Open Access Open Access  Restricted Access Subscription Access
Open Access Open Access Open Access  Restricted Access Restricted Access Subscription Access

Novel Approaches for Diabetes Mellitus: A Review


Affiliations
1 Department of Clinical Pharmacy, Shri Sarvajanik Pharmacy College, Nr. Arvind Baug, Mehsana - 384 001 Gujarat, India
2 Shri Sarvajanik Pharmacy College, Nr. Arvind Baug, Mehsana - 384 001, Gujarat, India
     

   Subscribe/Renew Journal


Diabetes mellitus is a major and growing public health problem of the developed country. Diabetes mellitus is also associated with disease like hypertension, chronic heart disease, blindness etc.. Now days drug that are available in the market are just to control the diabetes. There are several novel approaches which might cure the diabetes. Defective glucose-stimulated insulin secretion by pancreatic islet β cells could be cured with recombinant glucagon-like peptide 1 (GLP-1) or agonists of the GLP-1 receptor. Alternatively, decrease in GLP-1 clearance can be achieved with inhibition of Dipeptidylpeptidase IV (DP-IV) to reduce insulin resistance, enhanced insulin action. The role of peroxisome proliferator activated receptors (PPAR γ) in the regulation of lipid metabolism, insulin and triglycerides leads to the rationale design of several PPAR agonists. Gene therapy also generates greater hope for possible cure of diabetes. Sodium-Glucose Co-Transporter Inhibitor is also one of the novel target for lowering plasma glucose and improving insulin resistance by increasing renal glucose excretion. Under diabetic conditions, induced oxidative stress also activates the JNK pathway, which is involved in deterioration of pancreatic β-cell function found in diabetes. Treatment with antioxidants and/or suppression of the JNK pathway protect β-cells from some of the toxic effects of hyperglycemia could be the one of novel target therapy of diabetes mellitus.

Keywords

Diabetes Mellitus, Incretin, DPP-IV Inhibitor, Gene Therapy, Novel Approach.
Subscription Login to verify subscription
User
Notifications
Font Size


