Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar
Reach Us +44 1223 790975

GET THE APP

Journal of Alzheimers Disease & Parkinsonism - A Novel TREM2-Mediated Link between Diabetes and Cognitive Impairment: Recent Findings and Future Perspectives
ISSN: 2161-0460

Journal of Alzheimers Disease & Parkinsonism
Open Access

Like us on:

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)

A Novel TREM2-Mediated Link between Diabetes and Cognitive Impairment: Recent Findings and Future Perspectives

Masashi Tanaka, Hajime Yamakage, Takayuki Inoue, Shinya Masuda, Ryuji Ohue-Kitano, Toru Kusakabe and Noriko Satoh-Asahara*
Department of Endocrinology, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, 1-1 Fukakusa Mukaihata-cho, Fushimi-ku, Kyoto 612-8555, Japan
*Corresponding Author: Noriko Satoh-Asahara, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan, Tel: +81-75-641-9161, Fax: +81-75- 645-2781, Email: nsatoh@kuhp.kyoto-u.ac.jp

Received: 25-Sep-2017 / Accepted Date: 26-Sep-2017 / Published Date: 03-Oct-2017 DOI: 10.4172/2161-0460.1000380

Abstract

The protein triggering receptor expressed on myeloid cell 2 (TREM2) is a cell surface receptor exclusively expressed on microglia in the brain. Recent extensive studies reveal that the functions of TREM2 as well as the phenotypes of TREM2-expressing microglia are closely implicated in the pathogenesis of neurodegenerative diseases including Alzheimer’s disease (AD), one of the most common causes of dementia. Furthermore, several recent studies reported a possible novel pathological implication for TREM2 in cognitive impairment associated with diabetes, one of the several metabolic diseases that are reported risk factors of dementia. In this review, we summarize recent advances and future research directions on the pathophysiological significance of TREM2 in AD and other neurodegenerative diseases as well as in metabolic diseases with increased risk for dementia.

Keywords: Alzheimer’s disease; Cognitive impairment; Diabetes; Metabolic disease; Microglia; Neurodegenerative disease; TREM2

Abbreviations

ADAM10: A Disintegrin and Metalloproteinase Domain-containing Protein 10; AD: Alzheimer’s Disease; CSF: Cerebrospinal Fluid; sTREM2: Soluble Triggering Receptor Expressed on Myeloid Cell 2; TREM2: Triggering Receptor Expressed on Myeloid Cell 2; VaD: Vascular Dementia

Introduction

 

The prevalence of dementia is expanding worldwide, in conjunction with the increase in life expectancy. Dementia is a serious global health issue due to the associated disability and dependence and therefore is a significant economic, social and public health burden [1,2]. Development of predictive markers and effective treatments for dementia is thus urgently needed.

Recent epidemiological studies reported diabetes as a risk factor for dementia, including Alzheimer’s disease (AD) and vascular dementia (VaD), as well as cardiovascular diseases [2-4]. Possible underlying mechanisms of diabetes-related dementia include multifactorial pathways implicated in hyperglycemic toxicity, microvascular diseases, chronic inflammatory processes, and changes in insulin metabolism, which ultimately lead to small vessel infarcts, neuroinflammation, amyloid-β accumulation and neurodegeneration in the brain [2- 5]. Conversely, obesity has also been implicated to play a role in the development of dementia in later life [6]; however, a recent study reported an inverse monotonic association between body mass index (BMI) and dementia incidence [7]. Thus, the potential role of obesity in the development of dementia remains controversial. Although dementia might be prevented or at least delayed in some cases, factors exhibiting preventive effects on dementia remain to be elucidated [8].

Genome-wide association studies recently revealed novel significant associations between variants of the gene triggering receptor expressed on myeloid cell 2 (TREM2) and a high risk for AD and other neurodegenerative diseases [9-12]. Following these initial observations, an exponentially growing number of basic and clinical studies have focused on the pathological roles of TREM2 in the development and progression of dementia. More recently, we reported the first study delineating the pathological implications of serum TREM2 in cognitive impairment in non-obese type 2 diabetic patients [13]. Here, we review the characteristics and recent findings regarding TREM2 and discuss the possibility of TREM2 as a novel target to predict and treat cognitive dysfunction.

