We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Role of biomarkers in predicting diabetes complications with special reference to diabetic foot ulcers

    Sivakamasundari Pichu

    V Clinbio labs (P) Ltd, Sri Ramachandra University, Porur, Chennai, India

    AU-KBC, Anna University – MIT campus, Chromepet, Chennai – 44, India

    ,
    Bhoomika M Patel

    Institute of Pharmacy, Nirma University, Ahmedabad – 382481, India

    ,
    Subbu Apparsundaram

    V Clinbio labs (P) Ltd, Sri Ramachandra University, Porur, Chennai, India

    &
    Ramesh K Goyal

    *Author for correspondence:

    E-mail Address: goyalrk@gmail.com

    V Clinbio labs (P) Ltd, Sri Ramachandra University, Porur, Chennai, India

    Current Address: Delhi Pharmaceutical Sciences and Research University, Delhi 110017, India

    Search for more papers by this author

    Published Online:https://doi.org/10.2217/bmm-2016-0205

    Diabetic foot ulcer (DFU) is one of the major complications of diabetes and about 1% of people with diabetes have to go for lower limb amputation. With better understanding of the pathological basis of DFU, number of biomarkers like atrial natriuretic peptides, galectin-3, and cardiac troponins for diabetic cardiomyopathy, cystatin C for diabetics nephropathy and C-reactive protein for infection and procalcitonin could aid in early and noninvasive diagnosis especially when clinical signs are misleading. Predictive role of novel biomarkers in primary prevention however, requires additional studies considering sex, age and multiple complications in DFU. The current review provides an insight about the novel and emerging biomarkers of diabetes and its complications with special reference to DFUs.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 International Diabetes Federation. IDF Diabetes Atlas 7th Edition. www.diabetesatlas.org/resources/2015-atlas.html.
    • 2 Weerasuriya N, Siribaddana S, Dissanayake A et al. Long-term complications in newly diagnosed Sri Lankan patients with Type 2 diabetes mellitus. Q. J. Med. 91, 439–443 (1998).
    • 3 Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 69, 89–95 (2001).
    • 4 Goyal BR, Mehta AA. Diabetic cardiomyopathy: pathophysiological mechanisms and cardiac dysfunction. Hum. Exp. Toxicol. 32, 571–590 (2013).
    • 5 Patel BM, Mehta AA. Aldosterone and angiotensin: role in diabetes and cardiovascular diseases. Eur. J. Pharmacol. 697(1–3), 1–12 (2012).
    • 6 Patel BM, Mehta AA. The choice of anti-hypertensive agents in diabetic subjects. Diab. Vasc. Dis. Res. 10(50), 385–396 (2013).
    • 7 Raghunathan S, Patel BM. Therapeutic implications of small interfering RNA in cardiovascular diseases. Fund. Clin. Pharmacol. 27(1), 1–20 (2013).
    • 8 Hubbard LD, Brothers RJ, King WN et al. Methods for evaluation of retinal microvascular abnormalities associated with hypertension/sclerosis in the Atherosclerosis Risk in Communities study. Ophthalmology 106, 2269–2280 (1999).
    • 9 Gordois A, Scuffham P, Shearer A, Oglesby A, Tobian JA. The health care costs of diabetic peripheral neuropathy in the US. Diabetes Care 26, 1790–1795 (2003).
    • 10 Deshpande AD, Harris-Hayes M, Schootman M. Epidemiology of diabetes and diabetes-related complications. Phys. Ther. 88(11), 1254–1264 (2008).
    • 11 Habib AA, Brannagan TH 3rd. Therapeutic strategies for diabetic neuropathy. Curr. Neurol. Neurosci. Rep. 10, 92–100 (2010).