  • Amos A, McCarty D and Zimmet P. The rising global burden of diabetes and its complications: estimates and projections by 2010. Diabet. Med. 1997; 5: S5-S85.
  • Nagappa A. 16-Novel strategies for the therapeutic management of type 2 diabetes. 2000; 1-2: 58-68.
  • Shaw J et al. Type 2 Diabetes Worldwide According to the New Classification and Criteria. Diabetes Care 23 (Suppl. 2). 2000; 23: B5-B10.
  • Curtis L et al.Diabetes Mellitus. In DiPiro JT et al,Pharmacotherapy, Edited by McGraw-Hill, New York: 2005;6 th Edi: pp,1335-36
  • Harris M. National institute of health, diabetes and digestive and kidney diseases, "diabetes in america".Diabetes care.1995; 95-1468.
  • Moller DE. New drug targets for type 2 diabetes and the metabolic syndrome Nature. 2001; 414: 820-27.
  • Gautier JF, Choukem SP, Girard J, Physiology of incretins (GIP and GLP-1) and abnormalities in type 2 diabetes, Elsevier.Diabetes Metab.2008; 34: S65-S72.
  • Fehmann HC, Goke R, Goke B. Cell and molecular biology of the incretin hormones glucagon-like peptide-I and glucose-dependent insulin releasing polypeptide. Endocr Rev.1995; 16: 390-410.
  • Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132: 2131-57.
  • Egan JM et al. GLP-1 receptor agonists are growth and differentiation factors for pancreatic islet beta cells. Diabetes Metab Res Rev. 2003; 19: 115-23.
  • Naslund E et al. Energy intake and appetite are suppressed by glucagon-like peptide-1 (GLP-1) in obese men. Int J Obes Relat Metab Disord. 1999; 23: 304-11.
  • Gutzwiller JP et al. Glucagon-like peptide-1 promotes satiety and reduces food intake in patients with diabetes mellitus type 2. Am J Physiol. 1999; 276: R1541-4.
  • Nauck MA et al. Additive insulinotropic effects of exogenous synthetic human gastric inhibitory polypeptide and glucagon-like peptide-1-(7-36) amide infused at near-physiological insulinotropic hormone and glucose concentrations. J Clin Endocrinol Metab. 1993; 76: 912.
  • Kreymann B et al. Glucagon-like peptide-1 (7-36): a physiological incretin in man. Lancet. 1987; 2: 1300-4.
  • Zander M et al. Effect of 6-week course of glucagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function in type 2 diabetes: a parallel-group study. Lancet 2002; 359: 824- 30.
  • Hui H, Wright C, Perfetti R. Glucagon-like peptide 1 induces differentiation of islet duodenal homeobox-1-positive pancreatic ductal cells into insulin-secreting cells. Diabetes. 2001; 50: 785- 96.
  • Abraham EJ et al. Insulinotropic hormone glucagon-like peptide- 1 differentiation of human pancreatic islet-derived progenitor cells into insulin-producing cells. Endocrinology. 2002; 143: 3152- 61.
  • Hui H et al. Glucagon-like peptide-1 inhibits apoptosis of insulinsecreting cells via a cyclic 5V-adenosine monophosphatedependent protein kinase A- and a phosphatidylinositol 3- kinase-dependent pathway. Endocrinology. 2003; 144: 1444- 55.
  • Brubaker PL, Drucker DJ. Minireview: glucagon-like peptides regulate cell proliferation and apoptosis in the pancreas, gut, and central nervous system. Endocrinology. 2004; 145: 2653- 9 .
  • Xu G et al. Exendin-4 stimulates both beta-cell replication and neogenesis, resulting in increased beta-cell mass and improved glucose tolerance in diabetic rats. Diabetes. 1999; 48: 2270- 6.
  • Eng J et al. Isolation and characterization of exendin-4, an exendin-3 analogue from Heloderma suspectum venom. J Biol Chem. 1992; 267: 7402 -5.
  • Loretta L et al. Pharmacology of exenatide (synthetic exendin- 4): a potential therapeutic for improved glycemic control of type 2 diabetes , Regul. Pept 2004;117 :77-88.
  • Holz GG, Chepurny OG. Glucagon-like peptide-1 synthetic analogs: new therapeutic agents for use in the treatment of diabetes mellitus. Curr Med Chem. 2003; 10: 2471-83.
  • Mentlein R, Gallwitz B, Schmidt W. Dipeptidyl peptidase IV hydrolyzes gastric inhibitory polypeptide, glucagon-like peptide- 1(7-36), peptide histidine methionine and is responsible for their degradation in human serum. Eur J Biochem. 1993; 214: 829- 35.
  • Christopher HS, Demuth HU, Kim SJ. Applications of dipeptidyl peptidase IV inhibitors in diabetes mellitus, Int J Biochem Cell Bio. 2006; 38: 860-872.
  • Low SH et al. Apical cell surface expression of rat dipeptidyl peptidase IV in transfected Madin -Darby canine kidney cells. J Biol Chem. 1991; 266: 13391- 6.
  • Lambeir AM et al. Dipeptidyl-peptidase IV from bench to bedside: an update on structural properties, functions and clinical aspects of the enzyme DPP IV. Crit Rev Clin Lab Sci. 2003; 40: 209-94.
  • Meester DI et al. Dipeptidyl peptidase IV substrates. Adv Exp Med Biol. 2003; 524: 3 - 13.
  • Mentlein R. Dipeptidyl-peptidase IV (CD-26)-role in the inactivation of regulatory peptides. Regul Pept. 1999; 85: 9-24.
  • Augustyns K et al. The unique properties of dipeptidyl peptidase IV (DPP IV/ CD26) and the therapeutic potential of DPP IV inhibitors. Curr Med Chem. 1999; 6: 311 -27.
