Chronic low-level expression of HIV-1 Tat promotes a neurodegenerative phenotype with aging

Alex M Dickens, Seung Wan Yoo, Alfred C Chin, Jiadi Xu, Tory P Johnson, Amanda L Trout, Kurt F Hauser, Norman J Haughey, Alex M Dickens, Seung Wan Yoo, Alfred C Chin, Jiadi Xu, Tory P Johnson, Amanda L Trout, Kurt F Hauser, Norman J Haughey

Abstract

The widespread use of combinational antiretroviral therapies (cART) in developed countries has changed the course of Human Immunodeficiency Virus (HIV) infection from an almost universally fatal disease to a chronic infection for the majority of individuals. Although cART has reduced the severity of neurological damage in HIV-infected individuals, the likelihood of cognitive impairment increases with age, and duration of infection. As cART does not suppress the expression of HIV non-structural proteins, it has been proposed that a constitutive production of HIV regulatory proteins in infected brain cells may contribute to neurological damage. However, this assumption has never been experimentally tested. Here we take advantage of the leaky tetracycline promoter system in the Tat-transgenic mouse to show that a chronic very low-level expression of Tat is associated with astrocyte activation, inflammatory cytokine expression, ceramide accumulation, reductions in brain volume, synaptic, and axonal damage that occurs over a time frame of 1 year. These data suggest that a chronic low-level production of Tat may contribute to progressive neurological damage in virally suppressed HIV-infected individuals.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Brain volume loss is observed in doxycycline naïve rtTA-Tat mice at 12 months of age. qRT-PCR analysis showing expression of tat mRNA in cortex of (a) 3-month old control rtTA mice, and doxycycline naïve rtTA-Tat mice, and (b) 11–12 month old doxycycline treated control rtTA mice, doxycycline naïve rtTA-Tat mice, and doxycycline treated rtTA-Tat mice. (c) Tat protein expression in cortex of 11–12 month old doxycycline treated control rtTA mice, doxycycline naïve rtTA-Tat mice, and doxycycline treated rtTA-Tat mice. (d) Representative T2 weighted images from 3–5 and 11–12 month old mice of the indicated genotype and treatment conditions showing enlargement of ventricles in 11–12 month old doxycycline naïve and doxycycline treated rtTA-Tat mice compare with doxycycline treated rtTA control mice. (e,f) Bar graphs show volumetric quantification of the ventricle, hippocampus, dentate gyrus, striatum, and motor cortex of 3–5 month animals and (g,h) of 11–12 month animals. (i) Representative fiber tracking maps of the corpus callosum calculated from the diffusion weighted MRI images. Quantification of the fractional anisotropy (j) and parallel eigenvalue (k) the diffusion weighted images in 11–12 month old animals. Data are mean ± SEM of n = 6–7 animals/group. ANOVA with Tukey post hoc comparisons, *p < 0.05 compared to control, ***p < 0.001 compared to rtTA control.
Figure 2
Figure 2
Evidence for axonal and synaptic damage in doxycycline naïve rtTA-Tat mice. Immunoblots and density quantitation of (a) the axonal marker βIII-tubulin, (b) the presynaptic marker Synaptophysin, and (c) the postsynaptic marker PSD95 in cortex of 11–12 month old mice with the indicated genotype and treatment condition. (d) qRT-PCR analysis showing expression of the indicated cytokines in cortex of 11–12 month old mice with the indicated genotype and treatment condition. Data are expressed as mean ± SEM. N = 3 mice per condition for western blots, and n = 6–7 per group for qRT-PCR. ***p < 0.001, **p < 0.01, *p < 0.05, and #p < 0.05 compared to rtTA control group.
Figure 3
Figure 3
Accumulation of ceramide in cortex of rtTA-Tat mice. Concentrations of (a) dihydroceramides, (b) ceramides, (c) monohexosylceramides, and (d) dihexosylceramides, were determined in cortex of mice with the indicated genotypes. Hierarchal clustering analysis is shown to the left of each class of lipid. Blue colors indicates increase and red colors indicates decrease compared to rtTA control mice. Data are expressed as mean ± SEM, n = 6–7 per group. Bold text in table indicates p < 0.05 compared to rtTA control group. ANOVA with Tukey post hoc comparisons.
