Increased tau phosphorylation and receptor for advanced glycation endproducts (RAGE) in the brain of mice infected with Leishmania amazonensis

Leishmaniasis is a parasitosis caused by several species of the genus Leishmania, an obligate intramacrophagic parasite. Although neurologic symptoms have been observed in human cases of leishmaniasis, the manifestation of neurodegenerative processes is poorly studied. The aim of the present work was to investigate if peripheral infection of BALB/c mice with Leishmania amazonensis affects tau phosphorylation and RAGE protein content in the brain, which represent biochemical markers of neurodegenerative processes observed in diseases with a pro-inflammatory component, including Alzheimer's disease and Down syndrome. Four months after a single right hind footpad subcutaneous injection of L. amazonensis, the brain cortex of BALB/c mice was isolated. Western blot analysis indicated an increase in tau phosphorylation (Ser(396)) and RAGE immunocontent in infected animals. Brain tissue TNF-α, IL-1β, and IL-6 levels were not different from control animals; however, increased protein carbonylation, decreased IFN-γ levels and impairment in antioxidant defenses were detected. Systemic antioxidant treatment (NAC 20mg/kg, i.p.) inhibited tau phosphorylation and recovered IFN-γ levels. These data, altogether, indicate an association between impaired redox state, tau phosphorylation and RAGE up-regulation in the brain cortex of animals infected with L. amazonensis. In this context, it is possible that neurologic symptoms associated to chronic leishmaniasis are associated to disruptions in the homeostasis of CNS proteins, such as tau and RAGE, as consequence of oxidative stress. This is the first demonstration of alterations in biochemical parameters of neurodegeneration in an experimental model of Leishmania infection.

[1]  R. Badaró,et al.  Leishmaniasis in Bahia, Brazil: evidence that Leishmania amazonensis produces a wide spectrum of clinical disease. , 1991, The American journal of tropical medicine and hygiene.

[2]  M. Barral-Netto,et al.  Hormone levels are associated with clinical markers and cytokine levels in human localized cutaneous leishmaniasis , 2011, Brain, Behavior, and Immunity.

[3]  C. Petersen,et al.  Neurologic Manifestations of Leishmania spp. Infection. , 2011, Journal of neuroparasitology.

[4]  Tilman Grune,et al.  Protein oxidative modification in the aging organism and the role of the ubiquitin proteasomal system. , 2011, Current pharmaceutical design.

[5]  H. Aebi,et al.  Catalase in vitro. , 1984, Methods in enzymology.

[6]  C. R. Alves,et al.  Immunological characteristics of experimental murine infection with Leishmania (Leishmania) amazonensis. , 2008, Veterinary parasitology.

[7]  G. Ellman,et al.  Tissue sulfhydryl groups. , 1959, Archives of biochemistry and biophysics.

[8]  M. C. Brelaz-de-Castro,et al.  Cellular immune response evaluation of cutaneous leishmaniasis patients cells stimulated with Leishmania (Viannia) braziliensis antigenic fractions before and after clinical cure. , 2012, Cellular immunology.

[9]  J. Berman,et al.  Advances in leishmaniasis , 2005, The Lancet.

[10]  E. Lissi,et al.  Luminol luminescence induced by 2,2'-Azo-bis(2-amidinopropane) thermolysis. , 1992, Free radical research communications.

[11]  J. Cano,et al.  Leishmaniasis Worldwide and Global Estimates of Its Incidence , 2012, PloS one.

[12]  E. Kojro,et al.  Regulated proteolysis of RAGE and AbetaPP as possible link between type 2 diabetes mellitus and Alzheimer's disease. , 2009, Journal of Alzheimer's disease : JAD.

[13]  C. Petersen,et al.  Neurologic Manifestations ofLeishmania spp.Infection , 2011 .

[14]  P. Machado,et al.  The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of human tegumentary leishmaniasis☆ , 2014, Cytokine.

[15]  E. Shacter,et al.  Differential susceptibility of plasma proteins to oxidative modification: examination by western blot immunoassay. , 1994, Free radical biology & medicine.

[16]  W. Tourtellotte,et al.  Amyloid-beta peptide-receptor for advanced glycation endproduct interaction elicits neuronal expression of macrophage-colony stimulating factor: a proinflammatory pathway in Alzheimer disease. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[17]  C. Brodskyn,et al.  Toward a Novel Experimental Model of Infection To Study American Cutaneous Leishmaniasis Caused by Leishmania braziliensis , 2005, Infection and Immunity.