  • Villhauer EB et al. 1-[[(3-Hydroxy-1-adamantyl)amino] acetyl]2- cyano-(S)-pyrrolidine: a potent, selective, and orally bioavailable dipeptidyl peptidase IV inhibitor with antihyperglycemic properties. J Med Chem. 46: 2774-2789.
  • Dardik B et al. A dipeptidyl peptidase IV inhibitor, improves glucose tolerance and delays gastric emptying in obese insulin resistant cynomolgus monkeys. Diabetes. 2003 ;52: A322.
  • Ahren B et al. Inhibition of dipeptidyl peptidase-4 reduces glycemia, sustains insulin levels, and reduces glucagon levels in type 2 diabetes. J Clin Endocrinol Metab.2004; 89: 2078-2084.
  • Ahren B et al . Twelve- and 52-week efficacy of the dipeptidyl peptidase IV inhibitor LAF237 in metformin-treated patients with type 2 diabetes.Diabetes Care.2004; 27:2874-2880.
  • Balas BN et al. The Dipeptidyl Peptidase IV Inhibitor Vildagliptin Suppresses Endogenous Glucose Production and Enhances Islet Function after Single-Dose Administration in Type 2 Diabetic Patients; The Journal of Clinical Endocrinology & Metabolism. 2009; 92(4): 1249-1255.
  • Issemann I and Green S. An oxysterol signalling pathway mediated by the nuclear receptor LXR α .Nature. 1990; 347: 645-650.
  • Lemberger T, Desvergne B, Wahli W. Peroxisome proliferatoractivated receptors: A Nuclear Receptor Signaling Pathway In Lipid Physiology. Annu Rev Cell Dev Biol. 1996;12:335-363.
  • Vamecq J and Latruffe N. Medical significance of peroxisome proliferator-activated receptors. Diabetes. 1999; 354: 141-148.
  • Auboeuf, D et al. Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor-α in humans. No alteration in adipose tissue of obese and NIDDM patients.Diabetes.1997; 48: 1319-1327.
  • Park, KS et al. Troglitazone Effects on Gene Expression in Human Skeletal Muscle of Type II Diabetes Involve Up- Regulation of Peroxisome Proliferator-Activated Receptor-g. Diabetes. 1997; 46: 1230- 1234.
  • Loviscach M et al. Distribution of peroxisome proliferator-activated receptors (PPARs) in human skeletal muscle and adipose tissue: relation to insulin action. Diabetologia. 2000;43: 304-311.
  • Martin, G et al. PPARγ activators improve glucose homeostasis by stimulating fatty acid uptake in the adipocytes. Atherosclerosis J. Biol. Chem.1997;272: 28210-28217.
  • Berger, J, Moller, DE. Mechanism of PPAR action. Annu. Rev. Med. 2002;53:409-35.
  • Oakes ND et al. A new antidiabetic agent, BRL 49653, reduces lipid availability and improves insulin action and gluco regulation in the rat. Diabetes. 1994; 43: 1203-1210.
  • Osada, T et al. Identification of an extended half-site motif required for the function of peroxisome proliferator-activated receptor alpha Genes Cells. 1997; 2:315-327.
  • Lavinsky RM et al. Diverse singaling pathway modulate nuclear receptor recruitment of N-CR and SMART complex Proc. Natl Acad. Sci.1998; 95: 2920-2925.
  • Glass CK, Rose DW and Rosenfeld, M. G., Nuclear receptor co activators and corepressors. Curr. Opin. Cell. Biol.1997; 9: 222-232.
  • Esterbauer H et al. Human peroxisome proliferator activated receptor gamma co activator 1 (PPARGC1) gene: c DNA sequence, genomic organization, chromosomal localization, and tissue expression. Genomics. 1999; 62: 98-102.
  • Pazin MJ and Kadonaga, JT, What's up and down with histone deacetylation and transcription? Cell.1997; 89: 325-328.
  • Shao D et al. Interdomain communication regulating ligand binding by PPAR-gamma. Nature.1998; 396: 377-380.
  • Camp HS, Tafuri SR.Regulation of peroxisome proliferatoractivated receptor gamma activity by mitogen-activated protein kinase J. Biol. Chem.1997; 272: 10811-10816.
  • Dieterle C et al. Therapy of diabetes mellitus. Pancreas transplantation, islet transplantation, stem cell and gene therap. Internist (Berl). 2006; 47: 489-496.
  • Samson SL, Chan L. Gene therapy for diabetes: reinventing the islet. Trends .Endocrinol Metab. 2006; 17: 92-100.
  • Fukushima M et al. Adiponectin gene therapy of streptozotocininduced diabetic mice using hydrodynamic injection. J Gene Med. 2007; 9: 976-985.
  • Lu YC et al. Release of transgenic human insulin from gastric g cells: a novel approach for the amelioration of diabetes. Endocrinology. 2005; 146: 2610-2619.
  • Cho YM et al. Betacellulin and nicotinamide sustain PDX1 expression and induce pancreatic beta-cell differentiation in human embryonic stem cells. Biochem Biophys Res Commun. 2008; 366: 129-134.
  • Serafimidis I et al. Novel effectors of directed and Ngn3- mediated differentiation of mouse embryonic stem cells into endocrine pancreas progenitors. Stem Cells. 2008; 26: 3-16.
  • Korbling M. Estrov, Z. Adult stem cells for tissue repair - a new therapeutic concept? N. Engl. J. Med. 2003; 349: 570-582.
  • Street CN. Stem cell-based approaches to solving the problem of tissue supply for islet transplantation in Type 1 diabetes. Int. J. Biochem. Cell Biol. 2004; 36: 667-683.
  • Herzog EL, Chai L, Krause DS. Plasticity of marrow-derived stem cells. Blood. 2003; 102: 3483-93.
  • Theise ND et al. Liver from bone marrow in humans. Hepatology. 2000; 32: 11-16.