Figure 4
Figure 4
Accumulation of sphingomyelin in cortex of rtTA-Tat mice. Concentrations of sphingomyelins were determined in cortex of mice with the indicated genotypes. Hierarchal clustering analysis is shown to the left of each class of lipid. Blue colors indicates increase and red colors indicates decrease compared to rtTA control mice. Data are expressed as mean ± SEM, n = 6–7 per group. Bold text in table indicates p < 0.05 and italicized entries indicates p < 0.001 compared to rtTA control group. ANOVA with Tukey post hoc comparisons.
Figure 5
Figure 5
The increase in dentate gyrus volume is linked to an increase in reactive astrocytes in this brain region. (a) Representative images and (b) quantitative analysis of GFAP fluorescence from the dentate gyrus of 11–12 month old mice with the indicated genotypes. (c) qRT-PCR analysis showing expression of the indicated cytokines in hippocampus of 11–12 month old mice with the indicated genotype and treatment condition. Data are expressed as mean ± SEM for n = 6–7 per group. * p < 0.05, **p < 0.01. ANOVA with Tukey post hoc comparisons.

References

    1. Heaton R, et al. HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy CHARTER Study. Neurology. 2010;75:2087–2096. doi: 10.1212/WNL.0b013e318200d727.
    1. Heaton R, et al. HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. Journal of NeuroVirology. 2011;17:3–16. doi: 10.1007/s13365-010-0006-1.
    1. Cysique LA, Maruff P, Brew BJ. Prevalence and pattern of neuropsychological impairment in human immunodeficiency virus-infected/acquired immunodeficiency syndrome (HIV/AIDS) patients across pre-and post-highly active antiretroviral therapy eras: A combined study of two cohorts Clinical Report. Journal of Neurovirology. 2004;10:350–357. doi: 10.1080/13550280490521078.
    1. Tozzi V, et al. Persistence of neuropsychologic deficits despite long-term highly active antiretroviral therapy in patients with HIV-related neurocognitive impairment: prevalence and risk factors. Journal of acquired immune deficiency syndromes. 2007;45:174–182. doi: 10.1097/QAI.0b013e318042e1ee.
    1. Simioni S, et al. Cognitive dysfunction in HIV patients despite long-standing suppression of viremia. Aids. 2010;24:1243–1250.
    1. Kaul M, Garden GA, Lipton SA. Pathways to neuronal injury and apoptosis in HIV-associated dementia. Nature Genetics. 2001;2:988–994.
    1. Valcour V, et al. Central nervous system viral invasion and inflammation during acute HIV infection. J Infect Dis. 2012;206:275–282. doi: 10.1093/infdis/jis326.
    1. Spudich S, et al. Central nervous system immune activation characterizes primary human immunodeficiency virus 1 infection even in participants with minimal cerebrospinal fluid viral burden. The Journal of infectious diseases. 2011;204:753–760. doi: 10.1093/infdis/jir387.
    1. Ragin AB, et al. Brain alterations within the first 100 days of HIV infection. Annals of clinical and translational neurology. 2015;2:12–21. doi: 10.1002/acn3.136.
    1. Ragin AB, et al. Structural brain alterations can be detected early in HIV infection. Neurology. 2012;79:2328–2334. doi: 10.1212/WNL.0b013e318278b5b4.
    1. Wang X, et al. Abnormalities in resting-state functional connectivity in early human immunodeficiency virus infection. Brain connectivity. 2011;1:207–217. doi: 10.1089/brain.2011.0016.
    1. Chang L, et al. Persistent brain abnormalities in antiretroviral-naive HIV patients 3 months after HAART. Antiviral therapy. 2003;8:17–26.
    1. Hestad K, et al. Regional brain atrophy in HIV‐1 infection: association with specific neuropsychological test performance. Acta Neurologica Scandinavica. 1993;88:112–118. doi: 10.1111/j.1600-0404.1993.tb04201.x.
    1. Jernigan TL, et al. Clinical factors related to brain structure in HIV: the CHARTER study. Journal of neurovirology. 2011;17:248–257. doi: 10.1007/s13365-011-0032-7.
    1. Cohen RA, et al. Effects of nadir CD4 count and duration of human immunodeficiency virus infection on brain volumes in the highly active antiretroviral therapy era. Journal of neurovirology. 2010;16:25–32. doi: 10.3109/13550280903552420.
    1. Paul R, Cohen R, Navia B, Tashima K. Relationships between cognition and structural neuroimaging findings in adults with human immunodeficiency virus type-1. Neuroscience & Biobehavioral Reviews. 2002;26:353–359. doi: 10.1016/S0149-7634(02)00006-4.
