期刊论文详细信息
Molecular Neurodegeneration
Neuroinflammation and neurologic deficits in diabetes linked to brain accumulation of amylin
Florin Despa2  Linda J Van Eldik3  Kathryn E Saatman5  Xinmin Simon Xie6  Conrado Pascual6  Jennifer M Brelsfoard1  Adam B Bachstetter4  Savita Sharma2  Sarah Srodulski2 
[1] Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA;Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA;Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY, USA;Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA;Department of Physiology, University of Kentucky, Lexington, KY, USA;AfaSci Research Laboratories, AfaSci, Inc., Redwood City, CA, USA
关键词: Behavior;    Inflammation;    Insulin Resistance;    Pre-diabetes;    Amylin;    Alzheimer’s Disease;    Diabetes;   
Others  :  1138752
DOI  :  10.1186/1750-1326-9-30
 received in 2014-06-10, accepted in 2014-08-18,  发布年份 2014
PDF
【 摘 要 】

Background

We recently found that brain tissue from patients with type-2 diabetes (T2D) and cognitive impairment contains deposits of amylin, an amyloidogenic hormone synthesized and co-secreted with insulin by pancreatic β-cells. Amylin deposition is promoted by chronic hypersecretion of amylin (hyperamylinemia), which is common in humans with obesity or pre-diabetic insulin resistance. Human amylin oligomerizes quickly when oversecreted, which is toxic, induces inflammation in pancreatic islets and contributes to the development of T2D. Here, we tested the hypothesis that accumulation of oligomerized amylin affects brain function.

Methods

In contrast to amylin from humans, rodent amylin is neither amyloidogenic nor cytotoxic. We exploited this fact by comparing rats overexpressing human amylin in the pancreas (HIP rats) with their littermate rats which express only wild-type (WT) non-amyloidogenic rodent amylin. Cage activity, rotarod and novel object recognition tests were performed on animals nine months of age or older. Amylin deposition in the brain was documented by immunohistochemistry, and western blot. We also measured neuroinflammation by immunohistochemistry, quantitative real-time PCR and cytokine protein levels.

Results

Compared to WT rats, HIP rats show i) reduced exploratory drive, ii) impaired recognition memory and iii) no ability to improve the performance on the rotarod. The development of neurological deficits is associated with amylin accumulation in the brain. The level of oligomerized amylin in supernatant fractions and pellets from brain homogenates is almost double in HIP rats compared with WT littermates (P < 0.05). Large amylin deposits (>50 μm diameter) were also occasionally seen in HIP rat brains. Accumulation of oligomerized amylin alters the brain structure at the molecular level. Immunohistochemistry analysis with an ED1 antibody indicates possible activated microglia/macrophages which are clustering in areas positive for amylin infiltration. Multiple inflammatory markers are expressed in HIP rat brains as opposed to WT rats, confirming that amylin deposition in the brain induces a neuroinflammatory response.

Conclusions

Hyperamylinemia promotes accumulation of oligomerized amylin in the brain leading to neurological deficits through an oligomerized amylin-mediated inflammatory response. Additional studies are needed to determine whether brain amylin accumulation may predispose to diabetic brain injury and cognitive decline.

【 授权许可】

   
2014 Srodulski et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20150320092044198.pdf 1466KB PDF download
Figure 6. 64KB Image download
Figure 5. 74KB Image download
Figure 4. 64KB Image download
Figure 3. 56KB Image download
Figure 2. 52KB Image download
Figure 1. 57KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

Figure 6.

