Neurofilament light: a possible prognostic biomarker for treatment of vascular contributions to cognitive impairment and dementia

Christina Hoyer-Kimura, John P Konhilas, Heidi M Mansour, Robin Polt, Kristian P Doyle, Dean Billheimer, Meredith Hay, Christina Hoyer-Kimura, John P Konhilas, Heidi M Mansour, Robin Polt, Kristian P Doyle, Dean Billheimer, Meredith Hay

Abstract

Background: Decreased cerebral blood flow and systemic inflammation during heart failure (HF) increase the risk for vascular contributions to cognitive impairment and dementia (VCID) and Alzheimer disease-related dementias (ADRD). We previously demonstrated that PNA5, a novel glycosylated angiotensin 1-7 (Ang-(1-7)) Mas receptor (MasR) agonist peptide, is an effective therapy to rescue cognitive impairment in our preclinical model of VCID. Neurofilament light (NfL) protein concentration is correlated with cognitive impairment and elevated in neurodegenerative diseases, hypoxic brain injury, and cardiac disease. The goal of the present study was to determine (1) if treatment with Ang-(1-7)/MasR agonists can rescue cognitive impairment and decrease VCID-induced increases in NfL levels as compared to HF-saline treated mice and, (2) if NfL levels correlate with measures of cognitive function and brain cytokines in our VCID model.

Methods: VCID was induced in C57BL/6 male mice via myocardial infarction (MI). At 5 weeks post-MI, mice were treated with daily subcutaneous injections for 24 days, 5 weeks after MI, with PNA5 or angiotensin 1-7 (500 microg/kg/day or 50 microg/kg/day) or saline (n = 15/group). Following the 24-day treatment protocol, cognitive function was assessed using the Novel Object Recognition (NOR) test. Cardiac function was measured by echocardiography and plasma concentrations of NfL were quantified using a Quanterix Simoa assay. Brain and circulating cytokine levels were determined with a MILLIPLEX MAP Mouse High Sensitivity Multiplex Immunoassay. Treatment groups were compared via ANOVA, significance was set at p < 0.05.

Results: Treatment with Ang-(1-7)/MasR agonists reversed VCID-induced cognitive impairment and significantly decreased NfL levels in our mouse model of VCID as compared to HF-saline treated mice. Further, NfL levels were significantly negatively correlated with cognitive scores and the concentrations of multiple pleiotropic cytokines in the brain.

Conclusions: These data show that treatment with Ang-(1-7)/MasR agonists rescues cognitive impairment and decreases plasma NfL relative to HF-saline-treated animals in our VCID mouse model. Further, levels of NfL are significantly negatively correlated with cognitive function and with several brain cytokine concentrations. Based on these preclinical findings, we propose that circulating NfL might be a candidate for a prognostic biomarker for VCID and may also serve as a pharmacodynamic/response biomarker for therapeutic target engagement.

Keywords: Angiotensin-(1–7); Biomarker; Inflammation; Neurofilament light (NfL); PNA5; Vascular contributions to cognitive impairment and dementia.