Structure, Function and Features of TREM2

TREM2 is a 230-amino-acid V-type immunoglobulin domaincontaining transmembrane protein consisting of an extracellular domain, a transmembrane domain, and a short cytosolic tail lacking an obvious amino acid-based signaling motif [14]. TREM2 is exclusively expressed on myeloid lineage cells including dendritic cells, tissue macrophages, osteoclasts, and microglia [11,12]. Many potential ligands for TREM2 have been proposed, which are characterized based on the type of anionic and/or lipidic species, such as bacterial components, mammalian cellular membrane components and lipoproteins [15]. TREM2 interacts with the adaptor protein DNAX-activating protein 12 (DAP12)/TYRO protein kinase binding protein (TYROBP) via its transmembrane domain. Following binding to its ligands, TREM2 facilitates DAP12 phosphorylation on tyrosine residues within its immunoreceptor tyrosine-based activation motif region, thereby mediating downstream signaling that leads to various cellular functions including survival, phagocytosis and inflammation [10- 12,16]. One remarkable characteristic of TREM2 is the release of its soluble ectodomain fragment, soluble TREM2 (sTREM2), into the extracellular space via proteolytic cleavage at the site in the juxtamembrane region by shaddases such as a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) [17-20]. Although not much is known about the pathophysiological significance of sTREM2 in comparison with TREM2, recent studies demonstrated several novel aspects of sTREM2, which are reviewed in the next section.

Emerging Implications of TREM2 and STREM2 in Neurodegenerative Diseases

Pathophysiological significance of TREM2 and sTREM2 in AD and other neurodegenerative diseases

TREM2 is selectively expressed on microglia in the brain, the main cell type responsible for maintaining brain homeostasis that also plays a role in inflammatory response. Accordingly, recent studies reporting that the R47H TREM2 mutation was associated with an approximately 3-fold increase in AD risk in humans [9-12] had a deep impact and ripple effect based on the possibility that TREM2 and TREM2-expressing microglia might be novel key targets for elucidating mechanisms underlying AD pathogenesis. Recent accumulating evidence reveals an association between a diverse array of TREM2 variants and risk for AD and other neurodegenerative diseases [10-12]. These variants include mutations affecting TREM2 structure/function such as the generation of a truncated protein (W44X or W78X variants) [21], inability to associate with its intracellular adaptor, DAP12/TYROBP (K186N variant) [21], reduction in ligand binding ability (R47H variant) [22- 24], alteration of subcellular localization (reduction on cell surface and increase in endoplasmic reticulum in T66M or Y38C variants) [18,25] and accelerated proteolytic loss from the cell surface (H157Y variant) [19,20]. Therefore, TREM2-related microglial dysfunction can potentially lead to the impairment of brain homeostasis including amyloid-β clearance, possibly leading to neuronal injury and cognitive dysfunction. Therefore, further characterization of TREM2 will allow us to gain a better understanding of its pathological roles in neurodegenerative diseases.

It is increasingly evident that microglia phenotypes are much more complex than previously thought, irrespective of whether they express wildtype or variant TREM2. TREM2 is assumed to exhibit anti-inflammatory roles, mainly based on in vitro analyses; however, recent growing evidence highlights the pro-inflammatory roles of TREM2 in in vivo disease settings [12] and suggest pathological implications of TREM2-expressing microglia in neuroinflammation and concomitant neurodegeneration, with a shift in microglial phenotypes from homeostasis to disease states.

