor related protein.

In recent finding approximately 40 miRNAs have been shown to be dysregulated [67-69] in diabetic individuals. The identification of specific dietary supplements that may regulate DNA or microRNA metabolism has become important with nutritional interventions essential for rapid reversal for the severity of diabetes. Diets that do not contain therapeutic food supplements promote Sirt 1 downregulation and disturb p53/mi RNA metabolism [70] with relevance to toxic effects of drugs on insulin therapy in diabetes. Several miRNA have been shown to effect drug metabolism with nuclear receptors involved in the regulation of drug and xenobiotic metabolism [71]. Elevated xenobiotic cell levels have been shown to interfere with miRNA expression [72] with relevance to nuclear Sirt 1 activity.

Nutrigenomic diets activate Sirt 1 and accelerate the hepatic metabolism of toxins such as bacterial lipopolysaccharides (LPS) and mycotoxins that inhibit Sirt 1 and promote insulin resistance with relevance to brain, liver and pancreatic disease [73]. Nutrigenomic diets that do not contain LPS have become important since LPS effects on membrane asymmetry [74,75] involve membrane calcium levels/flux that determine severity of insulin resistance, abnormal drug transport with amyloid beta dyshomeostasis [13] connected to Type 3 diabetes.

Conclusion

Nutritional interventions have become important for the reversal of Type 3 diabetes associated with accelerated aging and linked to Type 1 and Type 2 diabetes. Therapeutic foods that contain anti-aging dietary components activate the anti-aging gene Sirt 1 with relevance to cell glucose dyshomeostasis linked to heart, pancreas, liver and brain diseases. Nutrigenomic diets that activate hepatic Sirt 1 improve drug metabolism with relevance to insulin therapy in diabetes. Lifestyle changes such as drug, environment, diet and stress are involved with the metabolism of therapeutic nutrients that are depleted from the brain with extended workload/exercise treatment periods associated with the downregulation of brain nuclear receptors with relevance to Type 3 diabetes and the severity of insulin resistance in various global communities.

Acknowledgements

This work was supported by grants from Edith Cowan University, the McCusker Alzheimer’s Research Foundation and the National Health and Medical Research Council.