Conflict of interest statement

COI, Dr. Meredith Hay is the founder of ProNeurogen, Inc. that holds exclusive rights to the clinical development of the Ang-(1–7)/MasR agonists described herein.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Experiment timeline. Three-month-old male mice were acclimated for one week before MI or Control surgeries. 5 weeks following recovery, mice were treated with Saline, Ang-(1–7) or PNA5 at 2 concentrations (50 and 500 ug/kg) for 24 days. During the last 3 days of treatment mice underwent Novel Object Recognition testing. Following Novel Object Recognition testing, mice underwent echocardiographs before killing. At killing, the plasma, brain, and heart were collected. MI myocardial infarct, S.C. subcutaneous injection
Fig. 2
Fig. 2
Echocardiograms and histology to confirm myocardial infarctions in the heart failure model. A Simpson Ejection Fraction was calculated using V-mode left ventricle calculations treatment groups: heart failure (HF)-A50 (Ang-(1–7)50 ug/kg), HF-P50 (PNA5 50ug/kg), HF-A500 (Ang-(1–7) 500 ug/kg), HF-P500 (PNA5 500 ug/kg), HF-Saline, Control-Saline. Significant difference between Control-Saline and the following treatment groups are as listed: HF-Saline, HF-A50 (Ang-(1–7)50 ug/kg), HF-P50 (PNA5 50 ug/kg), HF-A500 (Ang-(1–7)500 ug/kg), HF-P500 (PNA5 500 ug/kg), P < 0.05 ANOVA, Dunnett’s post. B Simpson End Systolic Volume was calculated using V-mode left ventricle calculation. Significant difference between Control-Saline and the following treatment groups are as listed: HF-Saline p = 0.001, HF-P50 (PNA5 50 ug/kg) p = 0.0001, and HF-A500 (Ang-(1–7)500 ug/kg) p = 0.015, ANOVA, Dunnett’s post. C Examples of hematoxylin and eosin staining of paraffin embedded formalin fixed hearts demonstrates differences in wall morphology between Control animals and HF-saline, PNA5, and Ang-(1–7) treatment mice. Left ventricle wall is indicated with black arrow. Images were taken at 5 × magnification
Fig. 3
Fig. 3
Ang-(1–7)/MasR agonists rescue cognitive impairment in a VCID mice. A Discrimination ratio = (Time spent at new object- Time spent at familiar object)/ total exploration time. Heart failure (HF) mice were treated with saline PNA5 or Ang-(1–7) at two different concentrations of either 50ug/kg, (P50, A50) or 500ug/kg (P500, A500). ****p < 0.001 Control-Saline vs HF-Saline. *p < 0.05 HF-Saline vs HF-P50, HF-P500, HF-A500. Data are represented with mean and ± SE. Difference in groups were tested by ANOVA, Dunnett’s post, significance p < 0.05. B No significant differences are noted in total exploration time in the familiarization test for all treatment groups. Data are represented with mean and ± SE. Significancy were tested by ANOVA
Fig. 4
Fig. 4
Treatment with Ang-(1–7)/Mas R agonists decreases plasma NfL levels. A Data are represented with mean and ± SE. Compares NfL levels between control saline and HF-saline mice, p = 0.08, Welch’s t-test. This difference did not reach significance. B PNA5 at 50ug/kg and Ang-(1–7) at 50 ug/kg, and 500 ug/kg significantly reduced NfL plasma concentration in VCID mice compared to HF-saline treated mice, Kruskal–Wallis ANOVA, Dunn’s post, p < 0.05. C NfL plasma levels significantly negatively correlated with Discrimination ratios. Correlation R values were produced using Pearson’s correlation analysis and the fit line was determined using simple linear regression
Fig. 5
Fig. 5
Increased TNFα in VCID models are inhibited by PNA5 and Ang-(1–7). Plasma TNFα of Control-Saline, HF-Saline, HF-A50 (Ang-(1–7)50 ug/kg), HF-P50 (PNA5 50ug/kg), HF-A500 (Ang-(1–7)500ug/kg), HF-P500 (PNA5 500ug/kg), were measured via MILLIPLEX MAP Mouse High Sensitivity Multiplex Immunoassay. Significance were observed between HF-Saline vs Control-Saline treated mice (3.50± SE 0.15, n = 6 vs mean 3.12, ± SE 0.01, n = 6, respectively, p = 0.003;), PNA5, and Ang-(1–7) (HF-P50 mean 3.12 pg/mL± SE 0.00, n = 5 p = 0.004; HF-A50 mean 3.12 pg/mL± SE 0.00, n = 6, p = 0.003; HF-P500 mean 3.12 pg/mL± SE 0.00, n = 6, p = 0.003; ANOVA, Dunnett’s post; values that were at or below assay threshold of 3.12 pg/mL were represented as 3.12 pg/mL)
Fig. 6
Fig. 6
Effect of treatment with Ang-(1–7)/MasR agonists on brain cytokine levels. Whole brain lysate cytokine levels were measured using MILLIPLEX MAP Mouse High Sensitivity Multiplex Immunoassay. AF HF-Saline treated VCID mice had significantly lower IL-1α, IL-2, IL-17, and IP-10 levels in comparison to Control-Saline treated mice. PNA5 and Ang-(1–7) significantly increased cytokines levels as compared to HF-saline treated animals. Data are represented with mean and ± SE. Differences in groups were tested by ANOVA, Dunnett’s post hoc, significance set a p < 0.05
Fig. 7
Fig. 7
Relationship between brain cytokine levels and NfL levels. AE The relationship between brain cytokines IL-2, IL-5, IL-13, IL-17, IP-10, and plasma NfL levels were analyzed using Pearson correlation. F Brain cytokine composite score of IL-2, IL-13, IL-17 were significantly negatively correlated with NfL plasma concentrations
Fig. 8
Fig. 8
Pearson’s correlation matrix of the relationship between plasma NfL and brain cytokine levels. The relationship between the brain cytokines IL-2, IL-5, IL-13, IL-17, IP-10, the composite cytokine z-score and plasma NfL levels were analyzed using Pearson correlation. Blue demonstrates positive correlation and red represents negative correlation. Values closer to 1 or -1 indicate more closely correlated variables
Fig. 9
Fig. 9
Longitudinal comparison of plasma cytokines in VCID mouse model. AC Plasma inflammatory cytokine levels from mice with 3 weeks of HF, and 8 weeks of HF, in both Control-Saline, and HF-Saline treated mice were measured via MILLIPLEX MAP Mouse High Sensitivity Multiplex Immunoassay. At 3 weeks of HF, HF-Saline mice had significantly higher IL-1α, MIP-1α, and MIP-2 concentrations than age-matched Control-Saline treated mice. A significant decrease in cytokine concentrations was observed in HF-Saline-treated mice from 3 to 8 weeks of HF. Data are represented with mean and ± SE. Difference in groups were tested by ANOVA