sTREM2 is detected in human blood and cerebrospinal fluid (CSF), and CSF sTREM2 levels are elevated in patients with neurodegenerative diseases compared to healthy controls [18,26-30]. Whereas CSF sTREM2 is a topic of great interest as a potential marker for neurodegenerative diseases, the pathophysiological significance of serum blood sTREM2 remains unclear. Additionally, the function of sTREM2 has not been elucidated, although elevated sTREM2 levels in CSF have been suggested to reflect microglial activation in response to neuronal degeneration [27-31]. In this respect, recent studies uncovered sTREM2 as not just an inactive end-product but also a signaling molecule [15] that promotes macrophage survival by preventing apoptosis [32] and activates microglia to ultimately trigger inflammatory responses and prolong survival [33]. These findings hint at the pathological implications of sTREM2 in chronic inflammation, and further elucidation of the pathophysiological roles of CSF and blood sTREM2 will provide significant novel clues on the regulation of central and systemic inflammation by targeting sTREM2 and/or sTREM2-related processes such as proteolytic production.

Pathophysiological significance of TREM2 and sTREM2 in metabolic diseases with increased risk for neurodegenerative diseases

Not much is known about the pathophysiological significance of TREM2 and sTREM2 in cognitive impairment in patients with metabolic diseases, despite studies demonstrating metabolic diseases as risk factors for AD and VaD. In mouse models of insulin resistance/ diabetes, an early study showed elevated TREM2 expression in mesenteric adipose tissue [34]. Further, activated TREM2-expressing microglia/monocytes were found to accumulate in hippocampus in response to a high-fat diet in aging mice and in a mouse model of AD [35]. In humans, a recent study reported that serum sTREM2 levels were associated with the exacerbation of glucose/lipid metabolism in diabetic conditions [36]. However, there are currently no studies elucidating the pathophysiological roles of TREM2 and sTREM2 in cognitive impairment in patients with metabolic diseases.

We recently provided the first report that addressed these issues in a cross-sectional approach using a database of a multicenter prospective observational cohort study [13]. Our analysis revealed that cognitive function was more exacerbated in non-obese type 2 diabetic patients compared to obese type 2 diabetic patients. In addition, whereas serum sTREM2 levels did not differ between the two patient groups, the elevation in serum sTREM2 levels was significantly associated with the risk of cognitive impairment in non-obese diabetic patients. Importantly, no such significant association was found in obese diabetic patients in whom elevated systolic blood pressure was associated with an increased risk of cognitive impairment. Moreover, we found that serum sTREM2 levels were positively correlated with diabetes-associated risk factors of dementia, including hyperglycemia and exacerbation of inflammation, and negatively correlated with adiponectin level in nonobese diabetic patients. However, these correlations were not observed in obese diabetic patients. Overall, these findings suggest that serum sTREM2 levels might be a potential novel marker of the diabetesassociated cognitive impairment in non-obese diabetic patients. Additional studies are critical to elucidate the mechanisms underlying the relationship between sTREM2 and cognitive dysfunction in diabetic patients. In this context, our recent findings provide potential novel strategies for reducing the risk of cognitive impairment and reveal that serum sTREM2 levels might have utility as a marker in prevention and early management hyperglycemia and aggravation of inflammation in non-obese diabetic patients. In addition, early control of systolic blood pressure might be effective in obese diabetic patients.

Future Perspectives

Recent remarkable advances in TREM2 research led to novel insights into the pathological significance of TREM2 and sTREM2 in AD and other neurodegenerative diseases as well as in metabolic diseases with increased risk for neurodegenerative diseases. These revelations further hint at the possible implications of TREM2 and sTREM2 in other cognitive dysfunction-causing diseases such as cerebral amyloid angiopathy [37]. Accordingly, future findings are expected to give prominence to the significance of TREM2 and sTREM2 as key targets in prediction, prevention and/or improvement of cognitive dysfunction.

Emerging pathological roles of TREM2 and sTREM2 raise new issues that need to be addressed. One instance is the mechanistic details underlying the phenotypic shift in microglia from an antiinflammatory to a pro-inflammatory state, which might trigger chronic neuroinflammation and account for inconsistencies reported regarding the phenotypes of TREM2-expressing microglia. Another important issue is the elucidation of the mechanisms regulating ADAM10- mediated sTREM2 release under steady-state and disease conditions. ADAM10 is also involved in amyloid precursor protein processing to release amyloid-β and ADAM10 mutations confer increased risk of AD [38,39]. Elucidation of the effect of ADAM10 mutations on sTREM2 production as well as the impact of TREM2-expressing microglia on their pro-inflammatory status and subsequent sTREM2 release should reveal novel pathological implications of ADAM10 in TREM2- expressing microglia-associated neuroinflammation.