References
  1. Greenberg H, Raymond, SU, Leeder SR (2005) Cardiovascular disease and global health: threat and opportunity. Health Aff (Millwood). [Ref.]
  2. Bukhman G, Kidder A (2008) Cardiovascular disease and global health equity: lessons from tuberculosis control then and now. Am J Public Health 98: 44-54. [Ref.]
  3. Mathew B, Francis L, Kayalar A, Cone J (2008) Obesity: effects on cardiovascular disease and its diagnosis. J Am Board Fam Med 21: 562-568. [Ref.]
  4. Lavie CJ, Mehra, MR, Milani RV (2005) Obesity and heart failure prognosis: paradox or reverse epidemiology?. Eur Heart J 26: 5-7. [Ref.]
  5. James PT, Rigby N, Leach R (2004) The obesity epidemic, metabolic syndrome and future prevention strategies. Eur J Cardiovasc Prev Rehabil 11: 3-8. [Ref.]
  6. Flegal KM, Kit BK, Orpana H, Graubard BI (2013) Association of allcause mortality with overweight and obesity using standard body mass index categories: a systematic review and meta-analysis. JAMA 309: 71-82. [Ref.]
  7. Martins IJ (2015) Nutritional diets accelerate amyloid beta metabolism and prevent the induction of chronic diseases and Alzheimer’s disease. Photon ebooks. UBN: 015-A94510112017. [Ref.]
  8. Jeanrenaud B, Halimi S, Van der Werve G (2009) Neuroendocrine disorders seen as triggers of the triad: Obesity-insulin resistanceabnormal glucose tolerance. Diabetes Metab Rev 1: 261-291. [Ref.]
  9. Susaki E, Nakayama KI (2010) An animal model manifesting neurodegeneration and obesity. Aging (Albany NY) 2: 453-456. [Ref.]
  10. Bjorntorp P (1999) Neuroendocrine perturbations as a cause of insulin resistance. Diabetes Metab Res Rev 15: 427-441. [Ref.]
  11. Bjorntorp P (1995) Insulin resistance: the consequence of a neuroendocrine disturbance?. Int J Obes Relat Metab Disord 19: S6- S10. [Ref.]
  12. Sjostrand M, Eriksson JW (2009) Neuroendocrine mechanisms in insulin resistance. Mol Cell Endocrinol 297: 104-111. [Ref.]
  13. Martins IJ, Creegan R (2014) Links between Insulin Resistance, Lipoprotein Metabolism and Amyloidosis in Alzheimer’s disease. Health 6: 1549-1579. [Ref.]
  14. Martins IJ, Creegan R, Lim WLF, Martins RN (2013) Molecular insights into appetite control and neuroendocrine disease as risk factors for chronic diseases in Western countries. OJEMD 3: 11-33. [Ref.]
  15. Martins IJ, Hone E, Foster JK, Sunram-Lea SI, Gnjec A, et al. (2006) Apolipoprotein E, Cholesterol Metabolism, Diabetes and the Convergence of Risk Factors for Alzheimer’s Disease and Cardiovascular Disease. Mol Psychiatry 11: 721-736. [Ref.]
  16. Martins IJ, Berger T, Sharman MJ, Verdile G, Fuller SJ, et al. (2009) Cholesterol Metabolism and Transport in the Pathogenesis of Alzheimer’s Disease. J Neurochem 111: 1275-1308. [Ref.]
  17. Martins IJ (2013) Increased risk for obesity and diabetes with neurodegeneration in developing countries. J Genet Mol Med S1: 001. [Ref.]
  18. Rose RL, Hodgson E (2004) Chemical and Physiological Influences onXenobiotic Metabolism. In: Hodgson E (eds) Chapter 9, A Textbook of Modern Toxicology. 3rd edition, 163-201. [Ref.]
  19. Yang CS, Brady JF, Hong JY (1992) Dietary effects on cytochromes P450, xenobiotic metabolism, and toxicity. FASEB J 6: 737-744. [Ref.]
  20. Jandacek RJ, Tso P (2001) Factors Affecting the Storage and Excretion of Toxic Lipophilic Xenobiotics. Lipids 12: 1289-1305. [Ref.]
  21. Jirtle RL, Skinner MK (2007) Environmental epigenomics and disease susceptibility. Nature Genetics 8: 253-262. [Ref.]
  22. Casals-Casas C, Desvergne B (2011) Endocrine Disruptors: From Endocrine to Metabolic Disruption. Annu Rev Physiol 73: 135-162. [Ref.]
  23. Martins IJ (2016) Anti-Aging Genes Improve Appetite Regulation and Reverse Cell Senescence and Apoptosis in Global Populations. Adv Aging Res 5: 9-26. [Ref.]
  24. Martins IJ (2015) Unhealthy Nutrigenomic Diets Accelerate NAFLD and Adiposity in Global communities. J Mol Genet Med 9: 1-11. [Ref.]
  25. Liang F, Kume S, Koya D (2009) SIRT1 and insulin resistance. Nat Rev Endocrinol 5: 367-373. [Ref.]
  26. Martins IJ, Lim WL, Wilson A, Laws S, Martins RN (2013) The acceleration of aging and Alzheimer’s disease through the biological mechanisms behind obesity and type II diabetes. Health 5: 913-920. [Ref.]
  27. Martins IJ, Wilson AC, Lim WLF, Laws SM, Laws SM, et al. (2012) Sirtuin 1 mediates the obesity induced risk of common degenerative diseases: Alzheimer’s disease, coronary artery disease and type 2 diabetes. Health 4: 1448-1456. [Ref.]