    • 12 Kiernan MC. Emergence of a predictive clinical biomarker for diabetic neuropathy. Diabetes 61(6), 1346–1347 (2012).
    • 13 Galkowska H, Olszewski WL, Wojewodzka U, Rosinski G, Karnafel W. Neurogenic factors in the impaired healing of diabetic foot ulcers. J. Surg. Res. 134(2), 252–258 (2006).
    • 14 Goren I, Muller E, Pfeilschifter J, Frank S. Severely impaired insulin signaling in chronic wounds of diabetic ob/ob mice: a potential role of tumor necrosis factor-alpha. Am. J. Pathol. 168, 765–777 (2006).
    • 15 Falanga V. Wound healing and its impairment in the diabetic foot. Lancet 366, 1736–1743 (2005).
    • 16 Galiano RD, Tepper OM, Pelo CR et al. Topical vascular endothelial growth factor accelerates diabetic wound healing through increased angiogenesis and by mobilizing and recruiting bone marrow-derived cells. Am. J. Pathol. 164, 1935–1947 (2004).
    • 17 Maruyama K, Asai J, Ii M, Thorne T, Losordo DW, D'Amore PA. Decreased macrophage number and activation lead to reduced lymphatic vessel formation and contribute to impaired diabetic would healing. Am. J. Pathol. 170, 1178–1191 (2007).
    • 18 Gibran NS, Jang YC, Isik FF et al. Diminished neuropeptide levels contribute to the impaired cutaneous healing response associated with diabetes mellitus. J. Surg. Res. 108, 122–128 (2002).
    • 19 Lobmann R, Ambrosch A, Schultz G, Waldmann K, Schiweck S, Lehnert H . . Expression of matrix-metalloproteinases and their inhibitors in the wounds of diabetic and non-diabetic patients. Diabetologia 45, 1011–1016 (2002).
    • 20 Deshpande SD, Putta S, Wang M et al. Transforming growth factor-beta induced cross talk between p53 and a microRNA in the pathogenesis of diabetic nephropathy. Diabetes 62(9), 3151–62 (2013).
    • 21 Kato M, Arce L, Wang M et al. A microRNA circuit mediates transforming growth factor-beta1 auto regulation in renal glomerular mesangial cells. Kidney Int. 80, 358–368 (2011).
    • 22 Wang Q, Wang Y, Minto AW et al. MicroRNA-377 is up-regulated and can lead to increased fibronectin production in diabetic nephropathy. FASEB J. 22, 4126–4135 (2008).
    • 23 Zhang Z, Peng H, Chen J et al. MicroRNA-21 protects from mesangial cell proliferation induced by diabetic nephropathy in db/db mice. FEBS Lett. 583, 2009–2014 (2009).
    • 24 Zhong X, Chung AC, Chen HY et al. miR-21 is a key therapeutic target for renal injury in a mouse model of Type 2 diabetes. Diabetologia 56, 663–674 (2013).
    • 25 Long J, Wang Y, Wang W et al. MicroRNA-29c is a signature microRNA under high glucose conditions that targets Sprouty homolog 1, and its in vivo knockdown prevents progression of diabetic nephropathy. J. Biol. Chem. 286, 11837–11848 (2011).
    • 26 Mu J, Pang Q, Guo YH et al. Functional implications of microRNA-215 in TGF-beta1-induced phenotypic transition of mesangial cells by targeting CTNNBIP1. PLoS ONE 8, e58622 (2013).
    • 27 Dey N, Das F, Mariappan MM et al. MicroRNA-21 orchestrates high glucose-induced signals to TOR complex 1, resulting in renal cell pathology in diabetes. J. Biol. Chem. 286, 25586–25603 (2011).
    • 28 Feng B, Chen S, McArthur K et al. miR-146a mediated extracellular matrix protein production in chronic diabetes complications. Diabetes 60, 2975–2984 (2011).
    • 29 McArthur K, Feng B, Wu Y et al. MicroRNA-200b regulates vascular endothelial growth factor-mediated alterations in diabetic retinopathy. Diabetes 60, 1314–1323 (2011).