Recent findings suggest several directions for future research regarding potential factors affecting microglial phenotypes. Microglial maturation and function were reported to be influenced by gut microbiota-derived metabolites, albeit through unidentified pathways [40]. In addition, the diversity and stability of gut microbiota were shown to alter in conjunction with aging or metabolic diseases such as diabetes [41,42]. These alterations in gut microbiota in turn increase the permeability of gut and blood-brain barriers, thereby triggering systemic and central inflammation and leading to the development of neurodegenerative diseases [42,43]. Accordingly, it remains possible that TREM2 might function as a receptor for gut microbiota-derived molecules during aging or metabolic diseases and might modulate microglial phenotypes to push towards a pro-inflammatory axis. In this respect, gut microbiota-targeted intervention might contribute to TREM2-mediated improvement of microglial phenotypes.

Conclusion

Recent research on TREM2 has provided a deeper understanding of the mechanisms underlying the development and progression of cognitive dysfunction in patients with AD and other neurodegenerative diseases as well as metabolic diseases with increased risk for neurodegenerative diseases. Future basic and clinical research elucidating the pathophysiological significance of TREM2 will reveal new avenues for preemptive medicine for dementia.

Acknowledgement

We wish to thank Mr. Kazuya Muranaka at Kyoto Medical Center for his technical assistance.

Funding

 

