1
|
Krumm L, Pozner T, Zagha N, Coras R, Arnold P, Tsaktanis T, Scherpelz K, Davis MY, Kaindl J, Stolzer I, Süß P, Khundadze M, Hübner CA, Riemenschneider MJ, Baets J, Günther C, Jayadev S, Rothhammer V, Krach F, Winkler J, Winner B, Regensburger M. Neuroinflammatory disease signatures in SPG11-related hereditary spastic paraplegia patients. Acta Neuropathol 2024; 147:28. [PMID: 38305941 PMCID: PMC10837238 DOI: 10.1007/s00401-023-02675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 02/03/2024]
Abstract
Biallelic loss of SPG11 function constitutes the most frequent cause of complicated autosomal recessive hereditary spastic paraplegia (HSP) with thin corpus callosum, resulting in progressive multisystem neurodegeneration. While the impact of neuroinflammation is an emerging and potentially treatable aspect in neurodegenerative diseases and leukodystrophies, the role of immune cells in SPG11-HSP patients is unknown. Here, we performed a comprehensive immunological characterization of SPG11-HSP, including examination of three human postmortem brain donations, immunophenotyping of patients' peripheral blood cells and patient-specific induced pluripotent stem cell-derived microglia-like cells (iMGL). We delineate a previously unknown role of innate immunity in SPG11-HSP. Neuropathological analysis of SPG11-HSP patient brain tissue revealed profound microgliosis in areas of neurodegeneration, downregulation of homeostatic microglial markers and cell-intrinsic accumulation of lipids and lipofuscin in IBA1+ cells. In a larger cohort of SPG11-HSP patients, the ratio of peripheral classical and intermediate monocytes was increased, along with increased serum levels of IL-6 that correlated with disease severity. Stimulation of patient-specific iMGLs with IFNγ led to increased phagocytic activity compared to control iMGL as well as increased upregulation and release of proinflammatory cytokines and chemokines, such as CXCL10. On a molecular basis, we identified increased STAT1 phosphorylation as mechanism connecting IFNγ-mediated immune hyperactivation and SPG11 loss of function. STAT1 expression was increased both in human postmortem brain tissue and in an Spg11-/- mouse model. Application of an STAT1 inhibitor decreased CXCL10 production in SPG11 iMGL and rescued their toxic effect on SPG11 neurons. Our data establish neuroinflammation as a novel disease mechanism in SPG11-HSP patients and constitute the first description of myeloid cell/ microglia activation in human SPG11-HSP. IFNγ/ STAT1-mediated neurotoxic effects of hyperreactive microglia upon SPG11 loss of function indicate that immunomodulation strategies may slow down disease progression.
Collapse
Affiliation(s)
- Laura Krumm
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Tatyana Pozner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Naime Zagha
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Coras
- Department of Neuropathology, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Thanos Tsaktanis
- Department of Neurology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Kathryn Scherpelz
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Marie Y Davis
- Department of Neurology, University of Washington Medical Center, Seattle, WA, USA
- VA Puget Sound Healthcare System, Seattle, WA, USA
| | - Johanna Kaindl
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Iris Stolzer
- Department of Medicine 1, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Patrick Süß
- Department of Neurology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Mukhran Khundadze
- Institute of Human Genetics, Jena University Hospital Friedrich-Schiller-University Jena, Jena, Germany
| | - Christian A Hübner
- Institute of Human Genetics, Jena University Hospital Friedrich-Schiller-University Jena, Jena, Germany
- Center for Rare Diseases, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | | | - Jonathan Baets
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Claudia Günther
- Department of Medicine 1, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Kussmaulallee 4, 91054, Erlangen, Germany
| | - Suman Jayadev
- Department of Neurology, University of Washington Medical Center, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Veit Rothhammer
- Department of Neurology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Florian Krach
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Winkler
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
- Department of Molecular Neurology, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Regensburger
- Department of Stem Cell Biology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Kussmaulallee 4, 91054, Erlangen, Germany.
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany.
- Department of Molecular Neurology, FAU Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
2
|
Liu J, Zhang Y, Lai CJS, Xie J. Multitarget Protective Effects of JUB on Aβ-Induced Neurotoxicity and the Mechanism Predication Using Network Pharmacology Analysis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:20724-20734. [PMID: 38098161 DOI: 10.1021/acs.jafc.3c06430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Amyloid-β (Aβ) is one of the core factors in the pathogenesis of Alzheimer's disease (AD), and the accumulation of its aggregates in the brain can form age-related plaques, leading to brain cell damage and intellectual decline, which may be the common intersection of all causes of neurotoxicity. Jujuboside B (JUB) has many characteristics such as hypnosis, sedation, antianxiety, and antioxidant stress. However, it is still unclear whether JuB can alleviate the neurotoxicity caused by Aβ. Our study demonstrates that JUB improves learning and memory deficits in the nematode model. At the same time, JUB increases the antioxidant activity, prevents excessive accumulation of lipid synthesis, and resists endogenous lipofuscin deposition, thereby inhibiting the toxic effect of Aβ. In vitro, JUB can improve Aβ1-42-induced neuronal apoptosis level through the Bax/Bcl-2/caspase-3 signaling pathway and restore mitochondrial function in SH-SY5Y cells. The network pharmacology has been used to predict the potential neuroprotective mechanism of JUB. In summary, JUB exhibits neuroprotective properties employing both a neural cell and a nematode, which provides a basis for screening candidate ingredients for preventing AD.