  28. Herskovits AZ, Guarente L (2014) SIRT1 in neurodevelopment and brain senescence. Neuron 81: 471-483. [Ref.]
  29. Martins IJ (2015) Nutritional and genotoxic stress contributes to diabetes and neurodegenerative diseases such as Parkinson’s and Alzheimer’s diseases. In: Frontiers in Clinical Drug Research— CNS and Neurological Disorders, Atta-ur-Rahman, London, United Kingdom 3: 158-192. [Ref.]
  30. Lu P, Liu J, Melikishvili M, Fried MG, Chi YI (2008) Crystallization of hepatocyte nuclear factor 4 alpha (HNF4 alpha) in complex with the HNF1 alpha promoter element. Acta Crystallogr Sect F Struct Biol Cryst Commun 64: 313-317. [Ref.]
  31. Grimm AA, Brace CS, Wang T, Stormo GD, Imai S (2011) A nutrientsensitive interaction between Sirt1 and HNF-1α regulates Crp expression. Aging Cell 10: 305-317. [Ref.]
  32. Martins IJ (2016) The role of clinical proteomics, lipidomics and genomics in the diagnosis of Alzheimer’s disease. Manuscript ID: proteomes-119802, Proteomes. [Ref.]
  33. Liang Q, Zhong L, Zhang J, Wang Y, Bornstein SR, et al. (2014) FGF21 maintains glucose homeostasis by mediating the cross talk between liver and brain during prolonged fasting. Diabetes 63: 4064- 4075. [Ref.]
  34. Cheung EC, Vousden KH (2010) The role of p53 in glucose metabolism. Curr Opin Cell Biol 22: 186-191. [Ref.]
  35. Madan E, Gogna R, Bhatt M, Pati U, Kuppusamy P, et al. (2011) Regulation of glucose metabolism by p53: emerging new roles for the tumor suppressor. Oncotarget 2: 948-957. [Ref.]
  36. Moore KJ (2013) microRNAs: small regulators with a big impact on lipid metabolism. J Lipid Res 54: 1159-1160. [Ref.]
  37. Mercado C, Eades G, Zhou Q (2013) MicroRNAs: A New Class of Master Regulators of Adipogenesis. Human Genet Embryol 3: 108. [Ref.]
  38. Xie H, Sun L, Lodish HF (2009) Targeting MicroRNAs in Obesity. Expert Opin Therap Targets 13: 1227-1238. [Ref.]
  39. Rottiers V, Näär AM (2012) MicroRNAs in metabolism and metabolic disorders. Nat Rev Mol Cell Biol 13: 239-250. [Ref.]
  40. Zhu H, Leung SW (2015) Identification of microRNA biomarkers in type 2 diabetes: a meta-analysis of controlled profiling studies. Diabetologia 58: 900-911. [Ref.]
  41. McClelland AD, Kantharidis P (2014) microRNA in the development of diabetic complications. Clin Sci (Lond) 126: 95-110. [Ref.]
  42. Lee J, Kemper JK (2010) Controlling SIRT1 Expression by microRNAs in Health and Metabolic Disease. Aging (Albany NY) 2: 527-534. [Ref.]
  43. Yamakuchi M, Lowenstein CJ (2009) MiR-34, SIRT1 and p53: the feedback loop. Cell Cycle 8: 712-715. [Ref.]
  44. Tsai KL, Chen LH, Chen YC, Kao CL, Chen LK, et al. (2011) The Role of microRNAs in Modulating Sirtuin 1 Expression. J Clin Geront Geriat 2: 71-75. [Ref.]
  45. Miyazaki Y, Li R, Rezk A, Misirliyan H, Moore C, et al. (2014) A Novel MicroRNA-132-Surtuin-1 Axis Underlies Aberrant B-cell Cytokine Regulation in Patients with Relapsing-Remitting Multiple Sclerosis. PLoS One 9: e105421 [Ref.]
  46. Liu Q, Yuan B, Lo KA, Patterson HC, Sun Y, et al. (2012) Adiponectin regulates expression of hepatic genes critical for glucose and lipid metabolism. Proc Natl Acad Sci U S A 109: 14568-14573. [Ref.]
  47. Yahagi N, Shimano H, Matsuzaka T, Sekiya M, Najima Y, et al (2004) p53 Involvement in the Pathogenesis of Fatty Liver Disease. J Biol Chem 279: 20571-20575. [Ref.]
  48. Brenmoehl J, Hoeflich A (2013) Dual control of mitochondrial biogenesis by sirtuin 1 and sirtuin 3. Mitochondrion 13: 755-761. [Ref.]
  49. Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, et al. (2007) Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell 26: 745-752. [Ref.]
  50. Rokavec M, Li H, Jiang L, Hermeking H (2014) The p53/miR-34 axis in development and disease. J Mol Cell Biol 6: 214-230. [Ref.]
  51. Martins IJ, Fernando WMADB (2014) High fibre diets and Alzheimer’s disease. Food and Nutrition Sciences 5: 410-424. [Ref.]
  52. Suchankova G, Nelson LE, Gerhart-Hines Z, Kelly M, Gauthier MS, et al. (2009) Concurrent regulation of AMP-activated protein kinase and SIRT1 in mammalian cells. Biochem Biophys Res Commun 378: 836-841. [Ref.]
  53. Ojha S, Goyal S, Kumari S, Arya DS (2012) Pyruvate attenuates cardiac dysfunction and oxidative stress in isoproterenol-induced cardiotoxicity. Exp Toxicol Pathol 64: 393-399. [Ref.]