References

    1. Qiu C, Winblad B, Marengoni A, Klarin I, Fastbom J, Fratiglioni L. Heart failure and risk of dementia and Alzheimer disease: A population-based cohort study. Arch Intern Med. 2006;166(9):1003–1008. doi: 10.1001/archinte.166.9.1003.
    1. Audia C, Santos Y, Snyder PJ, Wu W-C, Zhang M, Echeverria A, et al. Pathophysiologic relationship between Alzheimer’s disease, cerebrovascular disease, and cardiovascular risk: a review and synthesis. Alzheimers Dement. 2017 doi: 10.1016/j.dadm.2017.01.005.
    1. Corriveau RA, Bosetti F, Emr M, Gladman JT, Koenig JI, Moy CS, et al. The science of vascular contributions to cognitive impairment and dementia (vcid): a framework for advancing research priorities in the cerebrovascular biology of cognitive decline. Cell Mol Neurobiol. 2016;36:281–288. doi: 10.1007/s10571-016-0334-7.
    1. Mp M, Ra C, Dm W. Vascular contributions to cognitive impairment and dementia (VCID) Biochim Biophys Acta. 2016;1862(5):857–859. doi: 10.1016/j.bbadis.2016.02.010.
    1. Rodgers KE, Oliver J, DiZerega GS. Phase I/II dose escalation study of angiotensin 1–7 [A(1–7)] administered before and after chemotherapy in patients with newly diagnosed breast cancer. Cancer Chemother Pharmacol. 2006;57(5):559–568. doi: 10.1007/s00280-005-0078-4.
    1. Hay M, Vanderah TW, Samareh-Jahani F, Constantopoulos E, Uprety AR, Barnes CA, et al. Cognitive impairment in heart failure: a protective role for angiotensin-(1–7) Behav Neurosci. 2017;131(1):99–114. doi: 10.1037/bne0000182.
    1. Hay M, Polt R, Heien ML, Vanderah TW, Largent-Milnes TM, Rodgers K, et al. A novel angiotensin-(1–7) glycosylated mas receptor agonist for treating vascular cognitive impairment and inflammation-related memory dysfunction. J Pharmacol Exp Ther. 2019;369:9–25. doi: 10.1124/jpet.118.254854.
    1. Jiang T, Yu JT, Zhu XC, Zhang QQ, Tan MS, Cao L, et al. Angiotensin-(1–7) induces cerebral ischaemic tolerance by promoting brain angiogenesis in a Mas/eNOS-dependent pathway. Br J Pharmacol. 2014;171(18):4222–4232. doi: 10.1111/bph.12770.
    1. Ferrario CM. Angiotensin-converting enzyme 2 and angiotensin-(1–7): an evolving story in cardiovascular regulation. Hypertension. 2006 doi: 10.1161/01.HYP.0000196268.08909.fb.
    1. Zetterberg H. Neurofilament light: a dynamic cross-disease fluid biomarker for neurodegeneration. Neuron. 2016 doi: 10.1016/j.neuron.2016.06.030.
    1. Ashton NJ, Leuzy A, Lim YM, Troakes C, Hortobágyi T, Höglund K, et al. Increased plasma neurofilament light chain concentration correlates with severity of post-mortem neurofibrillary tangle pathology and neurodegeneration. Acta Neuropathol Commun. 2019;7(1):5. doi: 10.1186/s40478-018-0649-3.
    1. Gaetani L, Salvadori N, Lisetti V, Eusebi P, Mancini A, Gentili L, et al. Cerebrospinal fluid neurofilament light chain tracks cognitive impairment in multiple sclerosis. J Neurol. 2019;266(9):2157–2163. doi: 10.1007/s00415-019-09398-7.
    1. Villar LM, Picón C, Costa-Frossard L, Alenda R, García-Caldentey J, Espiño M, et al. Cerebrospinal fluid immunological biomarkers associated with axonal damage in multiple sclerosis. Eur J Neurol. 2015;22(8):1169–1175. doi: 10.1111/ene.12579.
    1. Thelin E, Al Nimer F, Frostell A, Zetterberg H, Blennow K, Nyström H, et al. A Serum protein biomarker panel improves outcome prediction in human traumatic brain injury. J Neurotrauma. 2019;36(20):2850–2862. doi: 10.1089/neu.2019.6375.