Collapse
Affiliation(s)
- Jinrui Liu
- College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Yanqing Zhang
- College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Chang-Jiang-Sheng Lai
- State Key Laboratory Breeding Base of Dao - di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junbo Xie
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
3
|
Gagné F, Roubeau-Dumont E, André C, Auclair J. Micro and Nanoplastic Contamination and Its Effects on Freshwater Mussels Caged in an Urban Area. J Xenobiot 2023; 13:761-774. [PMID: 38132709 PMCID: PMC10744427 DOI: 10.3390/jox13040048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Plastic-based contamination has become a major cause of concern as it pervades many environments such as air, water, sediments, and soils. This study sought to examine the presence of microplastics (MPs) and nanoplastics (NPs) in freshwater mussels placed at rainfall/street runoff overflows, downstream (15 km) of the city centre of Montréal, and 8 km downstream of a municipal effluent dispersion plume. MPs and NPs were determined using flow cytometry and size exclusion chromatography using fluorescence detection. Following 3 months of exposure during the summer season, mussels contained elevated amounts of both MPs and NPs. The rainfall overflow and downstream of the city centre were the most contaminated sites. Lipid peroxidation, metallothioneins, and protein aggregates (amyloids) were significantly increased at the most contaminated sites and were significantly correlated with NPs in tissues. Based on the levels of MPs and NPs in mussels exposed to municipal effluent, wastewater treatment plants appear to mitigate plastic contamination albeit not completely. In conclusion, the data support the hypothesis that mussels placed in urbanized areas are more contaminated by plastics, which are associated with oxidative damage. The highest responses observed at the overflow site suggest that tire wear and/or asphalt (road) erosion MPs/NPs represent important sources of contamination for the aquatic biota.
Collapse
Affiliation(s)
- François Gagné
- Aquatic Contaminants Research Division, Environment and Climate Change Canada, Montréal, QC H2Y 2E7, Canada; (E.R.-D.); (C.A.); (J.A.)
| | | | | | | |
Collapse
|
4
|
Yang Y, Yu Q, Li B, Li S, Yang Z, Yuan F, Liu Z. A single dose of lipopolysaccharide elicits autofluorescence in the mouse brain. Front Aging Neurosci 2023; 15:1126273. [PMID: 37020861 PMCID: PMC10067636 DOI: 10.3389/fnagi.2023.1126273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 03/03/2023] [Indexed: 03/22/2023] Open
Abstract
One hallmark of aging is autofluorescence (AF) in the brain. However, the underlying mechanism for inducing AF remains unknown. This study aims to determine the cause(s) of this phenomenon. The endogenous expression pattern of AF in mice was examined at differing ages. Intraperitoneal injection of a single dose of lipopolysaccharide (LPS) was performed to induce AF. Copper sulfate was applied to remove AF to allow for further immunofluorescence staining. AF appeared in the mouse brain as early as 3 months of age. In the cortex, AF occurs in the lysosomes of microglia, astrocytes, endothelial cells, and oligodendrocyte lineage cells and its prevalence increases with age. Interestingly, AF never occurs in the pericytes of young or aged brains. LPS administration resulted in a rapid and marked induction of brain AF, similar to the normal aging process. Finally, age-related and induced AF can be eliminated by low concentrations of copper sulfate solution. This pre-treatment is safe for aging and lineage tracing studies. These findings depict that AF in the brain could be associated with the innate immune response against Gram-negative bacteria infection.
Collapse
Affiliation(s)
- Yanzhuo Yang
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Qingting Yu
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Bin Li
- Zhoushan Institute for Food and Drug Control, Zhoushan, China
| | - Shijia Li
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Zuisu Yang
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Falei Yuan
- Department of Pharmacy, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
- Falei Yuan,
| | - Zhongliang Liu
- Department of Oncology, Zhoushan Hospital of Traditional Chinese Medicine, Zhoushan, China
- *Correspondence: Zhongliang Liu,
| |
Collapse
|
5
|
Modulation of the Microglial Nogo-A/NgR Signaling Pathway as a Therapeutic Target for Multiple Sclerosis. Cells 2022; 11:cells11233768. [PMID: 36497029 PMCID: PMC9737582 DOI: 10.3390/cells11233768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Current therapeutics targeting chronic phases of multiple sclerosis (MS) are considerably limited in reversing the neural damage resulting from repeated inflammation and demyelination insults in the multi-focal lesions. This inflammation is propagated by the activation of microglia, the endogenous immune cell aiding in the central nervous system homeostasis. Activated microglia may transition into polarized phenotypes; namely, the classically activated proinflammatory phenotype (previously categorized as M1) and the alternatively activated anti-inflammatory phenotype (previously, M2). These transitional microglial phenotypes are dynamic states, existing as a continuum. Shifting microglial polarization to an anti-inflammatory status may be a potential therapeutic strategy that can be harnessed to limit neuroinflammation and further neurodegeneration in MS. Our research has observed that the obstruction of signaling by inhibitory myelin proteins such as myelin-associated inhibitory factor, Nogo-A, with its receptor (NgR), can regulate microglial cell function and activity in pre-clinical animal studies. Our review explores the microglial role and polarization in MS pathology. Additionally, the potential therapeutics of targeting Nogo-A/NgR cellular mechanisms on microglia migration, polarization and phagocytosis for neurorepair in MS and other demyelination diseases will be discussed.