[18]  T. Montine,et al.  Free radical-mediated damage to brain in Alzheimer's disease and its transgenic mouse models. , 2008, Free radical biology & medicine.

[19]  P. Nawroth,et al.  Bench-to-bedside review: The inflammation-perpetuating pattern-recognition receptor RAGE as a therapeutic target in sepsis , 2008, Critical care.

[20]  P. Desjeux Leishmaniasis: current situation and new perspectives. , 2004, Comparative immunology, microbiology and infectious diseases.

[21]  Urs Meyer,et al.  Systemic immune challenges trigger and drive Alzheimer-like neuropathology in mice , 2012, Journal of Neuroinflammation.

[22]  G. Perry,et al.  Phosphorylation of tau protein at sites Ser396–404 is one of the earliest events in Alzheimer's disease and Down syndrome , 2014, Neuropathology and applied neurobiology.

[23]  J. Götz,et al.  Insights into mitochondrial dysfunction: aging, amyloid-β, and tau-A deleterious trio. , 2012, Antioxidants & redox signaling.

[24]  I. Grundke‐Iqbal,et al.  Abnormal hyperphosphorylation of tau: sites, regulation, and molecular mechanism of neurofibrillary degeneration. , 2012, Journal of Alzheimer's disease : JAD.

[25]  I. Fridovich,et al.  The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. , 1972, The Journal of biological chemistry.

[26]  T. Suuronen,et al.  Inflammation in Alzheimer's disease: Amyloid-β oligomers trigger innate immunity defence via pattern recognition receptors , 2009, Progress in Neurobiology.

[27]  E. Stadtman,et al.  Determination of carbonyl content in oxidatively modified proteins. , 1990, Methods in enzymology.

[28]  A. Satoskar,et al.  Leishmaniasis: clinical syndromes and treatment. , 2014, QJM : monthly journal of the Association of Physicians.

[29]  H. H. Draper,et al.  Malondialdehyde determination as index of lipid peroxidation. , 1990, Methods in enzymology.

[30]  M. M. Bradford A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. , 1976, Analytical biochemistry.

[31]  Ying-Dong Zhang,et al.  Anti-TNF-α reduces amyloid plaques and tau phosphorylation and induces CD11c-positive dendritic-like cell in the APP/PS1 transgenic mouse brains , 2011, Brain Research.

[32]  C. Barbiéri,et al.  Glycosphingolipid antigens of Leishmania (Leishmania) amazonensis amastigotes identified by use of a monoclonal antibody , 1993, Infection and immunity.

[33]  S. Barger,et al.  Interleukin-1 Mediates Pathological Effects of Microglia on Tau Phosphorylation and on Synaptophysin Synthesis in Cortical Neurons through a p38-MAPK Pathway , 2003, The Journal of Neuroscience.

[34]  S. Phipps,et al.  RAGE and TLRs: relatives, friends or neighbours? , 2013, Molecular immunology.

[35]  S. Yan,et al.  RAGE and Alzheimer's disease: a progression factor for amyloid-beta-induced cellular perturbation? , 2009, Journal of Alzheimer's disease : JAD.

[36]  A. Coyle,et al.  HMGB1 and RAGE in inflammation and cancer. , 2010, Annual review of immunology.

[37]  V. Srikanth,et al.  Advanced glycation endproducts and their receptor RAGE in Alzheimer's disease , 2011, Neurobiology of Aging.

[38]  I. Aroch,et al.  Canine leishmaniasis: a diagnostic and clinical challenge. , 2008, Veterinary journal.

[39]  R. Ader,et al.  Brain, behavior, and immunity , 1987, Brain, Behavior, and Immunity.

[40]  Xiongwei Zhu,et al.  Phosphorylation of Tau Protein as the Link between Oxidative Stress, Mitochondrial Dysfunction, and Connectivity Failure: Implications for Alzheimer's Disease , 2013, Oxidative medicine and cellular longevity.

[41]  P. Kaye,et al.  Macrophages, pathology and parasite persistence in experimental visceral leishmaniasis. , 2004, Trends in parasitology.