    1. Shahim P, Politis A, van der Merwe A, Moore B, Chou YY, Pham DL, et al. Neurofilament light as a biomarker in traumatic brain injury. Neurology. 2020;95(6):e610–e622. doi: 10.1212/WNL.0000000000009983.
    1. Nielsen HH, Soares CB, Høgedal SS, Madsen JS, Hansen RB, Christensen AA, et al. Acute neurofilament light chain plasma levels correlate with stroke severity and clinical outcome in ischemic stroke patients. Front Neurol. 2020;11:11. doi: 10.3389/fneur.2020.00448.
    1. Wiberg S, Holmgaard F, Blennow K, Nilsson JC, Kjaergaard J, Wanscher M, et al. Associations between mean arterial pressure during cardiopulmonary bypass and biomarkers of cerebral injury in patients undergoing cardiac surgery: secondary results from a randomized controlled trial. Interact Cardiovasc Thorac Surg. 2020 doi: 10.1093/icvts/ivaa264.
    1. Gao XM, Dart AM, Dewar E, Jennings G, Du XJ. Serial echocardiographic assessment of left ventricular dimensions and function after myocardial infarction in mice. Cardiovasc Res. 2000;45(2):330–338. doi: 10.1016/S0008-6363(99)00274-6.
    1. 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. doi: 10.1016/0166-4328(88)90157-X.
    1. Rumberger JA. Ventricular dilatation and remodeling after myocardial infarction. Mayo Clinic Proc. 1994 doi: 10.1016/S0025-6196(12)61345-7.
    1. Kapasi A, Schneider JA. Vascular contributions to cognitive impairment, clinical Alzheimer’s disease, and dementia in older persons. Biochim Biophys Acta Mol Basis Dis. 2016;1862(5):878–886. doi: 10.1016/j.bbadis.2015.12.023.
    1. Toledo JB, Toledo E, Weiner MW, Jack CR, Jagust W, Lee VMY, et al. Cardiovascular risk factors, cortisol, and amyloid-β deposition in Alzheimer’s Disease Neuroimaging Initiative. Alzheimer’s Dement. 2012;8(6):483–489. doi: 10.1016/j.jalz.2011.08.008.
    1. Yarchoan M, Xie SX, Kling MA, Toledo JB, Wolk DA, Lee EB, et al. Cerebrovascular atherosclerosis correlates with Alzheimer pathology in neurodegenerative dementias. Brain. 2012;135(12):3749–3756. doi: 10.1093/brain/aws271.
    1. van Oijen M, de Jong FJ, Witteman JCM, Hofman A, Koudstaal PJ, Breteler MMB. Atherosclerosis and risk for dementia. Ann Neurol. 2007;61(5):403–410. doi: 10.1002/ana.21073.
    1. Gorelick PB, Furie KL, Iadecola C, Smith EE, Waddy SP, Lloyd-Jones DM, et al. Defining optimal brain health in adults: a presidential advisory from the American Heart Association/American Stroke Association. Stroke. 2017;48(10):e284–303. doi: 10.1161/STR.0000000000000148.
    1. Santos CY, Snyder PJ, Wu W-C, Zhang M, Echeverria A, Alber J. Pathophysiologic relationship between Alzheimer’s disease, cerebrovascular disease, and cardiovascular risk: a review and synthesis. Alzheimers Dement. 2017;7(1):69–87.
    1. Canobbio I, Abubaker AA, Visconte C, Torti M, Pula G. Role of amyloid peptides in vascular dysfunction and platelet dysregulation in Alzheimer’s disease. Front Cell Neurosci. 2015 doi: 10.3389/fncel.2015.00065.
    1. Janota C, Lemere CA, Brito MA. Dissecting the Contribution of Vascular Alterations and Aging to Alzheimer’s Disease. Mol Neurobiol. 2015;53(6):3793–3811. doi: 10.1007/s12035-015-9319-7.
    1. Sweeney MD, Montagne A, Sagare AP, Nation DA, Schneider LS, Chui HC, et al. Vascular dysfunction—the disregarded partner of Alzheimer’s disease. Alzheimer’s Dement. 2019;15(1):158–167. doi: 10.1016/j.jalz.2018.07.222.