Collapse
|
6
|
Ferroptosis as a Novel Determinant of β-Cell Death in Diabetic Conditions. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3873420. [PMID: 35320979 PMCID: PMC8938062 DOI: 10.1155/2022/3873420] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 12/21/2022]
Abstract
The main pathological hallmark of diabetes is the loss of functional β-cells. Among several types of β-cell death in diabetes, the involvement of ferroptosis remains elusive. Therefore, we investigated the potential of diabetes-mimicking factors: high glucose (HG), proinflammatory cytokines, hydrogen peroxide (H2O2), or diabetogenic agent streptozotocin (STZ) to induce ferroptosis of β-cells in vitro. Furthermore, we tested the contribution of ferroptosis to injury of pancreatic islets in an STZ-induced in vivo diabetic model. All in vitro treatments increased loss of Rin-5F cells along with the accumulation of reactive oxygen species, lipid peroxides and iron, inactivation of NF-E2-related factor 2 (Nrf2), and decrease in glutathione peroxidase 4 expression and mitochondrial membrane potential (MMP). Ferrostatin 1 (Fer-1), ferroptosis inhibitor, diminished the above-stated effects and rescued cells from death in case of HG, STZ, and H2O2 treatments, while failed to increase MMP and to attenuate cell death after the cytokines' treatment. Moreover, Fer-1 protected pancreatic islets from STZ-induced injury in diabetic in vivo model, since it decreased infiltration of macrophages and accumulation of lipid peroxides and increased the population of insulin-positive cells. Such results revealed differences between diabetogenic stimuli in determining the destiny of β-cells, emerging HG, H2O2, and STZ, but not cytokines, as contributing factors to ferroptosis and shed new light on an antidiabetic strategy based on Nrf2 activation. Thus, targeting ferroptosis in diabetes might be a promising new approach for preservation of the β-cell population. Our results obtained from in vivo study strongly justify this approach.
Collapse
|
7
|
Abstract
Ageing, death, and potential immortality lie at the heart of biology, but two seemingly incompatible paradigms coexist in different research communities and have done since the nineteenth century. The universal senescence paradigm sees senescence as inevitable in all cells. Damage accumulates. The potential immortality paradigm sees some cells as potentially immortal, especially unicellular organisms, germ cells and cancerous cells. Recent research with animal cells, yeasts and bacteria show that damaged cell constituents do in fact build up, but can be diluted by growth and cell division, especially by asymmetric cell division. By contrast, mammalian embryonic stem cells and many cancerous and 'immortalized' cell lines divide symmetrically, and yet replicate indefinitely. How do they acquire their potential immortality? I suggest they are rejuvenated by excreting damaged cell constituents in extracellular vesicles. If so, our understanding of cellular senescence, rejuvenation and potential immortality could be brought together in a new synthesis, which I call the cellular rejuvenation hypothesis: damaged cell constituents build up in all cells, but cells can be rejuvenated either by growth and cell division or, in 'immortal' cell lines, by excreting damaged cell constituents. In electronic supplementary material, appendix, I outline nine ways in which this hypothesis could be tested.
Collapse
|
8
|
Fang Y, Taubitz T, Tschulakow AV, Heiduschka P, Szewczyk G, Burnet M, Peters T, Biesemeier A, Sarna T, Schraermeyer U, Julien-Schraermeyer S. Removal of RPE lipofuscin results in rescue from retinal degeneration in a mouse model of advanced Stargardt disease: Role of reactive oxygen species. Free Radic Biol Med 2022; 182:132-149. [PMID: 35219849 DOI: 10.1016/j.freeradbiomed.2022.02.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 11/30/2022]
Abstract
Accumulation of lipofuscin in the retinal pigment epithelium (RPE) is a hallmark of aging and is associated with retinal degeneration encountered in age-related macular degeneration (AMD) and Stargardt disease (SD). Currently, treatment for lipofuscin-induced retinal degeneration is unavailable. Here, we report that Remofuscin (INN: soraprazan, a tetrahydropyridoether small molecule) reverses lipofuscin accumulation in aged primary human RPE cells and is non-cytotoxic in aged SD mouse RPE cells in vitro. In addition, we show that the removal of lipofuscin after a single intravitreal injection of Remofuscin results in a rescue from retinal degeneration in a mouse model of advanced SD which is even accompanied by an amelioration of the retinal dysfunction. Finally, we demonstrate that the mechanism causing lipofuscinolysis may involve the reactive oxygen species generated via the presence of Remofuscin. These data suggest a possible therapeutic approach to untreatable lipofuscin-mediated diseases like AMD, SD and lipofuscinopathies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuan Fang
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Tatjana Taubitz
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Alexander V Tschulakow
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany; STZ Ocutox, Preclinical Drug Assessment, Hechingen, Germany
| | - Peter Heiduschka
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Grzegorz Szewczyk
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | | | - Tobias Peters
- Centre for Ophthalmology, University of Tuebingen, Tuebingen, Germany
| | - Antje Biesemeier
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Tadeusz Sarna
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ulrich Schraermeyer
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany; STZ Ocutox, Preclinical Drug Assessment, Hechingen, Germany
| | - Sylvie Julien-Schraermeyer
- Division of Experimental Vitreoretinal Surgery, Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany; STZ Ocutox, Preclinical Drug Assessment, Hechingen, Germany.