【 参考文献 】
  • [1]Luchsinger JA: Type 2 diabetes and cognitive impairment: linking mechanisms. J Alzheimers Dis 2012, 30(2):S185-198.
  • [2]Craft S: The role of metabolic disorders in Alzheimer disease and vascular dementia: two roads converged. Arch Neurol 2009, 66(3):300-305.
  • [3]Nelson PT, Smith CD, Abner EA, Schmitt FA, Scheff SW, Davis GJ, Keller JN, Jicha GA, Davis D, Wang-Xia W, Hartman A, Katz DG, Markesbery WR: Human cerebral neuropathology of Type 2 diabetes mellitus. Biochim Biophys Acta 2009, 1792(5):454-469.
  • [4]Ott A, Stolk RP, Van Harskamp F, Pols HA, Hofman A, Breteler MM: Diabetes mellitus and the risk of dementia: The Rotterdam Study. Neurology 1999, 53(9):1937-1942.
  • [5]Jackson K, Barisone G, Jin L-W, DeCarli C, Despa F: Amylin deposition in the brain: A second amyloid in Alzheimer disease? Ann Neurol 2013, 74(4):517-526.
  • [6]Kahn SE, D'Alessio DA, Schwartz MW, Fujimoto WY, Ensinck JW, Taborsky GJ Jr, Porte D Jr: Evidence of cosecretion of islet amyloid polypeptide and insulin by beta-cells. Diabetes 1990, 39(5):634-638.
  • [7]Westermark P, Andersson A, Westermark GT: Islet amyloid polypeptide, islet amyloid, and diabetes mellitus. Physiol Rev 2011, 91(3):795-826.
  • [8]Molina JM, Cooper GJS, Leighton B, Olefsky JM: Induction of insulin resistance in vivo by amylin and calcitonin gene-related peptide. Diabetes 1990, 39(2):260-265.
  • [9]Leighton B, Cooper GJS: Pancreatic amylin and calcitonin gene-related peptide causes resistance to insulin in skeletal muscle in vitro. Nature 1988, 355(6191):632-635.
  • [10]Zierath JR, Galuska D, Engstrom A, Johnson KH, Betsholtz C, Westermark P, Wallberg-Henriksson H: Human islet amyloid polypeptide at pharmacological levels inhibits insulin and phorbol ester-stimulated glucose transport in vitro incubated human muscle strips. Diabetologia 1992, 35(1):26-31.
  • [11]Cooper ME, McNally PG, Phillips PA, Johnston CI: Amylin stimulates plasma renin concentration in humans. Hypertension 1995, 26(3):460-464.
  • [12]Westfall TC, Curfman-Falvey M: Amylin-induced relaxation of the perfused mesenteric arterial bed: meditation by calcitonin gene-related peptide receptors. J Cardiovasc Pharmacol 1995, 26(6):932-936.
  • [13]Edvinsson L, Goadsby PJ, Uddman R: Amylin: localization, effects on cerebral arteries and on local cerebral blood flow in the cat. Scientific World J 2001, 1(1):168-180.
  • [14]Banks WA, Kastin AJ: Differential permeability of the blood–brain barrier to two pancreatic peptides: insulin and amylin. Peptides 1998, 19(5):883-889.
  • [15]Baldo BA, Kelley AE: Amylin infusion into rat nucleus accumbens potently depresses motor activity and ingestive behavior. Am J Physiol Regul Integr Comp Physiol 2001, 281(4):R1232-R1242.
  • [16]Potes CS, Lutz TA, Riediger T: Identification of central projections from amylin-activated neurons to the lateral hypothalamus. Brain Res 2010, 1334:31-44.
  • [17]Lutz TA: Control of energy homeostasis by amylin. Cell Mol Life Sci 2012, 69(12):1947-1965.
  • [18]Beaumont K, Kenney MA, Young AA, Rink TJ: High affinity amylin binding sites in rat brain. Mol Pharmacol 1993, 44(3):493-497.
  • [19]Zhu H, Wang X, Wallack M, Li H, Carreras I, Dedeoglu A, Hur JY, Zheng H, Li H, Fine R, Mwamburi M, Sun X, Kowall N, Stern RA, Qiu WQ: Intraperitoneal injection of the pancreatic peptide amylin potently reduces behavioral impairment and brain amyloid pathology in murine models of Alzheimer's disease. Mol Psychiatry 2014. doi:10.1038/mp.2014.17