    1. Paul RH, et al. Relative sensitivity of magnetic resonance spectroscopy and quantitative magnetic resonance imaging to cognitive function among nondemented individuals infected with HIV. Journal of the International Neuropsychological Society. 2008;14:725–733. doi: 10.1017/S1355617708080910.
    1. Garden GA. Microglia in human immunodeficiency virus-associated neurodegeneration. Glia. 2002;40:240–251. doi: 10.1002/glia.10155.
    1. Churchill MJ, et al. Extensive astrocyte infection is prominent in human immunodeficiency virus–associated dementia. Annals of neurology. 2009;66:253–258. doi: 10.1002/ana.21697.
    1. Saito Y, et al. Overexpression of nef as a marker for restricted HIV‐1 infection of astrocytes in postmortem pediatric central nervous tissues. Neurology. 1994;44:474–474. doi: 10.1212/WNL.44.3_Part_1.474.
    1. Brack-Werner R. Astrocytes: HIV cellular reservoirs and important participants in neuropathogenesis. AIDS. 1999;13:1–22. doi: 10.1097/00002030-199901140-00003.
    1. Neumann M, et al. Restriction of human immunodeficiency virus type 1 production in a human astrocytoma cell line is associated with a cellular block in Rev function. Journal of virology. 1995;69:2159–2167.
    1. Tornatore C, Meyers K, Atwood W, Conant K, Major E. Temporal patterns of human immunodeficiency virus type 1 transcripts in human fetal astrocytes. Journal of virology. 1994;68:93–102.
    1. Conant K, et al. In vivo and in vitro infection of the astrocyte by HIV-1. Adv Neuroimmunol. 1994;4:287–289. doi: 10.1016/S0960-5428(06)80269-X.
    1. Nath A. Human immunodeficiency virus (HIV) proteins in neuropathogenesis of HIV dementia. The Journal of infectious diseases. 2002;186(Suppl 2):S193–198. doi: 10.1086/344528.
    1. van Marle G, et al. Human immunodeficiency virus type 1 Nef protein mediates neural cell death: a neurotoxic role for IP-10. Virology. 2004;329:302–318. doi: 10.1016/j.virol.2004.08.024.
    1. Chompre G, et al. Astrocytic expression of HIV-1 Nef impairs spatial and recognition memory. Neurobiology of disease. 2013;49:128–136. doi: 10.1016/j.nbd.2012.08.007.
    1. Sabatier JM, et al. Evidence for neurotoxic activity of tat from human immunodeficiency virus type 1. Journal of virology. 1991;65:961–967.
    1. Haughey NJ, Holden CP, Nath A, Geiger JD. Involvement of inositol 1,4,5-trisphosphate-regulated stores of intracellular calcium in calcium dysregulation and neuron cell death caused by HIV-1 protein tat. J Neurochem. 1999;73:1363–1374. doi: 10.1046/j.1471-4159.1999.0731363.x.
    1. Ensoli B, et al. Release, uptake, and effects of extracellular human immunodeficiency virus type 1 Tat protein on cell growth and viral transactivation. Journal of virology. 1993;67:277–287.
    1. Chauhan A, et al. Intracellular human immunodeficiency virus Tat expression in astrocytes promotes astrocyte survival but induces potent neurotoxicity at distant sites via axonal transport. J Biol Chem. 2003;278:13512–13519. doi: 10.1074/jbc.M209381200.
    1. Fitting S, et al. Interactive comorbidity between opioid drug abuse and HIV-1 Tat: chronic exposure augments spine loss and sublethal dendritic pathology in striatal neurons. Am J Pathol. 2010;177:1397–1410. doi: 10.2353/ajpath.2010.090945.
    1. Fitting S, et al. Synaptic dysfunction in the hippocampus accompanies learning and memory deficits in human immunodeficiency virus type-1 Tat transgenic mice. Biological psychiatry. 2013;73:443–453. doi: 10.1016/j.biopsych.2012.09.026.
    1. Flexner C. HIV drug development: the next 25 years. Nat Rev Drug Discov. 2007;6:959–966. doi: 10.1038/nrd2336.
    1. Duncan MJ, et al. Effects of chronic expression of the HIV-induced protein, transactivator of transcription, on circadian activity rhythms in mice, with or without morphine. Am J Physiol Regul Integr Comp Physiol. 2008;295:R1680–1687. doi: 10.1152/ajpregu.90496.2008.