| |
Collapse
|
9
|
|
10
|
Sun Y, Xu S, Jiang M, Liu X, Yang L, Bai Z, Yang Q. Role of the Extracellular Matrix in Alzheimer's Disease. Front Aging Neurosci 2021; 13:707466. [PMID: 34512308 PMCID: PMC8430252 DOI: 10.3389/fnagi.2021.707466] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/04/2021] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease with complex pathological characteristics, whose etiology and pathogenesis are still unclear. Over the past few decades, the role of the extracellular matrix (ECM) has gained importance in neurodegenerative disease. In this review, we describe the role of the ECM in AD, focusing on the aspects of synaptic transmission, amyloid-β-plaque generation and degradation, Tau-protein production, oxidative-stress response, and inflammatory response. The function of ECM in the pathological process of AD will inform future research on the etiology and pathogenesis of AD.
Collapse
Affiliation(s)
- Yahan Sun
- College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, China
| | - Sen Xu
- College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, China
| | - Ming Jiang
- College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, China
| | - Xia Liu
- College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, China
| | - Liang Yang
- College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, China
| | - Zhantao Bai
- College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, China
| | - Qinghu Yang
- College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, China
| |
Collapse
|
11
|
Souder DC, Dreischmeier IA, Smith AB, Wright S, Martin SA, Sagar MAK, Eliceiri KW, Salamat SM, Bendlin BB, Colman RJ, Beasley TM, Anderson RM. Rhesus monkeys as a translational model for late-onset Alzheimer's disease. Aging Cell 2021; 20:e13374. [PMID: 33951283 PMCID: PMC8208787 DOI: 10.1111/acel.13374] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/22/2021] [Accepted: 04/11/2021] [Indexed: 11/28/2022] Open
Abstract
Age is a major risk factor for late-onset Alzheimer's disease (AD) but seldom features in laboratory models of the disease. Furthermore, heterogeneity in size and density of AD plaques observed in individuals are not recapitulated in transgenic mouse models, presenting an incomplete picture. We show that the amyloid plaque microenvironment is not equivalent between rodent and primate species, and that differences in the impact of AD pathology on local metabolism and inflammation might explain established differences in neurodegeneration and functional decline. Using brain tissue from transgenic APP/PSEN1 mice, rhesus monkeys with age-related amyloid plaques, and human subjects with confirmed AD, we report altered energetics in the plaque microenvironment. Metabolic features included changes in mitochondrial distribution and enzymatic activity, and changes in redox cofactors NAD(P)H that were shared among species. A greater burden of lipofuscin was detected in the brains from monkeys and humans of advanced age compared to transgenic mice. Local inflammatory signatures indexed by astrogliosis and microglial activation were detected in each species; however, the inflamed zone was considerably larger for monkeys and humans. These data demonstrate the advantage of nonhuman primates in modeling the plaque microenvironment, and provide a new framework to investigate how AD pathology might contribute to functional loss.
Collapse
Affiliation(s)
- Dylan C. Souder
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
| | | | - Alex B. Smith
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
| | - Samantha Wright
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
| | - Stephen A. Martin
- Biology of Aging Laboratory Center for American Indian and Rural Health Equity Montana State University Bozeman MT USA
| | - Md Abdul Kader Sagar
- Department of Biomedical Engineering University of Wisconsin Madison Madison WI USA
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering University of Wisconsin Madison Madison WI USA
| | - Shahriar M. Salamat
- Department of Pathology Laboratory Medicine University of Wisconsin Madison Madison WI USA
- Neurological Surgery University of Wisconsin Madison Madison WI USA
| | | | - Ricki J. Colman
- Wisconsin National Primate Research Center University of Wisconsin Madison Madison WI USA
| | - T. Mark Beasley
- Department of Biostatistics University of Alabama Birmingham AL USA
- GRECC Birmingham/Atlanta Veterans Administration Hospital Birmingham AL USA
| | - Rozalyn M. Anderson
- Division of Geriatrics Department of Medicine SMPH Madison WI USA
- GRECC William S. Middleton Memorial Veterans Hospital Madison WI USA
| |
Collapse
|
12
|
Mold MJ, O’Farrell A, Morris B, Exley C. Aluminum and Tau in Neurofibrillary Tangles in Familial Alzheimer's Disease. J Alzheimers Dis Rep 2021; 5:283-294. [PMID: 34113785 PMCID: PMC8150251 DOI: 10.3233/adr-210011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Familial Alzheimer's disease (fAD) is driven by genetic predispositions affecting the expression and metabolism of the amyloid-β protein precursor. Aluminum is a non-essential yet biologically-reactive metal implicated in the etiology of AD. Recent research has identified aluminum intricately and unequivocally associated with amyloid-β in senile plaques and, more tentatively, co-deposited with neuropil-like threads in the brains of a Colombian cohort of donors with fAD. OBJECTIVE Herein, we have assessed the co-localization of aluminum to immunolabelled phosphorylated tau to probe the potential preferential binding of aluminum to senile plaques or neurofibrillary tangles in the same Colombian kindred. METHODS Herein, we have performed phosphorylated tau-specific immunolabelling followed by aluminum-specific fluorescence microscopy of the identical brain tissue sections via a sequential labelling method. RESULTS Aluminum was co-localized with immunoreactive phosphorylated tau in the brains of donors with fAD. While aluminum was predominantly co-located to neurofibrillary tangles in the temporal cortex, aluminum was more frequently co-deposited with cortical senile plaques. CONCLUSION These data suggest that the co-deposition of aluminum with amyloid-β precedes that with neurofibrillary tangles. Extracellularly deposited amyloid-β may also be more immediately available to bind aluminum versus intracellular aggregates of tau. Therapeutic approaches to reduce tau have demonstrated the amelioration of its synergistic interactions with amyloid-β, ultimately reducing tau pathology and reducing neuronal loss. These data support the intricate associations of aluminum in the neuropathology of fAD, of which its subsequent reduction may further therapeutic benefits observed in ongoing clinical trials in vivo.