  • [20]Höppener JWM, Ahren B, Lips CJM: Islet amyloid and type 2 diabetes mellitus. N Engl J Med 2000, 343(6):411-419.
  • [21]Westermark P: Quantitative studies on amyloid in the islets of Langerhans. Ups J Med Sci 1972, 77(2):91-94.
  • [22]Bell ET: Hyalinization of the islets of Langerhans in nondiabetic individuals. Am J Pathol 1959, 35(4):801-805.
  • [23]Jurgens CA, Toukatly MN, Fligner CL, Udayasankar J, Subramanian SL, Zraika S, Aston-Mourney K, Carr DB, Westermark P, Westermark GT, Kahn SE, Hull RL: β-cell loss and β-cell apoptosis in human type 2 diabetes are related to islet amyloid deposition. Am J Pathol 2011, 178(6):2632-2640.
  • [24]Janson J, Ashley RH, Harrison D, McIntyre S, Butler PC: The mechanism of islet amyloid polypeptide toxicity is membrane disruption by intermediate-sized toxic amyloid particles. Diabetes 1999, 48(3):491-498.
  • [25]Udayasankar J, Kodama K, Hull RL, Zraika S, Aston-Mourney K, Subramanian SL, Tong J, Faulenbach MV, Vidal J, Kahn SE: Amyloid formation results in recurrence of hyperglycaemia following transplantation of human IAPP transgenic mouse islets. Diabetologia 2009, 52(1):145-153.
  • [26]Lorenzo A, Razzaboni B, Weir GC, Yankner BA: Pancreatic islet cell toxicity of amylin associated with type-2 diabetes mellitus. Nature 1994, 368(6473):756-760.
  • [27]Zraika S, Hull RL, Udayasankar J, Aston-Mourney K, Subramanian SL, Kisilevsky R, Szarek WA, Kahn SE: Oxidative stress is induced by islet amyloid formation and time-dependently mediates amyloid-induced beta cell apoptosis. Diabetologia 2009, 52(4):626-635.
  • [28]Janciauskiene S, Ahrén B: Fibrillar islet amyloid polypeptide differentially affects oxidative mechanisms and lipoprotein uptake in correlation with cytotoxicity in two insulin-producing cell lines. Biochem Biophys Res Commun 2000, 267(2):619-625.
  • [29]Westwell-Roper C, Dai DL, Soukhatcheva G, Potter KJ, Van Rooijen N, Ehses JA, Verchere CB: IL-1 blockade attenuates islet amyloid polypeptide-induced proinflammatory cytokine release and pancreatic islet graft dysfunction. J Immunol 2011, 187(5):2755-2765.
  • [30]Matveyenko AV, Butler PC: Islet amyloid polypeptide (IAPP) transgenic rodents as models for Type 2 Diabetes. ILAR J 2006, 47(3):225-233.
  • [31]Huang C-J, Haataja L, Gurlo T, Butler AE, Wu X, Soeller WC, Butler PC: Induction of endoplasmic reticulum stress-induced beta-cell apoptosis and accumulation of polyubiquitinated proteins by human islet amyloid polypeptide. Am J Physiol Endocrinol Metab 2007, 293(6):1656-1662.
  • [32]Aitken JF, Loomes KM, Scott DW, Reddy S, Phillips AR, Prijic G, Fernando C, Zhang S, Broadhurst R, L'Huillier P, Cooper GJ: Tetracycline treatment retards the onset and slows the progression of diabetes in human amylin/islet amyloid polypeptide transgenic mice. Diabetes 2010, 59(1):161-71.
  • [33]Gurlo T, Ryazantsev S, Huang CJ, Yeh MW, Reber HA, Hines OJ, O'Brien TD, Glabe CG, Butler PC: Evidence for proteotoxicity in beta cells in type 2 diabetes: toxic islet amyloid polypeptide oligomers form intracellularly in the secretory pathway. Am J Pathol 2010, 176(2):861-869.
  • [34]Laganowsky A, Liu C, Sawaya MR, Whitelegge JP, Park J, Zhao M, Pensalfini A, Soriaga AB, Landau M, Teng PK, Cascio D, Glabe C, Eisenberg D: Atomic view of a toxic amyloid small oligomer. Science 2012, 335(6073):1228-1231.
  • [35]Enoki S, Mitsukawa T, Takemura J, Nakazato M, Aburaya J, Toshimori H, Matsukara S: Plasma islet amyloid polypeptide levels in obesity, impaired glucose tolerance and non-insulin-dependent diabetes mellitus. Diabetes Res Clin Prac 1992, 15(1):97-102.