    1. Kim BO, et al. Neuropathologies in transgenic mice expressing human immunodeficiency virus type 1 Tat protein under the regulation of the astrocyte-specific glial fibrillary acidic protein promoter and doxycycline. Am J Pathol. 2003;162:1693–1707. doi: 10.1016/S0002-9440(10)64304-0.
    1. Haughey NJ, et al. Converging roles for sphingolipids and cell stress in the progression of neuro-AIDS. Front Biosci. 2008;13:5120–5130. doi: 10.2741/3068.
    1. Mathias S, et al. Activation of the sphingomyelin signaling pathway in intact EL4 cells and in a cell-free system by IL-1 beta. Science. 1993;259:519–522. doi: 10.1126/science.8424175.
    1. Figuera-Losada M, et al. Cambinol, a novel inhibitor of neutral sphingomyelinase 2 shows neuroprotective properties. PLoS One. 2015;10:e0124481. doi: 10.1371/journal.pone.0124481.
    1. Friis-Møller N, et al. Combination antiretroviral therapy and the risk of myocardial infarction. N Engl J Med. 2003;349:1993–2003. doi: 10.1056/NEJMoa030218.
    1. Sulkowski MS, Thomas DL, Chaisson RE, Moore RD. Hepatotoxicity associated with antiretroviral therapy in adults infected with human immunodeficiency virus and the role of hepatitis C or B virus infection. JAMA. 2000;283:74–80. doi: 10.1001/jama.283.1.74.
    1. Saylor D, et al. HIV-associated neurocognitive disorder - pathogenesis and prospects for treatment. Nature Reviews. Neurology. 2016;12:234–U220.
    1. Heaton RK, et al. Neurocognitive change in the era of HIV combination antiretroviral therapy: the longitudinal CHARTER study. Clinical Infectious Diseases. 2015;60:473–480. doi: 10.1093/cid/ciu862.
    1. Kelly SG, et al. Early suppressive antiretroviral therapy in HIV infection is associated with measurable changes in the corpus callosum. Journal of neurovirology. 2014;20:514–520. doi: 10.1007/s13365-014-0261-7.
    1. Li S, et al. Matrix metalloproteinase levels in early HIV infection and relation to in vivo brain status. Journal of neurovirology. 2013;19:452–460. doi: 10.1007/s13365-013-0197-3.
    1. Burdo TH, et al. Soluble CD163 made by monocyte/macrophages is a novel marker of HIV activity in early and chronic infection prior to and after anti-retroviral therapy. Journal of Infectious Diseases. 2011;204:154–163. doi: 10.1093/infdis/jir214.
    1. Sailasuta, N. et al. Change in brain magnetic resonance spectroscopy after treatment during acute HIV infection (2012).
    1. Kore I, et al. Neuropsychological Impairment in Acute HIV and the Effect of Immediate Antiretroviral Therapy. J Acquir Immune Defic Syndr. 2015;70:393–399. doi: 10.1097/QAI.0000000000000746.
    1. Dickens AM, et al. Cerebrospinal fluid metabolomics implicate bioenergetic adaptation as a neural mechanism regulating shifts in cognitive states of HIV-infected patients. AIDS. 2015;29:559–569.
    1. Stout JC, et al. Progressive cerebral volume loss in human immunodeficiency virus infection: a longitudinal volumetric magnetic resonance imaging study. HIV Neurobehavioral Research Center Group. Archives of neurology. 1998;55:161–168. doi: 10.1001/archneur.55.2.161.
    1. Becker JT, et al. Subcortical brain atrophy persists even in HAART-regulated HIV disease. Brain imaging and behavior. 2011;5:77–85. doi: 10.1007/s11682-011-9113-8.
    1. Thompson PM, et al. Thinning of the cerebral cortex visualized in HIV/AIDS reflects CD4+ T lymphocyte decline. Proc Natl Acad Sci USA. 2005;102:15647–15652. doi: 10.1073/pnas.0502548102.
    1. Behrman-Lay AM, et al. Human immunodeficiency virus has similar effects on brain volumetrics and cognition in males and females. J Neurovirol. 2016;22:93–103. doi: 10.1007/s13365-015-0373-8.
    1. Guha A, et al. Intrinsic network connectivity abnormalities in HIV-infected individuals over age 60. J Neurovirol. 2016;22:80–87. doi: 10.1007/s13365-015-0370-y.