  • Korbling M et al. Hepatocytes and epithelial cells of donor origin in recipients of peripheral blood stem cells. N Engl J Med. 2002; 346: 738-46.
  • Ianus A et al. In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion. J Clin Invest. 2003; 111: 843-50.
  • Jiang Y et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature .2002; 418: 41-49.
  • Hess D et al. Bone marrow-derived stem cells initiatepancreatic regeneration. Nat Biotechnol. 2003; 21: 763-70.
  • Teitelman, G. Induction of beta-cell neogenesis by islet injury. Diab Metab. Rev 1996; 12: 91-102.
  • Zalzman M et al. Reversal of hyperglycemia in mice by using human expandable insulin-producing cells differentiated from fetal liver progenitor cells. Proc Natl Acad Sci USA .2003; 100: 7253-58.
  • Meivar LI, Ferber S. New organs from our own tissues: liver-topancreas transdifferentiation. Trends Endocrinol Metab. 2003; 14: 460-66.
  • Kojima H et al. NeuroD-betacellulin gene therapy induces islet neogenesis in the liver and reverses diabetes in mice. Nat Med. 2003; 9: 596-603.
  • Dor Y et al. Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature. 2004; 429: 41-46.
  • Herold KC et al. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med. 2002; 346: 1692-98.
  • Bishop JH, Green R, and Thomas S. Free-flow reabsorption of glucose, sodium, osmoles and water in rat proximal convoluted tubule. J Physiol (Lond) .1979; 288:331- 351.
  • Silverman M and Turner RJ Glucose transport in the renal proximal tubule, in Handbook of Physiology. Section 8. Renal Physiology.1992; 2017-2038.
  • Kanai Y et al. The human kidney low affinity Na_/glucose cotransporter SGLT2: delineation of the major renal reabsorptive mechanism for D-glucose. J Clin Investig.1992; 93:397-404.
  • Van DH et al. Autosomal recessive renal glucosuria attributable to a mutation in the sodium glucose cotransporter (SGLT2). Hum Genet.2003; 111:544-547.
  • Pajor A and Wright EM. Cloning and functional expression of a mammalian Na_/nucleoside cotransporter. A member of the SGLT family. J Biol Chem.1992; 267: 3557-3560.
  • Turk E et al. Glucose/galactose malabsorption caused by a defect in the Na_/glucose cotransporter. Nature (Lond).1991; 350:354-356.
  • Wells RG et al. Cloning of a human kidney cDNA with similarity to the sodium-glucose cotransporter. Am J Physiol.1992; 263:F459-F465.
  • Ehrenkranz JR et al. A review.Diabetes Metab Res Rev;2005 21(1):31-38.
  • Thorens B. Glucose transporter in the regulation of intestinal,renal and liver glucose fluxes. Am J Physiolgastrointest. Liver Physiol. 1996; 270(33): G541-G543.
  • Rossetti L et al.Correction Of Hyperglycemia With Phlorizin Normalizes Tissue Sensitivity To Insuline In Diabeetic Rat.1987 ;79 :1510-15.
  • Katsuno et al. Sergliflozin A Novel Selective Inhibitor Of Low Affinity Sodium Glucose Cotransporter (Sglt2) Validates The Critical Role Of Sglt2 In Renal Glucose Reabsorption And Modulates Plasma Glucose.Level.J Pharmacol Exp Ther. 2007;320:323-330.
  • Wright Em,Turk E.The Sodium Glucose Cotransporter Family slc5 Pflugersarch;2004 447:510-18.
  • Nishimura S.Tissue Specific M-Rna Expression Profile Of Human Atp-Binding Cassette And Solute Carrier Transporter Superfamillies.Drug Meteb Pharmacokinetic ;2005 20:452-77.
  • Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipid metabolism. Nature.2001; 414: 799-806.
  • Weir GC et al. β- Cell adaptation and decompensation during the progression of diabetes. Diabetes. 2001; 50: S154-159.
  • Dandona P et al. Oxidative damage to DNA in diabetes mellitus. Lancet 1996; 347: 444-445.
  • Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001; 414: 813-820.
  • Kaneto H et al. Reducing sugars trigger oxidative modification and apoptosis in pancreatic b-cells by provoking oxidative stress through the glycation reaction. Biochem J. 1996; 320: 855-863.
  • Gorogawa S et al. Probucol preserves pancreatic b-cell function through reduction of oxidative stress in type 2 diabetes. Diabetes Res Clin Prac. 2002; 57: 1-10.
  • Sakai K et al. Mitochondrial reactive oxygen species reduce insulin secretion by pancreatic b-cells. Biochem Biophys Res Commun. 2003; 300: 216-222.
  • Miller CP, McGehee RE, Habener JF. IDX-1: a new homeodomain transcription factor expressed in rat pancreatic islets and duodenum that transactivates the somatostatin gene. EMBO J. 1994; 13: 1145-1156.
  • Dutta S.et al. Regulatory factor linked to late-onset diabetes? Nature. 1998; 392: 560.
  • Ahlgren U et al. β-Cellspecific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the β-cell phenotype and maturity onset diabetes. Genes Dev. 1998; 12: 1763-1768.
  • Stoffers DA et al. Early-onset type- II diabetes mellitus (MODY4) linked to IPF1. Nat Genet .1997; 17: 138-139.
  • Nakatani Y et al. Modulation of the JNK pathway in liver affects insulin resistance status. J Biol Chem .2004; 279: 45803-45809.
  • Kaneto H et al. Oxidative Stress and the JNK Pathway in Diabetes Current Diabetes Reviews, 2005; 1: 65-72.