This work was supported in part by a Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan to M.T. (grant number 16K09276), H.Y. (16K08959), T.I. (16H07498), S.M. (15K08211), T.K. (17K09827), and N.S.-A. (15K08634); grants from Takeda Science Foundation to M.T.; grants from Japan Research Foundation for Clinical Pharmacology to M.T.; grants from Kao Research Council for the Study of Healthcare Science to M.T.; and grants from the National Hospital Organization for collaborative clinical research to N.S.-A. (H26-NHO-02). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  1. Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, et al. (2013) The global prevalence of dementia: A systematic review and metaanalysis. Alzheimers Dement 9: 63-75.
  2. Chatterjee S, Peters SA, Woodward M, Mejia Arango S, Batty GD, et al. (2016) Type 2 diabetes as a risk factor for dementia in women compared with men: A pooled analysis of 2.3 million people comprising more than 100,000 cases of dementia. Diabetes Care 39: 300-307.
  3. Ohara T, Doi Y, Ninomiya T, Hirakawa Y, Hata J, et al. (2011) Glucose tolerance status and risk of dementia in the community: the Hisayama study. Neurology 77: 1126-1134.
  4. Morley JE (2017) Diabetes: The diabetic brain. Nat Rev Endocrinol 13: 570-571.
  5. Hirabayashi N, Hata J, Ohara T, Mukai N, Nagata M, et al. (2016) Association between diabetes and hippocampal atrophy in elderly japanese: The Hisayama study. Diabetes Care 39: 1543-1549.
  6. Kivipelto M, Ngandu T, Fratiglioni L, Viitanen M, KÃ¥reholt I, et al. (2005) Obesity and vascular risk factors at midlife and the risk of dementia and Alzheimer disease. Arch Neurol 62: 1556-1560.
  7. Qizilbash N, Gregson J, Johnson ME, Pearce N, Douglas I, et al. (2015) BMI and risk of dementia in two million people over two decades: A retrospective cohort study. Lancet Diabetes Endocrinol 3: 431-436.
  8. Satizabal CL, Beiser AS, Chouraki V, Chêne G, Dufouil C, et al. (2016) Incidence of dementia over three decades in the Framingham heart study. N Engl J Med 374: 523-532.
  9. Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, et al. (2013) TREM2 variants in Alzheimer's disease. N Engl J Med 368: 117-127.
  10. Yaghmoor F, Noorsaeed A, Alsaggaf S, Aljohani W, Scholtzova H, et al. (2014) The role of TREM2 in alzheimer's disease and other neurological disorders. J Alzheimers Dis Parkinsonism 4: 160.
  11. Yeh FL, Hansen DV, Sheng M (2017) TREM2, microglia and neurodegenerative diseases. Trends Mol Med 23: 512-533.
  12. Jay TR, von Saucken VE, Landreth GE (2017) TREM2 in neurodegenerative diseases. Mol Neurodegener 12: 56.
  13. Tanaka M, Yamakage H, Masuda S, Inoue T, Ohue-Kitano R, et al. (2017) Serum soluble TREM2 is a potential novel biomarker of cognitive impairment in Japanese non-obese patients with diabetes. Diabetes Metab pii: S1262-3636(17)30470-6.
  14. Bouchon A, Dietrich J, Colonna M (2000) Cutting edge: Inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol 164: 4991-4995.
  15. Kober DL, Brett TJ (2017) TREM2-Ligand interactions in health and disease. J Mol Biol 429: 1607-1629.
  16. Ulrich JD, Ulland TK, Colonna M, Holtzman DM (2017) Elucidating the Role of TREM2 in Alzheimer's disease. Neuron 94: 237-248.
  17. Wunderlich P, Glebov K, Kemmerling N, Tien NT, Neumann H, et al. (2013) Sequential proteolytic processing of the triggering receptor expressed on myeloid cells-2 (TREM2) protein by ectodomain shedding and γ-secretase-dependent intramembranous cleavage. J Biol Chem 288: 33027-33036.
  18. Kleinberger G, Yamanishi Y, Suárez-Calvet M, Czirr E, Lohmann E, et al. (2014) TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci Transl Med 6: 243ra86.
  19. Schlepckow K, Kleinberger G, Fukumori A, Feederle R, Lichtenthaler SF, et al. (2017) An Alzheimer-associated TREM2 variant occurs at the ADAM cleavage site and affects shedding and phagocytic function. EMBO Mol Med pii: e201707672.
  20. Thornton P, Sevalle J, Deery MJ, Fraser G, Zhou Y, et al. (2017) TREM2 shedding by cleavage at the H157-S158 bond is accelerated for the Alzheimer's disease-associated H157Y variant. EMBO Mol Med pii: e201707673.
  21. Paloneva J, Manninen T, Christman G, Hovanes K, Mandelin J, et al. (2002) Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am J Hum Genet 71: 656-662.
  22. Bailey CC, DeVaux LB, Farzan M. (2015) The triggering receptor expressed on myeloid cells 2 binds apolipoprotein E. J Biol Chem 290: 26033-26042.