    1. Ortega M, Brier MR, Ances BM. Effects of HIV and combination antiretroviral therapy on cortico-striatal functional connectivity. AIDS. 2015;29:703–712. doi: 10.1097/QAD.0000000000000611.
    1. Campbell JH, Hearps AC, Martin GE, Williams KC, Crowe SM. The importance of monocytes and macrophages in HIV pathogenesis, treatment, and cure. AIDS. 2014;28:2175–2187. doi: 10.1097/QAD.0000000000000408.
    1. Walsh JG, et al. Rapid inflammasome activation in microglia contributes to brain disease in HIV/AIDS. Retrovirology. 2014;11:35. doi: 10.1186/1742-4690-11-35.
    1. Schuenke K, Gelman BB. Human microglial cell isolation from adult autopsy brain: brain pH, regional variation, and infection with human immunodeficiency virus type 1. Journal of neurovirology. 2003;9:346–357. doi: 10.1080/13550280390201056.
    1. Brack-Werner R, et al. Infection of human brain cells by HIV-1: restricted virus production in chronically infected human glial cell lines. Aids. 1992;6:273–285. doi: 10.1097/00002030-199203000-00004.
    1. Tardieu M, Héry C, Peudenier S, Boespflug O, Montagnier L. Human immunodeficiency virus type 1-infected monocytic cells can destroy human neural cells after cell-to-cell adhesion. Ann Neurol. 1992;32:11–17. doi: 10.1002/ana.410320104.
    1. Thompson KA, McArthur JC, Wesselingh SL. Correlation between neurological progression and astrocyte apoptosis in HIV-associated dementia. Annals of neurology. 2001;49:745–752. doi: 10.1002/ana.1011.
    1. Messam CA, Major EO. Stages of restricted HIV-1 infection in astrocyte cultures derived from human fetal brain tissue. Journal of neurovirology. 2000;6(Suppl 1):S90–94.
    1. Churchill MJ, et al. Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia. Annals of neurology. 2009;66:253–258. doi: 10.1002/ana.21697.
    1. Kim HJ, Martemyanov KA, Thayer SA. Human immunodeficiency virus protein Tat induces synapse loss via a reversible process that is distinct from cell death. The. Journal of neuroscience: the official journal of the Society for Neuroscience. 2008;28:12604–12613. doi: 10.1523/JNEUROSCI.2958-08.2008.
    1. Zhou BY, Liu Y, Kim B, Xiao Y, He JJ. Astrocyte activation and dysfunction and neuron death by HIV-1 Tat expression in astrocytes. Mol Cell Neurosci. 2004;27:296–305. doi: 10.1016/j.mcn.2004.07.003.
    1. Pu H, et al. HIV-1 Tat protein upregulates inflammatory mediators and induces monocyte invasion into the brain. Molecular and cellular neurosciences. 2003;24:224–237. doi: 10.1016/S1044-7431(03)00171-4.
    1. Haughey NJ, et al. Perturbation of sphingolipid metabolism and ceramide production in HIV-dementia. Ann Neurol. 2004;55:257–267. doi: 10.1002/ana.10828.
    1. Nath A, et al. Identification of a human immunodeficiency virus type 1 Tat epitope that is neuroexcitatory and neurotoxic. J Virol. 1996;70:1475–1480.
    1. Haughey NJ, Nath A, Mattson MP, Slevin JT, Geiger JD. HIV-1 Tat through phosphorylation of NMDA receptors potentiates glutamate excitotoxicity. J Neurochem. 2001;78:457–467. doi: 10.1046/j.1471-4159.2001.00396.x.
    1. Hui, L., Chen, X., Haughey, N. J. & Geiger, J. D. Role of endolysosomes in HIV-1 Tat-induced neurotoxicity. ASN neuro4 (2012).
    1. Carey AN, Sypek EI, Singh HD, Kaufman MJ, McLaughlin JP. Expression of HIV-Tat protein is associated with learning and memory deficits in the mouse. Behav Brain Res. 2012;229:48–56. doi: 10.1016/j.bbr.2011.12.019.
    1. Carey AN, et al. Conditional Tat protein expression in the GT-tg bigenic mouse brain induces gray matter density reductions. Prog Neuropsychopharmacol Biol Psychiatry. 2013;43:49–54. doi: 10.1016/j.pnpbp.2012.12.018.