    1. Helman AM, Murphy MP. Vascular cognitive impairment: Modeling a critical neurologic disease in vitro and in vivo. Biochim Biophys Acta Mol Basis Dis. 2016;1862(5):975–982. doi: 10.1016/j.bbadis.2015.12.009.
    1. Wentzel C, Rockwood K, MacKnight C, Hachinski V, Hogan DB, Feldman H, et al. Progression of impairment in patients with vascular cognitive impairment without dementia. Neurology. 2001;57(4):714–716. doi: 10.1212/WNL.57.4.714.
    1. Hsiung GYR, Donald A, Grand J, Black SE, Bouchard RW, Gauthier SG, et al. Outcomes of cognitively impaired not demented at 2 years in the Canadian cohort study of cognitive impairment and related dementias. Dement Geriatr Cogn Disord. 2006;22(5–6):413–420. doi: 10.1159/000095751.
    1. Mattsson N, Andreasson U, Zetterberg H, Blennow K, Weiner MW, Aisen P, et al. Association of plasma neurofilament light with neurodegeneration in patients with Alzheimer disease. JAMA Neurol. 2017;74(5):557–566. doi: 10.1001/jamaneurol.2016.6117.
    1. Olsson B, Lautner R, Andreasson U, Öhrfelt A, Portelius E, Bjerke M, et al. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol. 2016;15(7):673–684. doi: 10.1016/S1474-4422(16)00070-3.
    1. Petzold A, Keir G, Warren J, Fox N, Rossor MN. A systematic review and meta-analysis of CSF neurofilament protein levels as biomarkers in dementia. Neurodegener Dis. 2007;4(2–3):185–194. doi: 10.1159/000101843.
    1. Rosengren LE, Karlsson JE, Sjögren M, Blennow K, Wallin A. Neurofilament protein levels in CSF are increased in dementia. Neurology. 1999;52(5):1090–1093. doi: 10.1212/WNL.52.5.1090.
    1. Skillbäck T, Farahmand B, Bartlett JW, Rosén C, Mattsson N, Nägga K, et al. CSF neurofilament light differs in neurodegenerative diseases and predicts severity and survival. Neurology. 2014;83(21):1945–1953. doi: 10.1212/WNL.0000000000001015.
    1. Ma W, Zhang J, Xu J, Feng D, Wang X, Zhang F. Elevated levels of serum neurofilament light chain associated with cognitive impairment in vascular dementia. Dis Markers. 2020 doi: 10.1155/2020/6612871.
    1. Cipollini V, Troili F, Giubilei F. Emerging biomarkers in vascular cognitive impairment and dementia: from pathophysiological pathways to clinical application. IJMS. 2019 doi: 10.3390/ijms20112812.
    1. FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools). Silver Spring (MD): Food and Drug Administration (US); 2016-. Available from: Co-published by National Institutes of Health (US), Bethesda (MD).
    1. Kuhle J, Kropshofer H, Haering DA, Kundu U, Meinert R, Barro C, et al. Blood neurofilament light chain as a biomarker of MS disease activity and treatment response. Neurology. 2019;92(10):e1007–e1015. doi: 10.1212/WNL.0000000000007032.
    1. Kapoor R, Smith KE, Allegretta M, Arnold DL, Carroll W, Comabella M, et al. Serum neurofilament light as a biomarker in progressive multiple sclerosis. Neurology. 2020;95(10):436–444. doi: 10.1212/WNL.0000000000010346.
    1. Varhaug KN, Torkildsen Ø, Myhr KM, Vedeler CA. Neurofilament light chain as a biomarker in multiple sclerosis. Front Neurol. 2019 doi: 10.3389/fneur.2019.00338.
    1. Price BR, Norris CM, Sompol P, Wilcock DM. An emerging role of astrocytes in vascular contributions to cognitive impairment and dementia. J Neurochem. 2018;144(5):644–650. doi: 10.1111/jnc.14273.
    1. Price BR, Wilcock DM, Weekman EM. Hyperhomocysteinemia as a risk factor for vascular contributions to cognitive impairment and dementia. Front Aging Neurosci. 2018;31:350. doi: 10.3389/fnagi.2018.00350.