Abstract Views: 276

PDF Views: 1




  • Novel Approaches for Diabetes Mellitus: A Review

Abstract Views: 276  |  PDF Views: 1

Authors

S. M. Bhanushali
Department of Clinical Pharmacy, Shri Sarvajanik Pharmacy College, Nr. Arvind Baug, Mehsana - 384 001 Gujarat, India
K. M. Modh
Shri Sarvajanik Pharmacy College, Nr. Arvind Baug, Mehsana - 384 001, Gujarat, India
I. S. Anand
Shri Sarvajanik Pharmacy College, Nr. Arvind Baug, Mehsana - 384 001, Gujarat, India
C. N. Patel
Shri Sarvajanik Pharmacy College, Nr. Arvind Baug, Mehsana - 384 001, Gujarat, India
J. B. Dave
Shri Sarvajanik Pharmacy College, Nr. Arvind Baug, Mehsana - 384 001, Gujarat, India

Abstract


Diabetes mellitus is a major and growing public health problem of the developed country. Diabetes mellitus is also associated with disease like hypertension, chronic heart disease, blindness etc.. Now days drug that are available in the market are just to control the diabetes. There are several novel approaches which might cure the diabetes. Defective glucose-stimulated insulin secretion by pancreatic islet β cells could be cured with recombinant glucagon-like peptide 1 (GLP-1) or agonists of the GLP-1 receptor. Alternatively, decrease in GLP-1 clearance can be achieved with inhibition of Dipeptidylpeptidase IV (DP-IV) to reduce insulin resistance, enhanced insulin action. The role of peroxisome proliferator activated receptors (PPAR γ) in the regulation of lipid metabolism, insulin and triglycerides leads to the rationale design of several PPAR agonists. Gene therapy also generates greater hope for possible cure of diabetes. Sodium-Glucose Co-Transporter Inhibitor is also one of the novel target for lowering plasma glucose and improving insulin resistance by increasing renal glucose excretion. Under diabetic conditions, induced oxidative stress also activates the JNK pathway, which is involved in deterioration of pancreatic β-cell function found in diabetes. Treatment with antioxidants and/or suppression of the JNK pathway protect β-cells from some of the toxic effects of hyperglycemia could be the one of novel target therapy of diabetes mellitus.

Keywords


Diabetes Mellitus, Incretin, DPP-IV Inhibitor, Gene Therapy, Novel Approach.

References