Collapse
Affiliation(s)
- Matthew John Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, UK
| | - Adam O’Farrell
- School of Life Sciences, Huxley Building, Keele University, Keele, Staffordshire, UK
| | - Benjamin Morris
- School of Life Sciences, Huxley Building, Keele University, Keele, Staffordshire, UK
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, UK
| |
Collapse
|
13
|
Abhijit S, Tripathi SJ, Shankaranarayana Rao B, Asha Devi S. Grape seed proanthocyanidin extract and swimming training enhances neuronal number in dorso-medial prefrontal cortex in middle-aged male rats by alleviating oxidative stress. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
14
|
Kakimoto Y, Okada C, Kawabe N, Sasaki A, Tsukamoto H, Nagao R, Osawa M. Myocardial lipofuscin accumulation in ageing and sudden cardiac death. Sci Rep 2019; 9:3304. [PMID: 30824797 PMCID: PMC6397159 DOI: 10.1038/s41598-019-40250-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/12/2019] [Indexed: 01/10/2023] Open
Abstract
Lipofuscin is an intracellular aggregate of highly oxidized proteins that cannot be digested in the ubiquitin-proteasome system and accumulate mainly in lysosomes, especially in aged cells and pathological conditions. However, no systematic study has evaluated the cardiac accumulation of lipofuscin during human ageing and sudden cardiac death (SCD). Age estimation in unidentified bodies and postmortem SCD diagnosis are important themes in forensics. Thus, we aimed to elucidate their correlations with myocardial lipofuscin accumulation. We collected 76 cardiac samples from autopsy patients aged 20–97 years. After histopathological examination, myocardial lipofuscin was measured using its autofluorescence. Lipofuscin accumulated mainly in the perinuclear zone, and its accumulation rate positively correlated with chronological ageing (r = 0.82). Meanwhile, no significant change in lipofuscin level was observed with different causes of death, including SCD. There was also no significant change in lipofuscin level in relation to body mass index, serum brain natriuretic peptide level, or heart weight. Moreover, we performed LC3 and p62 immunoblotting to evaluate autophagic activity, and no change was observed in ageing. Therefore, lipofuscin accumulation more directly reflects chronological ageing rather than human cardiac pathology. Our study reveals the stability and utility of cardiac lipofuscin measurement for age estimation during autopsy.
Collapse
Affiliation(s)
- Yu Kakimoto
- Department of Forensic Medicine, Tokai University School of Medicine, Kanagawa, Japan.
| | - Chisa Okada
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Noboru Kawabe
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Ayumi Sasaki
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Hideo Tsukamoto
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Ryoko Nagao
- Department of Forensic Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Motoki Osawa
- Department of Forensic Medicine, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
15
|
Perrotta I. Occurrence and characterization of lipofuscin and ceroid in human atherosclerotic plaque. Ultrastruct Pathol 2018; 42:477-488. [PMID: 30465462 DOI: 10.1080/01913123.2018.1544953] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Atherosclerotic plaque formation starts early in life, develops silently over decades, and often displays clear evidence of accelerated biological aging. Lipofuscin has been classically defined as "the most consistent and phylogenetically conserved cellular morphologic change of aging," however, despite this traditional view different lines of evidence have recently demonstrated that, besides aging, various noxious influences can engeder its accumulation in cells and also that specific experimental conditions can revert this effect. Lipofuscin has been also proven to interact with disease-related factors to enhance cell loss. Along with lipofuscin, ceroid, another autofluorescent lipopigment usually produced under various pathological conditions unrelated to aging, has been suggested to jeopardize cell performance and viability by inducing membrane fragility, mitochondrial dysfunction, DNA damage, and oxidative stress-induced apoptosis. With regard to atherosclerosis, very few investigations have been conducted to assess whether a link could exist between lipofuscin/ceroid accumulation and the progression of the disease and no information still exist regarding the anatomy and the ultrastructural diversification of lipofuscin and ceroid in the lesional vascular tissue. At the same time, data concerning their potential toxicity at the cellular level are fragmentary, dated, and scarce. The present study investigates the occurrence and distribution of lipofuscin and ceroid in human atherosclerotic plaque and adjacent healthy tissues and analyzes the ultrastructural changes associated with their accumulation within the cell.