  • [36]Johnson KH, O’Brien TD, Jordan K, Westermark P: Impaired glucose tolerance is associated with increased islet amyloid polypeptide (IAPP) immunoreactivity in pancreatic beta cells. Am J Pathol 1989, 135(2):245-250.
  • [37]Despa S, Margulies KB, Chen L, Knowlton AA, Havel PJ, Taegtmeyer H, Bers DM, Despa F: Hyperamylinemia contributes to heart dysfunction in obesity and diabetes, a study in humans and rats. Circ Res 2012, 110(4):598-608.
  • [38]Erickson JR, Pereira L, Wang L, Han G, Ferguson A, Dao K, Copeland RJ, Despa F, Hart GW, Ripplinger CM, Bers DM: Diabetic Hyperglycemia activates CaMKII and Arrhythmias by O linked Glycosylation. Nature 2013, 502(7471):372-376.
  • [39]Gong W, Liu ZH, Zeng CH, Peng A, Chen HP, Zhou H, Li LS: Amylin deposition in the kidney of patients with diabetic nephropathy. Kidney Int 2007, 72(2):213-218.
  • [40]Westermark P, Engström U, Johnson KH, Westermark GT, Betsholtz C: Islet amyloid polypeptide: pinpointing amino acid residues linked to amyloid fibril formation. Proc Natl Acad Sci U S A 1990, 87(13):5036-5040.
  • [41]Park SW, Goodpaster BH, Strotmeyer ES, Kuller LH, Broudeau R, Kammerer C, De Rekeneire N, Harris TB, Schwartz AV, Tylavsky FA, Cho Y-W: Newman AB for the Health, Aging, and Body Composition Study: Accelerated loss of skeletal muscle strength in older adults with type 2 diabetes: the Health, Aging, and Body Composition Study. Diabetes Care 2007, 30(6):1507-1512.
  • [42]Biessels GJ, Gispen WH: The impact of diabetes on cognition: what can be learned from rodent models? Neurobiol Aging 2005, 26(1):36-41.
  • [43]Craft S, Watson GS: Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol 2004, 3(3):169-178.
  • [44]Mattson MP, Goodman Y: Different amyloidogenic peptides share a similar mechanism of neurotoxicity involving reactive oxygen species and calcium. Brain Res 1995, 676(1):219-224.
  • [45]Kawahara M, Kuroda Y, Arispe N, Rojas E: Alzheimer’s beta-amyloid, human islet amylin, and prion protein fragment evoke intracellular free calcium elevations by a common mechanism in a hypothalamic GnRH neuronal cell line. J Biol Chem 2000, 275(19):14077-14083.
  • [46]Hu R, Zhang M, Patel K, Wang Q, Chang Y, Gong X, Zhang G, Zheng J: Cross-sequence interactions between human and rat islet amyloid polypeptides. Langmuir 2014, 30(18):5193-5201.
  • [47]Middleton CT, Marek P, Cao P, Chiu CC, Singh S, Woys AM, De Pablo JJ, Raleigh DP, Zanni MT: Two-dimensional infrared spectroscopy reveals the complex behaviour of an amyloid fibril inhibitor. Nat Chem 2012, 4(5):355-360.
  • [48]Bachstetter AD, Rowe RK, Kaneko M, Goulding D, Lifshitz J, Van Eldik LJ: The p38alpha MAPK regulates microglial responsiveness to diffuse traumatic brain injury. J Neurosci 2013, 33(14):6143-6153.
  • [49]Khroyan TV, Zhang J, Yang LY, Zou B, Xie JJ, Pascual C, Malik A, Xie JZ, Zaveri NT, Vazquez J, Polgar W, Toll L, Fang J, Xie XM: Rodent Motor and Neuropsychological Behavior Measured in Home Cages Using the Integrated Modular Platform – SmartCageTM. Clin Exp Pharmacol Physiol 2012, 39(7):614-622.
  • [50]Ennaceur A, Delacour J: A new one-trial test for neurobiological studies of memory in rats. 1; Behavioral data. Behav Brain Res 1988, 31(1):47-59.
  • [51]Schoch KM, Evans HN, Brelsfoard JM, Madathil SK, Takano J, Saido TC, Saatman KE: Calpastatin overexpression limits calpain-mediated proteolysis and behavioral deficits following traumatic brain injury. Exp Neurol 2012, 6(2):371-82.
  文献评价指标  
  下载次数:10次 浏览次数:5次