    1. Bruce-Keller AJ, et al. Morphine causes rapid increases in glial activation and neuronal injury in the striatum of inducible HIV-1 Tat transgenic mice. Glia. 2008;56:1414–1427. doi: 10.1002/glia.20708.
    1. Gelbard HA, et al. Neurotoxic effects of tumor necrosis factor alpha in primary human neuronal cultures are mediated by activation of the glutamate AMPA receptor subtype: implications for AIDS neuropathogenesis. Dev Neurosci. 1993;15:417–422. doi: 10.1159/000111367.
    1. Bal-Price A, Brown GC. Inflammatory neurodegeneration mediated by nitric oxide from activated glia-inhibiting neuronal respiration, causing glutamate release and excitotoxicity. J Neurosci. 2001;21:6480–6491.
    1. Flora G, et al. Proinflammatory synergism of ethanol and HIV-1 Tat protein in brain tissue. Experimental neurology. 2005;191:2–12. doi: 10.1016/j.expneurol.2004.06.007.
    1. El-Hage N, et al. Morphine exacerbates HIV-1 Tat-induced cytokine production in astrocytes through convergent effects on [Ca(2+)](i), NF-kappaB trafficking and transcription. PloS one. 2008;3:e4093. doi: 10.1371/journal.pone.0004093.
    1. Hahn YK, et al. beta-Chemokine production by neural and glial progenitor cells is enhanced by HIV-1 Tat: effects on microglial migration. J Neurochem. 2010;114:97–109.
    1. Bermel RA, Bakshi R. The measurement and clinical relevance of brain atrophy in multiple sclerosis. Lancet Neurol. 2006;5:158–170. doi: 10.1016/S1474-4422(06)70349-0.
    1. Rudick RA. Impact of disease-modifying therapies on brain and spinal cord atrophy in multiple sclerosis. J Neuroimaging. 2004;14:54S–64S. doi: 10.1111/j.1552-6569.2004.tb00279.x.
    1. Chen JT, et al. Brain atrophy after immunoablation and stem cell transplantation in multiple sclerosis. Neurology. 2006;66:1935–1937. doi: 10.1212/01.wnl.0000219816.44094.f8.
    1. Ngwainmbi J, et al. Effects of HIV-1 Tat on enteric neuropathogenesis. The. Journal of neuroscience: the official journal of the Society for Neuroscience. 2014;34:14243–14251. doi: 10.1523/JNEUROSCI.2283-14.2014.
    1. Guedia J, et al. HIV-1 Tat exacerbates lipopolysaccharide-induced cytokine release via TLR4 signaling in the enteric nervous system. Scientific reports. 2016;6:31203. doi: 10.1038/srep31203.
    1. Wall R, et al. Bacterial neuroactive compounds produced by psychobiotics. Advances in experimental medicine and biology. 2014;817:221–239. doi: 10.1007/978-1-4939-0897-4_10.
    1. Hahn YK, et al. Effects of chronic HIV-1 Tat exposure in the CNS: heightened vulnerability of males versus females to changes in cell numbers, synaptic integrity, and behavior. Brain structure & function. 2015;220:605–623. doi: 10.1007/s00429-013-0676-6.
    1. Carey AN, et al. Conditional Tat protein brain expression in the GT-tg bigenic mouse induces cerebral fractional anisotropy abnormalities. Curr HIV Res. 2015;13:3–9. doi: 10.2174/1570162X13666150126125244.
    1. Chakos MH, et al. Striatal enlargement in rats chronically treated with neuroleptic. Biol Psychiatry. 1998;44:675–684. doi: 10.1016/S0006-3223(98)00029-8.
    1. Johnson TP, et al. Induction of IL-17 and nonclassical T-cell activation by HIV-Tat protein. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:13588–13593. doi: 10.1073/pnas.1308673110.
    1. Bandaru VVR, et al. A lipid storage–like disorder contributes to cognitive decline in HIV-infected subjects. Neurology. 2013;81:1492–1499. doi: 10.1212/WNL.0b013e3182a9565e.
    1. Mielke MM, et al. Demographic and clinical variables affecting mid- to late-life trajectories of plasma ceramide and dihydroceramide species. Aging Cell. 2015;14:1014–1023. doi: 10.1111/acel.12369.
    1. Mielke MM, et al. Factors affecting longitudinal trajectories of plasma sphingomyelins: the Baltimore Longitudinal Study of Aging. Aging cell. 2015;14:112–121. doi: 10.1111/acel.12275.

Source: PubMed

3
Abonnere