    • 30 Silva VA, Polesskaya A, Sousa TA et al. Expression and cellular localization of microRNA-29b and RAX, an activator of the RNA-dependent protein kinase (PKR), in the retina of streptozotocin-induced diabetic rats. Mol. Vis. 17, 2228–2240 (2011).
    • 31 Shan ZX, Lin QX, Deng CY et al. miR-1/miR-206 regulate Hsp60 expression contributing to glucose mediated apoptosis in cardiomyocytes. FEBS Lett. 584, 3592–3600 (2010).
    • 32 Katare R, Caporali A, Zentilin L et al. Intravenous gene therapy with PIM-1 via a cardiotropic viral vector halts the progression of diabetic cardiomyopathy through promotion of prosurvival signaling. Circ. Res. 108, 1238–1251 (2011).
    • 33 Care A, Catalucci D, Felicetti F et al. MicroRNA-133 controls cardiac hypertrophy. Nat. Med. 13, 613–618 (2007).
    • 34 Horie T, Ono K, Nishi H. MicroRNA-133 regulates the expression of GLUT4 by targeting KLF15 and is involved in metabolic control in cardiac myocytes. Biochem. Biophys. Res. Commun. 389, 315–320 (2009).
    • 35 Shen E, Diao X, Wang X. MicroRNAs involved in the mitogen-activated protein kinase cascades pathway during glucose induced cardiomyocyte hypertrophy. Am. J. Pathol. 179, 639–650 (2011).
    • 36 van Rooij E, Sutherland LB, Liu N et al. A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure. Proc. Natl Acad. Sci. USA 103, 18255–18260 (2006).
    • 37 Thum T, Gross C, Fiedler J et al. MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature 456, 980–984 (2008).
    • 38 Duisters RF, Tijsen AJ, Schroen B et al. miR-133 and miR-30 regulate connective tissue growth factor: implications for a role of microRNAs in myocardial matrix remodeling. Circ. Res. 104, 170–178 (2009).
    • 39 Villeneuve LM, Kato M, Reddy MA et al. Enhanced levels of microRNA-125b in vascular smooth muscle cells of diabetic db/db mice lead to increased inflammatory gene expression by targeting the histone methyltransferase Suv39h1. Diabetes 59, 2904–2915 (2010).
    • 40 Reddy MA, Jin W, Villeneuve L et al. Pro inflammatory role of microRNA-200 in vascular smooth muscle cells from diabetic mice. Arterioscler. Thromb. Vasc. Biol. 32, 721–729 (2012).
    • 41 Wang XH, Qian RZ, Zhang W et al. MicroRNA-320 expression in myocardial microvascular endothelial cells and its relationship with insulin-like growth factor-1 in Type 2 diabetic rats. Clin. Exp. Pharmacol. Physiol. 36, 181–188 (2009).
    • 42 Caporali A, Meloni M, Vollenkle C. Deregulation of microRNA-503 contributes to diabetes mellitus-induced impairment of endothelial function and reparative angiogenesis after limb ischemia. Circulation 123, 282–291 (2011).
    • 43 Mustoe T. Understanding chronic wounds: a unifying hypothesis on their pathogenesis & implications for therapy. Am. J. Surg. 187(5A), S65–S70 (2004).
    • 44 Crovetti G, Martinelli G, Issi M et al. Platelet gel for healing cutaneous chronic wounds. Transfus. Apher. Sci. 30(2), 145–51 (2004).
    • 45 Watters C, DeLeon K, Trivedi U et al. Pseudomonas aeruginosa biofilms perturb wound resolution and antibiotic tolerance in diabetic mice. Med. Microbiol. Immunol. 202(2), 131–141 (2013).
    • 46 Halcón L, Milkus K. Staphylococcus aureus and wounds: a review of tea tree oil as a promising antimicrobial. Am. J. Infect. Control. 32(7), 402–408 (2004).
    • 47 Yates C, May K, Hale T et al. Wound chronicity, inpatient care, and chronic kidney disease predispose to MRSA infection in diabetic foot ulcers. Diabetes Care 32(10), 1907–1909 (2009).