Collapse
Affiliation(s)
- Ida Perrotta
- a Department of Biology, Ecology and Earth Sciences, Centre for Microscopy and Microanalysis, Transmission Electron Microscopy Laboratory , University of Calabria , Cosenza , Italy
| |
Collapse
|
16
|
Moreno-García A, Kun A, Calero O, Medina M, Calero M. An Overview of the Role of Lipofuscin in Age-Related Neurodegeneration. Front Neurosci 2018; 12:464. [PMID: 30026686 PMCID: PMC6041410 DOI: 10.3389/fnins.2018.00464] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/18/2018] [Indexed: 12/21/2022] Open
Abstract
Despite aging being by far the greatest risk factor for highly prevalent neurodegenerative disorders, the molecular underpinnings of age-related brain changes are still not well understood, particularly the transition from normal healthy brain aging to neuropathological aging. Aging is an extremely complex, multifactorial process involving the simultaneous interplay of several processes operating at many levels of the functional organization. The buildup of potentially toxic protein aggregates and their spreading through various brain regions has been identified as a major contributor to these pathologies. One of the most striking morphologic changes in neurons during normal aging is the accumulation of lipofuscin (LF) aggregates, as well as, neuromelanin pigments. LF is an autofluorescent lipopigment formed by lipids, metals and misfolded proteins, which is especially abundant in nerve cells, cardiac muscle cells and skin. Within the Central Nervous System (CNS), LF accumulates as aggregates, delineating a specific senescence pattern in both physiological and pathological states, altering neuronal cytoskeleton and cellular trafficking and metabolism, and being associated with neuronal loss, and glial proliferation and activation. Traditionally, the accumulation of LF in the CNS has been considered a secondary consequence of the aging process, being a mere bystander of the pathological buildup associated with different neurodegenerative disorders. Here, we discuss recent evidence suggesting the possibility that LF aggregates may have an active role in neurodegeneration. We argue that LF is a relevant effector of aging that represents a risk factor or driver for neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Alejandra Kun
- Biochemistry Section, Science School, Universidad de la República, Montevideo, Uruguay
- Protein and Nucleic Acids Department, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Olga Calero
- Chronic Disease Programme-CROSADIS, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Miguel Medina
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| | - Miguel Calero
- Chronic Disease Programme-CROSADIS, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| |
Collapse
|
17
|
Press M, Jung T, König J, Grune T, Höhn A. Protein aggregates and proteostasis in aging: Amylin and β-cell function. Mech Ageing Dev 2018; 177:46-54. [PMID: 29580826 DOI: 10.1016/j.mad.2018.03.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 03/22/2018] [Indexed: 01/07/2023]
Abstract
The ubiquitin-proteasomal-system (UPS) and the autophagy-lysosomal-system (ALS) are both highly susceptible for disturbances leading to the accumulation of cellular damage. A decline of protein degradation during aging results in the formation of oxidatively damaged and aggregated proteins finally resulting in failure of cellular functionality. Besides protein aggregation in response to oxidative damage, amyloids are a different type of protein aggregates able to distract proteostasis and interfere with cellular functionality. Amyloids are clearly linked to the pathogenesis of age-related degenerative diseases such as Alzheimer's disease. Human amylin is one of the peptides forming fibrils in β-sheet conformation finally leading to amyloid formation. In contrast to rodent amylin, human amylin is prone to form amyloidogenic aggregates, proposed to play a role in the pathogenesis of Type 2 Diabetes by impairing β-cell functionality. Since aggregates such as lipofuscin and β-amyloid are known to impair proteostasis, it is likely to assume similar effects for human amylin. In this review, we focus on the effects of IAPP on UPS and ALS and their role in amylin degradation, since both systems play a crucial role in maintaining proteome balance thereby influencing, at least in part, cellular fate and aging.
Collapse
Affiliation(s)
- Michaela Press
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 Muenchen-Neuherberg, Germany.
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Cardiovascular Research (DZHK), 10117 Berlin, Germany.
| | - Jeannette König
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany.