    1. Trollor JN, Smith E, Agars E, Kuan SA, Baune BT, Campbell L, et al. The association between systemic inflammation and cognitive performance in the elderly: The Sydney Memory and Ageing Study. Age (Omaha) 2012;34(5):1295–1308. doi: 10.1007/s11357-011-9301-x.
    1. Bruunsgaard H. A high plasma concentration of TNF-α is associated with dementia in centenarians. J Gerontol Ser A Biol Sci Med Sci. 1999 doi: 10.1093/gerona/54.7.M357.
    1. Yndestad A, Damås JK, Øie E, Yndestad A, et al. Systemic inflammation in heart failure—the whys and wherefores. Hear Fail Rev. 2006;11:83–92. doi: 10.1007/s10741-006-9196-2.
    1. Aukrust P, Ueland T, Lien E, Bendtzen K, Müller F, Andreassen AK, et al. Cytokine network in congestive heart failure secondary to ischemic or idiopathic dilated cardiomyopathy. Am J Cardiol. 1999;83(3):376–382. doi: 10.1016/S0002-9149(98)00872-8.
    1. Salmeron KE, Maniskas ME, Edwards DN, Wong R, Rajkovic I, Trout A, et al. Interleukin 1 alpha administration is neuroprotective and neuro-restorative following experimental ischemic stroke. J Neuroinflammation. 2019;16(1):222. doi: 10.1186/s12974-019-1599-9.
    1. Salmeron K, Aihara T, Redondo-Castro E, Pinteaux E, Bix G. IL-1alpha induces angiogenesis in brain endothelial cells in vitro: Implications for brain angiogenesis after acute injury. J Neurochem. 2016;136(3):573–580. doi: 10.1111/jnc.13422.
    1. Alves S, Churlaud G, Audrain M, Michaelsen-Preusse K, Fol R, Souchet B, et al. Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice. Brain. 2017;140(3):826–842.
    1. Schmitz M, Hermann P, Oikonomou P, Stoeck K, Ebert E, Poliakova T, et al. Cytokine profiles and the role of cellular prion protein in patients with vascular dementia and vascular encephalopathy. Neurobiol Aging. 2015;36(9):2597–2606. doi: 10.1016/j.neurobiolaging.2015.05.013.
    1. Zhou Y, Li C, Li D, Zheng Y, Wang J. IL-5 blocks apoptosis and tau hyperphosphorylation induced by Aβ25–35 peptide in PC12 cells. J Physiol Biochem. 2017;73(2):259–266. doi: 10.1007/s13105-017-0550-8.
    1. Miao W, Zhao Y, Huang Y, Chen D, Luo C, Su W, et al. IL-13 ameliorates neuroinflammation and promotes functional recovery after traumatic brain injury. J Immunol. 2020;204(6):1486–1498. doi: 10.4049/jimmunol.1900909.
    1. Morrison BE, Marcondes MCG, Nomura DK, Sanchez-Alavez M, Sanchez-Gonzalez A, Saar I, et al. Cutting edge: IL-13Rα1 expression in dopaminergic neurons contributes to their oxidative stress-mediated loss following chronic peripheral treatment with lipopolysaccharide. J Immunol. 2012;189(12):5498–5502. doi: 10.4049/jimmunol.1102150.
    1. Cipollini V, Anrather J, Orzi F, Iadecola C. Th17 and cognitive impairment: possible mechanisms of action. Front Neuroanat. 2019 doi: 10.3389/fnana.2019.00095.
    1. Bordon Y. IL-17: an immune mnemonic. Nat Rev Immunol. 2019 doi: 10.1038/s41577-019-0237-1.
    1. McKimmie C, Michlmayr D. Role of CXCL10 in central nervous system inflammation. Int J Interf Cytokine Mediat Res. 2014;6(1):1.
    1. Bettcher BM, Johnson SC, Fitch R, Casaletto KB, Heffernan KS, Asthana S, et al. CSF and plasma levels of inflammation differentially relate to CNS markers of Alzheimer’s disease pathology and neuronal damage hhs public access. J Alzheimers Dis. 2018;62(1):385–397. doi: 10.3233/JAD-170602.

Source: PubMed

3
Suscribir