    • 48 Foy Y, Li J, Kirsner R, Eaglstein W. Analysis of fibroblast defects in extracellular matrix production in chronic wounds. J. Am. Acad. Dermatol. 50(3), 168 (2004).
    • 49 Augustin M, Maier K. Psychosomatic aspects of chronic wounds. Dermatol. Psychosom. 4, 5–13 (2003).
    • 50 Mueller C, Muller B, Perruchoud AP. Biomarkers: past, present, and future. Swiss Med. Wkly. 138(15–16), 225–229 (2008). •• Explains about the general views of past, present and future biomarker studies.
    • 51 Domenici E, Willé DR, Tozzi F et al. Plasma protein biomarkers for depression and schizophrenia by multi analyte profiling of case–control collections. PLoS ONE 5(2), e9166 (2010).
    • 52 Yager DR, Kulina RA, Gilman LA. Wound fluids: a window into the wound environment? Int. J. Low Extrem. Wounds 6(4), 262–272 (2007).
    • 53 Schreml S, Szeimies RM, Prantl L, Karrer S, Landthaler BP. Oxygen in acute and chronic wound healing. Br. J. Dermatol. 163(2), 257–268 (2010).
    • 54 Massara M, Caridi GD, Serra R et al. The role of procalcitonin as a marker of diabetic foot ulcer infection. Int. Wound J. 14(1), 31–34 (2017). •• Explains about the recent advancement on the foot ulcers and identification of potential biomarker for infected diabetic foot ulcers.
    • 55 Franks R, Hargreaves R. Clinical biomarkers in drug discovery and development. Nat. Rev. Drug. Discov. 2, 566–568 (2003).
    • 56 Mayeux R. Biomarkers: potential uses and limitations. J. Am. Soc. Exp. Neurother. 1, 182–188 (2004). •• Explains about the general biomarker categories and how to choose potential biomarkers for certain diseases.
    • 57 Joseph V, Boykin J. Wound nitric oxide bioactivity: a promising diagnostic indicator for diabetic foot ulcer management. J. Wound Ostomy Continence Nurs. 37(1), 25–32 (2010).
    • 58 Soneja A, Drews M, Malinski T. Role of nitric oxide, nitroxidative and oxidative stress in wound healing. Pharmacol. Rep. 57(Suppl.), 108–119 (2005).
    • 59 Mendoza-Mari Y, Valdés-Pérez C, Rodríguez-Corrales E et al. Histological and transcriptional expression differences between diabetic foot and pressure ulcers. J. Diabetes Metab. 4, 296 (2013). • Interesting paper explaining about the genetic expression profile on different stages of wound healing.
    • 60 Loots MA, Lamme EN, Zeegelaar J et al. Differences in cellular infiltrate and extracellular matrix of chronic diabetic and venous ulcers versus acute wounds. J. Invest. Dermatol. 111, 850–857 (1998). • Interesting paper on matrix proteins involvement and their role on different ulcers.
    • 61 Piaggesi A, Viacava P, Rizzo L et al. Semiquantitative analysis of the histopathological features of the neuropathic foot ulcer: effects of pressure relief. Diabetes Care 26, 3123–3128 (2003).
    • 62 Eming AS, Koch M, Krieger A et al. Differential proteomic analysis distinguishes tissue repair biomarker signatures in wound exudates obtained from normal healing and chronic wounds. J. Proteome Res. 9, 4758–4766 (2010). • Explains about the current proteomics approach and its usage in identifying the biomarker candidates.
    • 63 Pichu S, Sathiyamoorthy J, Krishnamoorthy E, Umapathy D, Viswanathan V. Impact of the hypoxia inducible factor-1α (HIF-1α) pro582ser polymorphism and its gene expression on diabetic foot ulcers. Diabetes Res. Clin. Pract. 109(3), 533–540 (2015).