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 Muenchen-Neuherberg, Germany; German Center for Cardiovascular Research (DZHK), 10117 Berlin, Germany; NutriAct - Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany; Institute of Nutrition, University of Potsdam, 14558 Nuthetal, Germany.
| | - Annika Höhn
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 Muenchen-Neuherberg, Germany.
| |
Collapse
|
18
|
Kozlov S, Afonin A, Evsyukov I, Bondarenko A. Alzheimer's disease: as it was in the beginning. Rev Neurosci 2018; 28:825-843. [PMID: 28704198 DOI: 10.1515/revneuro-2017-0006] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/19/2017] [Indexed: 01/09/2023]
Abstract
Since Alzheimer's disease was first described in 1907, many attempts have been made to reveal its main cause. Nowadays, two forms of the disease are known, and while the hereditary form of the disease is clearly caused by mutations in one of several genes, the etiology of the sporadic form remains a mystery. Both forms share similar sets of neuropathological and molecular manifestations, including extracellular deposition of amyloid-beta, intracellular accumulation of hyperphosphorylated tau protein, disturbances in both the structure and functions of mitochondria, oxidative stress, metal ion metabolism disorders, impairment of N-methyl-D-aspartate receptor-related signaling pathways, abnormalities of lipid metabolism, and aberrant cell cycle reentry in some neurons. Such a diversity of symptoms led to proposition of various hypotheses for explaining the development of Alzheimer's disease, the amyloid hypothesis, which postulates the key role of amyloid-beta in Alzheimer's disease development, being the most prominent. However, this hypothesis does not fully explain all of the molecular abnormalities and is therefore heavily criticized. In this review, we propose a hypothetical model of Alzheimer's disease progression, assuming a key role of age-related mitochondrial dysfunction, as was postulated in the mitochondrial cascade hypothesis. Our model explains the connections between all the symptoms of Alzheimer's disease, with particular attention to autophagy, metal metabolism disorders, and aberrant cell cycle re-entry in neurons. Progression of the Alzheimer's disease appears to be a complex process involving aging and too many protective mechanisms affecting one another, thereby leading to even greater deleterious effects.
Collapse
|
19
|
Lee WH, Higuchi H, Ikeda S, Macke EL, Takimoto T, Pattnaik BR, Liu C, Chu LF, Siepka SM, Krentz KJ, Rubinstein CD, Kalejta RF, Thomson JA, Mullins RF, Takahashi JS, Pinto LH, Ikeda A. Mouse Tmem135 mutation reveals a mechanism involving mitochondrial dynamics that leads to age-dependent retinal pathologies. eLife 2016; 5:e19264. [PMID: 27863209 PMCID: PMC5117855 DOI: 10.7554/elife.19264] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/25/2016] [Indexed: 12/21/2022] Open
Abstract
While the aging process is central to the pathogenesis of age-dependent diseases, it is poorly understood at the molecular level. We identified a mouse mutant with accelerated aging in the retina as well as pathologies observed in age-dependent retinal diseases, suggesting that the responsible gene regulates retinal aging, and its impairment results in age-dependent disease. We determined that a mutation in the transmembrane 135 (Tmem135) is responsible for these phenotypes. We observed localization of TMEM135 on mitochondria, and imbalance of mitochondrial fission and fusion in mutant Tmem135 as well as Tmem135 overexpressing cells, indicating that TMEM135 is involved in the regulation of mitochondrial dynamics. Additionally, mutant retina showed higher sensitivity to oxidative stress. These results suggest that the regulation of mitochondrial dynamics through TMEM135 is critical for protection from environmental stress and controlling the progression of retinal aging. Our study identified TMEM135 as a critical link between aging and age-dependent diseases.
Collapse
Affiliation(s)
- Wei-Hua Lee
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, United States
| | - Hitoshi Higuchi
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, United States
| | - Sakae Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, United States
| | - Erica L Macke
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, United States
| | - Tetsuya Takimoto
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, United States
| | - Bikash R Pattnaik
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, United States
- Department of Pediatrics, University of Wisconsin-Madison, Madison, United States
| | - Che Liu
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, United States
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, United States
| | - Li-Fang Chu
- Morgridge Institute for Research, Madison, United States
| | - Sandra M Siepka
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Kathleen J Krentz
- Transgenic Mouse Facility, Biotechnology Center, University of Wisconsin-Madison, Madison, United States
| | - C Dustin Rubinstein
- Translational Genomics Facility, Biotechnology Center, University of Wisconsin-Madison, Madison, United States
| | - Robert F Kalejta
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, United States
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, United States
| | | | - Robert F Mullins
- Department of Ophthalmology and Visual, University of Iowa, Iowa City, United States
| | - Joseph S Takahashi
- Department of Neuroscience, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Lawrence H Pinto
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, United States
| |
Collapse
|
20
|
Exercise in an electrotactic flow chamber ameliorates age-related degeneration in Caenorhabditis elegans. Sci Rep 2016; 6:28064. [PMID: 27305857 PMCID: PMC4910109 DOI: 10.1038/srep28064] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/31/2016] [Indexed: 01/09/2023] Open
Abstract
Degeneration is a senescence process that occurs in all living organisms. Although tremendous efforts have been exerted to alleviate this degenerative tendency, minimal progress has been achieved to date. The nematode, Caenorhabditis elegans (C. elegans), which shares over 60% genetic similarities with humans, is a model animal that is commonly used in studies on genetics, neuroscience, and molecular gerontology. However, studying the effect of exercise on C. elegans is difficult because of its small size unlike larger animals. To this end, we fabricated a flow chamber, called “worm treadmill,” to drive worms to exercise through swimming. In the device, the worms were oriented by electrotaxis on demand. After the exercise treatment, the lifespan, lipofuscin, reproductive capacity, and locomotive power of the worms were analyzed. The wild-type and the Alzheimer’s disease model strains were utilized in the assessment. Although degeneration remained irreversible, both exercise-treated strains indicated an improved tendency compared with their control counterparts. Furthermore, low oxidative stress and lipofuscin accumulation were also observed among the exercise-treated worms. We conjecture that escalated antioxidant enzymes imparted the worms with an extra capacity to scavenge excessive oxidative stress from their bodies, which alleviated the adverse effects of degeneration. Our study highlights the significance of exercise in degeneration from the perspective of the simple life form, C. elegans.
