4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The behavioural and neuropathologic sexual dimorphism and absence of MIP-3α in tau P301S mouse model of Alzheimer’s disease

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Tau hyper-phosphorylation has been considered a major contributor to neurodegeneration in Alzheimer’s disease (AD) and related tauopathies, and has gained prominence in therapeutic development for AD. To elucidate the pathogenic mechanisms underlying AD and evaluate therapeutic approaches targeting tau, numerous transgenic mouse models that recapitulate critical AD-like pathology have been developed. Tau P301S transgenic mice is one of the most widely used mouse models in AD research. Extensive studies have demonstrated that sex significantly influences AD pathology, behavioral status, and therapeutic outcomes, suggesting that studies using mouse models of AD must consider sex- and age-related differences in neuropathology, behavior, and plasma content.

          Method

          We systematically investigated differences in tau P301S transgenic mice (PS19 line) and wildtype littermates of different sex behavioral performance, tau neuropathology, and biomarkers in plasma and brain.

          Results

          Male P301S transgenic mice exhibited significant changes in weight loss, survival rate, clasping, kyphosis, composite phenotype assessment, nest building performance, tau phosphorylation at Ser202/Thr205, and astrocyte activation compared to that of wild-type littermates. In contrast, female P301S transgenic mice were only sensitive in the Morris water maze and open field test. In addition, we characterized the absence of macrophage-inflammatory protein (MIP-3α) and the upregulation of interferon (IFN)-γ, interleukin (IL)-5, and IL-6 in the plasma of P301S transgenic mice, which can be served as potential plasma biomarkers in P301S Tg mice. Male P301S transgenic mice expressed more monokine induced by IFN-γ (MIG), tumor necrosis factor-α (TNF-α), IL-10, and IL-13 than those of female P301S mice.

          Conclusion

          Our findings highlight sexual dimorphism in the behavior, neuropathology, and plasma proteins in tau P301S transgenic AD mice, indicating that the use of male P301S transgenic mice may be more suitable for assessing anti-phosphorylated tau therapeutic strategies for AD and related tauopathies, and the MIP-3α may be a new potential plasma biomarker.

          Related collections

          Most cited references18

          • Record: found
          • Abstract: found
          • Article: not found

          Abundant tau filaments and nonapoptotic neurodegeneration in transgenic mice expressing human P301S tau protein.

          The identification of mutations in the Tau gene in frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) has made it possible to express human tau protein with pathogenic mutations in transgenic animals. Here we report on the production and characterization of a line of mice transgenic for the 383 aa isoform of human tau with the P301S mutation. At 5-6 months of age, homozygous animals from this line developed a neurological phenotype dominated by a severe paraparesis. According to light microscopy, many nerve cells in brain and spinal cord were strongly immunoreactive for hyperphosphorylated tau. According to electron microscopy, abundant filaments made of hyperphosphorylated tau protein were present. The majority of filaments resembled the half-twisted ribbons described previously in cases of FTDP-17, with a minority of filaments resembling the paired helical filaments of Alzheimer's disease. Sarkosyl-insoluble tau from brains and spinal cords of transgenic mice ran as a hyperphosphorylated 64 kDa band, the same apparent molecular mass as that of the 383 aa tau isoform in the human tauopathies. Perchloric acid-soluble tau was also phosphorylated at many sites, with the notable exception of serine 214. In the spinal cord, neurodegeneration was present, as indicated by a 49% reduction in the number of motor neurons. No evidence for apoptosis was obtained, despite the extensive colocalization of hyperphosphorylated tau protein with activated MAP kinase family members. The latter may be involved in the hyperphosphorylation of tau.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Burrowing and nest building behavior as indicators of well-being in mice.