  54. Izumi Y, Katsuki H, Zorumski CF (1997) Monocarboxylates (pyruvate and lactate) as alternative energy substrates for the induction of longterm potentiation in rat hippocampal slices. Neurosci Lett 232: 17-20. [Ref.]
  55. Martins IJ (2015) Diabetes and Organ Dysfunction in the Developing and Developed. World Global Journal of Medical Research: F Diseases 15. [Ref.]
  56. Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, et al. (2009) Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58: 1509-1517 [Ref.]
  57. Davie JR (2003) Inhibition of histone deacetylase activity by butyrate. J Nutr 133: 2485S-2493S. [Ref.]
  58. Engel N, Mahlknecht U (2008) Aging and anti-aging: unexpected side effects of everyday medication through sirtuin1 modulation. Int J Mol Med 21: 223-232. [Ref.]
  59. Lu M, Sarruf DA, Li P, Osborn O, Sanchez-Alavez M, et al. (2013) Neuronal Sirt1 deficiency increases insulin sensitivity in both brain and peripheral tissues. J Biol Chem 288: 10722-10735. [Ref.]
  60. Zhou XM, Zhang X, Zhang XS, Zhuang Z, Li W, et al. (2014) SIRT1 inhibition by sirtinol aggravates brain edema after experimental subarachnoid hemorrhage. J Neurosci Res 92: 714-722. [Ref.]
  61. Dumbreck S, Flynn A, Nairn M, Wilson M, Treweek S, et al. (2015) Drug-disease and drug-drug interactions: systematic examination of recommendations in 12 UK national clinical guidelines. BMJ 350: h949. [Ref.]
  62. Stamatovic SM, Keep RF, Andjelkovic AV (2008) Brain endothelial cell-cell junctions: how to “open” the blood brain barrier. Curr Neuropharmacol 6: 179-192. [Ref.]
  63. Lippmann ES, Azarin SM, Kay JE, Nessler RA, Wilson HK, et al. (2012) Derivation of blood-brain barrier endothelial cells from human pluripotent stem cells. Nat Biotechnol 30: 783-791. [Ref.]
  64. Lombardo L, Pellitteri R, Balazy M, Cardile V (2008) Induction of nuclear receptors and drug resistance in the brain microvascular endothelial cells treated with antiepileptic drugs. Curr Neurovasc Res 5: 82-92. [Ref.]
  65. Wang YQ, Cao Q, Wang F, Huang LY, Sang TT, et al. (2015) SIRT1 Protects Against Oxidative Stress-Induced Endothelial Progenitor Cells Apoptosis by Inhibiting FOXO3a via FOXO3a Ubiquitination and Degradation. J Cell Physiol 230: 2098-2107. [Ref.]
  66. Arunachalam G, Samuel SM, Marei I, Ding H, Triggle CR (2014) Metformin modulates hyperglycaemia-induced endothelial senescence and apoptosis through SIRT1. Br J Pharmacol 171: 523- 535. [Ref.]
  67. Yu AM (2009) Role of microRNAs in the regulation of drug metabolism and disposition. Expert Opin Drug Metab Toxicol 5:1513-1528. [Ref.]
  68. Schmidt MF (2014) Drug target miRNAs: chances and challenges. Trends Biotechnol 32: 578-585. [Ref.]
  69. Li Z, Rana TM (2014) Therapeutic targeting of microRNAs: current status and future challenges. Nat Rev Drug Discov1 3: 622-638. [Ref.]
  70. Liao JM, Cao B, Zhou X, Lu H (2014) New insights into p53 functions through its target microRNAs. J Mol Cell Biol 6: 206-213. [Ref.]
  71. Bolleyn J, De Kock J, Rodrigues RM, Vinken M, Rogiers V, et al. (2015) MicroRNAs as key regulators of xenobiotic biotransformation and drug response. Arch Toxicol 89:1523-1541. [Ref.]
  72. Rodrigues AC, Li X, Radecki L, Pan YZ, Winter JC, et al. (2011) MicroRNA expression is differentially altered by xenobiotic drugs in different human cell lines. Biopharm Drug Dispos 32: 355-367. [Ref.]
  73. Martins IJ (2015) Overnutrition Determines LPS Regulation of Mycotoxin Induced Neurotoxicity in Neurodegenerative Diseases. Int J Mol Sci. 16: 29554-29573. [Ref.]
  74. Li Y, Powell DA, Shaffer SA, Rasko DA, Pelletier MR, et al. (2012) LPS remodeling is an evolved survival strategy for bacteria. Proc Natl Acad Sci USA 109: 8716-8721. [Ref.]
  75. Clifton LA, Skoda MWA, Daulton EL, Hughes AV, Le Brun AP, et al. (2013) Asymmetric phospholipid: lipopolysaccharide bilayers; a Gram-negative bacterial outer membrane mimic. J R Soc Interface 10: 20130810. [Ref.]

Download Provisional PDF Here

 

Article Information

Article Type: Short Commentary

Citation: Martins IJ, Calderón AM (2016) Diet and Nutrition reverse Type 3 Diabetes and Accelerated Aging linked to Global chronic diseases. J Dia Res Ther 2(2): doi http://dx.doi.org/10.16966/2380-5544.117

Copyright: © 2016 Martins IJ, et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Publication history: 

  • Received date: 15 Feb 2016

  • Accepted date: 25 Feb 2016

  • Published date: 03 Mar 2016
  •