Collapse
|
21
|
Talwar P, Sinha J, Grover S, Rawat C, Kushwaha S, Agarwal R, Taneja V, Kukreti R. Dissecting Complex and Multifactorial Nature of Alzheimer's Disease Pathogenesis: a Clinical, Genomic, and Systems Biology Perspective. Mol Neurobiol 2015; 53:4833-64. [PMID: 26351077 DOI: 10.1007/s12035-015-9390-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/11/2015] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by loss of memory and other cognitive functions. AD can be classified into familial AD (FAD) and sporadic AD (SAD) based on heritability and into early onset AD (EOAD) and late onset AD (LOAD) based on age of onset. LOAD cases are more prevalent with genetically complex architecture. In spite of significant research focused on understanding the etiological mechanisms, search for diagnostic biomarker(s) and disease-modifying therapy is still on. In this article, we aim to comprehensively review AD literature on established etiological mechanisms including role of beta-amyloid and apolipoprotein E (APOE) along with promising newer etiological factors such as epigenetic modifications that have been associated with AD suggesting its multifactorial nature. As genomic studies have recently played a significant role in elucidating AD pathophysiology, a systematic review of findings from genome-wide linkage (GWL), genome-wide association (GWA), genome-wide expression (GWE), and epigenome-wide association studies (EWAS) was conducted. The availability of multi-dimensional genomic data has further coincided with the advent of computational and network biology approaches in recent years. Our review highlights the importance of integrative approaches involving genomics and systems biology perspective in elucidating AD pathophysiology. The promising newer approaches may provide reliable means of early and more specific diagnosis and help identify therapeutic interventions for LOAD.
Collapse
Affiliation(s)
- Puneet Talwar
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi, India.,Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India
| | - Juhi Sinha
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India
| | - Sandeep Grover
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India.,Department of Paediatrics, Division of Pneumonology-Immunology, Charité University Medical Centre, Berlin, Germany
| | - Chitra Rawat
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi, India.,Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India
| | - Suman Kushwaha
- Institute of Human Behaviour and Allied Sciences (IHBAS), Delhi, India
| | - Rachna Agarwal
- Institute of Human Behaviour and Allied Sciences (IHBAS), Delhi, India
| | - Vibha Taneja
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Ritushree Kukreti
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi, India. .,Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India.
| |
Collapse
|
22
|
A neuronal aging pattern unique to humans and common chimpanzees. Brain Struct Funct 2014; 221:647-64. [DOI: 10.1007/s00429-014-0931-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 10/24/2014] [Indexed: 12/27/2022]
|
23
|
He Y, Zhao H, Su G. Ginsenoside Rg1 decreases neurofibrillary tangles accumulation in retina by regulating activities of neprilysin and PKA in retinal cells of AD mice model. J Mol Neurosci 2013; 52:101-6. [PMID: 24287922 DOI: 10.1007/s12031-013-0173-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/04/2013] [Indexed: 10/26/2022]
Abstract
Neurofibrillary tangles (NFTs) are the major component of senile plaques in the brains of patients with Alzheimer's disease (AD). However, the mechanism causing NFTs accumulation in AD patients' retina is also elusive. Thus, we investigated the effects of ginsenoside Rg1 on NFTs accumulation in retinal pigment epithelial (RPE) cells isolated form double transgenic APP/PS1 mice model. NFTs amounts in culture supernatants were examined by enzyme-linked immunosorbent assay. Activity and mRNA transcription of enzymes and proteins that regulate NFTs accumulation were examined by activity assay and reverse transcription PCR. The expression of neprilysin (NEP) and neutral endopeptidase (PKA) were detected by western blot assay. Rg1 significantly decreased NFTs accumulation in isolated RPE cells. Activity of NEP was significantly increased, and activity of PKA was significantly decreased in cell lysates of Rg1-feeding APP/PS1 mice compared with non-Rg1-feeding mice. mRNA level of NEP was significantly higher and mRNA level of PKA was significantly lower in cells of Rg1-feeding mice than nonfeeding mice. The phosphorylation of tau at Thr231, Thr205, and Ser396 were significantly decreased in RPE of Rg1-feeding APP/PS1 mice compared with the non-Rg1-feeding mice. Rg1 decreased the NFTs production in RPE cell of APP/PS1 mice by modulating the expression and activity of NEP and PKA, which perform the function through downregulating the phosphorylation of tau protein.
Collapse
Affiliation(s)
- Yanhui He
- Department of Ophthalmology, Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, Jilin, 130041, China
| | | | | |
Collapse
|