            The assessment of pain, distress and suffering, as well as evaluation of the efficacy of stress-reduction strategies, is crucial in animal experimentation but can be challenging in laboratory mice. Nest building and burrowing performance, observed in the home cage, have proved to be valuable and easy-to-use tools to assess brain damage or malfunction as well as neurodegenerative diseases. Both behaviors are used as parameters in models of psychiatric disorders or to monitor sickness behavior following infection. Their use has been proposed in more realistic and clinically relevant preclinical models of disease, and reduction of these behaviors seems to be especially useful as an early sign of dysfunction and to monitor disease progression. Finally, both behaviors are reduced by pain and stress. Therefore, in combination with specific disease markers, changes in nest building and burrowing performance may help provide a global picture of a mouse's state, and thus aid monitoring to ensure well-being in animal experimentation. Copyright © 2014 Elsevier B.V. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Sex and the development of Alzheimer's disease.

              Men and women exhibit differences in the development and progression of Alzheimer's disease (AD). The factors underlying the sex differences in AD are not well understood. This Review emphasizes the contributions of sex steroid hormones to the relationship between sex and AD. In women, events that decrease lifetime exposure to estrogens are generally associated with increased AD risk, whereas estrogen-based hormone therapy administered near the time of menopause may reduce AD risk. In men, estrogens do not exhibit age-related reduction and are not significantly associated with AD risk. Rather, normal age-related depletions of testosterone in plasma and brain predict enhanced vulnerability to AD. Both estrogens and androgens exert numerous protective actions in the adult brain that increase neural functioning and resilience as well as specifically attenuating multiple aspects of AD-related neuropathology. Aging diminishes the activational effects of sex hormones in sex-specific manners, which is hypothesized to contribute to the relationship between aging and AD. Sex steroid hormones may also drive sex differences in AD through their organizational effects during developmental sexual differentiation of the brain. Specifically, sex hormone actions during early development may confer inherent vulnerability of the female brain to development of AD in advanced age. The combined effects of organizational and activational effects of sex steroids yield distinct sex differences in AD pathogenesis, a significant variable that must be more rigorously considered in future research. © 2016 Wiley Periodicals, Inc.
                Bookmark

                Author and article information

                Contributors
                topwuhui@jlu.edu.cn
                weikong@jlu.edu.cn
                Journal
                J Neuroinflammation
                J Neuroinflammation
                Journal of Neuroinflammation
                BioMed Central (London )
                1742-2094
                24 February 2020
                24 February 2020
                2020
                : 17
                : 72
                Affiliations
                [1 ]GRID grid.64924.3d, ISNI 0000 0004 1760 5735, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, , Jilin University, ; Changchun, 130012 China
                [2 ]GRID grid.9227.e, ISNI 0000000119573309, State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, , Chinese Academy of Sciences, ; Beijing, 100101 China
                [3 ]GRID grid.452829.0, Department of Pathology, , The Second Hospital of Jilin University, ; Changchun, 130041 Jilin China
                [4 ]GRID grid.464353.3, ISNI 0000 0000 9888 756X, Laboratory of Pathogenic Microbiology and Immunology, College of life science, , Jilin Agricultural University, ; Changchun, 130012 China
                [5 ]GRID grid.64924.3d, ISNI 0000 0004 1760 5735, Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, , Jilin University, ; Changchun, 130012 China
                Article
                1749
                10.1186/s12974-020-01749-w
                7041244
                32093751
                ecd12969-8aa6-4a46-ab9e-4f5ecb2d29c2
                © The Author(s) 2020

                Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

                History
                : 19 November 2019
                : 17 February 2020
                Funding
                Funded by: Jilin Scientific and Technological Development Program (CN)
                Award ID: 20190201185JC
                Award Recipient :
                Funded by: National Natural Science Foundation of China (CN)
                Award ID: 31971142
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100012226, Fundamental Research Funds for the Central Universities;
                Award ID: 2019JCXK-55
                Award Recipient :
                Categories
                Research
                Custom metadata
                © The Author(s) 2020

                Neurosciences
                alzheimer’s disease,tauopathies,sexual dimorphism,tau p301s transgenic mice,behavior,tau hyper-phosphorylation,plasma biomarker

                Comments

                Comment on this article