  23. Yeh FL, Wang Y, Tom I, Gonzalez LC, Sheng M (2016) TREM2 binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facilitates uptake of amyloid-beta by microglia. Neuron 91: 328-340.
  24. Kober DL, Alexander-Brett JM, Karch CM, Cruchaga C, Colono M, et al. (2016) Neurodegenerative disease mutations in TREM2 reveals a functional surface and distinct loss-of-function mechanisms. Elife 5: e20391.
  25. Sirkis DW, Bonham LW, Aparicio RE, Geier EG, Ramos EM, et al. (2016) Rare TREM2 variants associated with Alzheimer's disease display reduced cell surface expression. Acta Neuropathol Commun 4: 98.
  26. Piccio L, Buonsanti C, Cella M, Tassi I, Schmidt RE, et al. (2008) Identification of soluble TREM-2 in the cerebrospinal fluid and its association with multiple sclerosis and CNS inflammation. Brain 131: 3081-3091.
  27. Piccio L, Deming Y, Del-Águila JL, Ghezzi L, Holtzman DM, et al. (2016) Cerebrospinal fluid soluble TREM2 is higher in Alzheimer disease and associated with mutation status. Acta Neuropathol 131: 925-933.
  28. Suárez-Calvet M, Kleinberger G, Araque Caballero MÁ, Brendel M, Rominger A, et al. (2016) sTREM2 cerebrospinal fluid levels are a potential biomarker for microglia activity in early-stage Alzheimer's disease and associate with neuronal injury markers. EMBO Mol Med 8: 466-476.
  29. Henjum K, Almdahl IS, Ã…rskog V, Minthon L, Fladby T, et al. (2016) Cerebrospinal fluid soluble TREM2 in aging and Alzheimer's disease. Alzheimers Res Ther 8: 17.
  30. Öhrfelt A, Axelsson M, Malmeström C, Novakova L, Heslegrave A, et al. (2016) Soluble TREM-2 in cerebrospinal fluid from patients with multiple sclerosis treated with natalizumab or mitoxantrone. Mult Scler 22: 1587-1595.
  31. Heslegrave A, Heywood W, Paterson R, Magdalinou N, Svensson J, et al. (2016) Increased cerebrospinal fluid soluble TREM2 concentration in Alzheimer's disease. Mol Neurodegener 11: 3.
  32. Wu K, Byers DE, Jin X, Agapov E, Alexander-Brett J, et al. (2015) TREM-2 promotes macrophage survival and lung disease after respiratory viral infection. J Exp Med 212: 681-697.
  33. Zhong L, Chen XF, Wang T, Wang Z, Liao C, et al. (2017) Soluble TREM2 induces inflammatory responses and enhances microglial survival. J Exp Med 214: 597-607.
  34. Fujimoto S, Goda T, Mochizuki K (2011) in vivo evidence of enhanced di-methylation of histone H3 K4 on upregulated genes in adipose tissue of diabetic db/db mice. Biochem Biophys Res Commun 404: 223-227.
  35. Graham LC, Harder JM, Soto I, de Vries WN, John SW, et al. (2016) Chronic consumption of a western diet induces robust glial activation in aging mice and in a mouse model of Alzheimer's disease. Sci Rep 6: 21568.
  36. Subramanian S, Pallati PK, Rai V, Sharma P, Agrawal DK, et al. (2017) Increased expression of triggering receptor expressed on myeloid cells-1 in the population with obesity and insulin resistance. Obesity (Silver Spring) 25: 527-538.
  37. Saito S, Ihara M (2014) New therapeutic approaches for Alzheimer's disease and cerebral amyloid angiopathy. Front Aging Neurosci 6: 290.
  38. Kim M, Suh J, Romano D, Truong MH, Mullin K, et al. (2009) Potential late-onset Alzheimer's disease-associated mutations in the ADAM10 gene attenuate {alpha}-secretase activity. Hum Mol Genet 18: 3987-3996.
  39. Suh J, Choi SH, Romano DM, Gannon MA, Lesinski AN, et al. (2013) ADAM10 missense mutations potentiate ß-amyloid accumulation by impairing prodomain chaperone function. Neuron 80: 385-401.
  40. Fung TC, Olson CA, Hsiao EY (2017) Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci 20: 145-155.
  41. Sonnenburg JL, Bäckhed F (2016) Diet-microbiota interactions as moderators of human metabolism. Nature 535: 56-64.
  42. Alkasir R, Li J, Li X, Jin M, Zhu B, et al. (2017) Human gut microbiota: The links with dementia development. Protein Cell 8: 90-102.
  43. Jiang C, Li G, Huang P, Liu Z, Zhao B (2017) The gut microbiota and Alzheimer’s disease. J Alzheimers Dis 58: 1-15.

Citation: Tanaka M, Yamakage H, Inoue T, Masuda S, Ohue-Kitano R, et al. (2017) A Novel TREM2-Mediated Link between Diabetes and Cognitive Impairment: Recent Findings and Future Perspectives. J Alzheimers Dis Parkinsonism 7: 380. DOI: 10.4172/2161-0460.1000380

Copyright: © 2017 Tanaka M, 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.

https://bahigox.fun/ https://bahisjet.fun/ https://bahsegel.fun/ https://betboo.fun/ https://betdoksan.xyz/ https://betebetgiris.xyz/ https://betexper.fun/ https://betgram.fun/ https://betkanyon.fun/ https://betkolik.fun/
Top