[42]  M. Cooper,et al.  Renal microvascular injury in diabetes: RAGE and redox signaling. , 2006, Antioxidants & redox signaling.

[43]  Z. Guo,et al.  Advanced oxidation protein products activate vascular endothelial cells via a RAGE-mediated signaling pathway. , 2008, Antioxidants & redox signaling.

[44]  J. Alexander,et al.  T helper (h)1/Th2 and Leishmania: paradox rather than paradigm. , 2005, Immunology letters.

[45]  R. Mortara,et al.  Central nervous system involvement in experimental infection with Leishmania (Leishmania) amazonensis. , 2003, The American journal of tropical medicine and hygiene.

[46]  C. Heizmann,et al.  S100B and S100A6 Differentially Modulate Cell Survival by Interacting with Distinct RAGE (Receptor for Advanced Glycation End Products) Immunoglobulin Domains* , 2007, Journal of Biological Chemistry.

[47]  Xi Chen,et al.  RAGE potentiates Aβ‐induced perturbation of neuronal function in transgenic mice , 2004, The EMBO journal.

[48]  P. Veras,et al.  Immune and inflammatory responses to Leishmania amazonensis isolated from different clinical forms of human leishmaniasis in CBA mice. , 2011, Memorias do Instituto Oswaldo Cruz.

[49]  R. Cecchini,et al.  OXIDATIVE STRESS OF LIVER IN HAMSTERS INFECTED WITH LEISHMANIA (L.) CHAGASI , 2000, The Journal of parasitology.

[50]  Bradley T. Hyman,et al.  Tau pathophysiology in neurodegeneration: a tangled issue , 2009, Trends in Neurosciences.

[51]  A. Pontecorvi,et al.  Hormones and antioxidant systems: Role of pituitary and pituitary-dependent axes , 2010, Journal of endocrinological investigation.

[52]  J. Fietto,et al.  Immune response induced by New World Leishmania species in C57BL/6 mice , 2004, Parasitology Research.

[53]  Barry Halliwell,et al.  Oxidative stress and neurodegeneration: where are we now? , 2006, Journal of neurochemistry.

[54]  H. Goto,et al.  Cutaneous and mucocutaneous leishmaniasis. , 2012, Infectious disease clinics of North America.

[55]  P. Sawchenko,et al.  Lipopolysaccharide‐induced tau phosphorylation and kinase activity – modulation, but not mediation, by corticotropin‐releasing factor receptors , 2011, The European journal of neuroscience.

[56]  W. Wahli,et al.  Tau hyperphosphorylation and increased BACE1 and RAGE levels in the cortex of PPARβ/δ-null mice. , 2013, Biochimica et biophysica acta.

[57]  V. M. Lima,et al.  Pro-inflammatory cytokines predominate in the brains of dogs with visceral leishmaniasis: a natural model of neuroinflammation during systemic parasitic infection. , 2013, Veterinary parasitology.

[58]  R. Mortara,et al.  Leishmania amazonensis META2 protein confers protection against heat shock and oxidative stress. , 2011, Experimental parasitology.

[59]  L. Fenart,et al.  Apical-to-basolateral transport of amyloid-β peptides through blood-brain barrier cells is mediated by the receptor for advanced glycation end-products and is restricted by P-glycoprotein. , 2010, Journal of Alzheimer's disease : JAD.

[60]  D. Gelain,et al.  Vitamin A (retinol) downregulates the receptor for advanced glycation endproducts (RAGE) by oxidant-dependent activation of p38 MAPK and NF-kB in human lung cancer A549 cells. , 2013, Cellular signalling.

[61]  Leonard Petrucelli,et al.  The role of tau in neurodegeneration , 2009, Molecular Neurodegeneration.

[62]  G. Münch,et al.  Inflammation and the Redox‐sensitive AGE–RAGE Pathway as a Therapeutic Target in Alzheimer's Disease , 2008, Annals of the New York Academy of Sciences.

[63]  Xiao-Hong Li,et al.  AGEs induce Alzheimer-like tau pathology and memory deficit via RAGE-mediated GSK-3 activation , 2012, Neurobiology of Aging.

[64]  B. Rossi-Bergmann,et al.  Hyperbaric oxygen therapy reduces the size of Leishmania amazonensis-induced soft tissue lesions in mice. , 2006, Acta tropica.