1
|
McFadden SA, Peck MR, Sime LN, Cox MF, Ikiz ED, Findley CA, Quinn K, Fang Y, Bartke A, Hascup ER, Hascup KN. Thermotherapy has sexually dimorphic responses in APP/PS1 mice. Aging (Albany NY) 2024; 16:13237-13251. [PMID: 39614130 DOI: 10.18632/aging.206156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 11/08/2024] [Indexed: 12/01/2024]
Abstract
A thermoregulatory decline occurs with age due to changes in muscle mass, vasoconstriction, and metabolism that lowers core body temperature (Tc). Although lower Tc is a biomarker of successful aging, we have previously shown this worsens cognitive performance in the APP/PS1 mouse model of Alzheimer's disease (AD). We hypothesized that elevating Tc with thermotherapy would improve metabolism and cognition in APP/PS1 mice. From 6-12 months of age, male and female APP/PS1 and C57BL/6 mice were chronically housed at 23 or 30°C. At 12 months of age, mice were assayed for insulin sensitivity, glucose tolerance, and spatial cognition. Plasma, hippocampal, and peripheral (adipose, hepatic, and skeletal muscle) samples were procured postmortem and tissue-specific markers of amyloid accumulation, metabolism, and inflammation were assayed. Chronic 30°C exposure increased Tc in all groups except female APP/PS1 mice. All mice receiving thermotherapy had either improved glucose tolerance or insulin sensitivity, but the underlying processes responsible for these effects varied across sexes. In males, glucose regulation was influenced predominantly by hormonal signaling in plasma and skeletal muscle glucose transporter 4 expression, whereas in females, this was modulated at the tissue level. Thermotherapy improved spatial navigation in male C57BL/6 and APP/PS1 mice, with the later attributed to reduced hippocampal soluble amyloid-β (Aβ)42. Female APP/PS1 mice exhibited worse spatial memory recall after chronic thermotherapy. Together, the data highlights the metabolic benefits of passive thermotherapy, but future studies are needed to determine therapeutic benefits for those with AD.
Collapse
Affiliation(s)
- Samuel A McFadden
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
| | - Mackenzie R Peck
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
| | - Lindsey N Sime
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
| | - MaKayla F Cox
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
| | - Erol D Ikiz
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
| | - Caleigh A Findley
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Kathleen Quinn
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
| | - Yimin Fang
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Erin R Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Kevin N Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| |
Collapse
|
2
|
Tang M, Guo JJ, Guo RX, Xu SJ, Lou Q, Hu QX, Li WY, Yu JB, Yao Q, Wang QW. Progress of research and application of non-pharmacologic intervention in Alzheimer's disease. J Alzheimers Dis 2024; 102:275-294. [PMID: 39573867 DOI: 10.1177/13872877241289396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by amyloid-β (Aβ) deposition and neurofibrillary tangles formed by high phosphorylation of tau protein. At present, drug therapy is the main strategy of AD treatment, but its effects are limited to delaying or alleviating AD. Recently, non-pharmacologic intervention has attracted more attention, and more studies have confirmed that non-pharmacologic intervention in AD can improve the patient's cognitive function and quality of life. This paper summarizes the current non-pharmacologic intervention in AD, to provide useful supplementary means for AD intervention.
Collapse
Affiliation(s)
- Min Tang
- Ningbo Rehabilitation Hospital, Ningbo, Zhejiang, China
| | - Jie-Jie Guo
- The First People's Hospital of Wenling, Taizhou, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Rong-Xia Guo
- School of Teacher Education, Ningbo University, Ningbo, Zhejiang, China
| | - Shu-Jun Xu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Qiong Lou
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qiao-Xia Hu
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Wan-Yi Li
- Ningbo Rehabilitation Hospital, Ningbo, Zhejiang, China
| | - Jing-Bo Yu
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qi Yao
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qin-Wen Wang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
3
|
McFadden SA, Peck MR, Sime LN, Cox MF, Ikiz ED, Findley CA, Quinn K, Fang Y, Bartke A, Hascup ER, Hascup KN. Thermotherapy has Sexually Dimorphic Responses in APP/PS1 Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586836. [PMID: 38586039 PMCID: PMC10996586 DOI: 10.1101/2024.03.26.586836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
A thermoregulatory decline occurs with age due to changes in muscle mass, vasoconstriction, and metabolism that lowers core body temperature (Tc). Although lower Tc is a biomarker of successful aging, we have previously shown this worsens cognitive performance in the APP/PS1 mouse model of Alzheimer's disease (AD) [1]. We hypothesized that elevating Tc with thermotherapy would improve metabolism and cognition in APP/PS1 mice. From 6-12 months of age, male and female APP/PS1 and C57BL/6 mice were chronically housed at 23 or 30°C. At 12 months of age, mice were assayed for insulin sensitivity, glucose tolerance, and spatial cognition. Plasma, hippocampal, and peripheral (adipose, hepatic, and skeletal muscle) samples were procured postmortem and tissue-specific markers of amyloid accumulation, metabolism, and inflammation were assayed. Chronic 30°C exposure increased Tc in all groups except female APP/PS1 mice. All mice receiving thermotherapy had either improved glucose tolerance or insulin sensitivity, but the underlying processes responsible for these effects varied across sexes. In males, glucose regulation was influenced predominantly by hormonal signaling in plasma and skeletal muscle glucose transporter 4 expression, whereas in females, this was modulated at the tissue level. Thermotherapy improved spatial navigation in male C57BL/6 and APP/PS1 mice, with the later attributed to reduced hippocampal soluble amyloid-β (Aβ)42. Female APP/PS1 mice exhibited worse spatial memory recall after chronic thermotherapy. Together, the data highlights the metabolic benefits of passive thermotherapy, but future studies are needed to determine therapeutic benefits for those with AD.
Collapse
Affiliation(s)
- Samuel A. McFadden
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Mackenzie R. Peck
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Lindsey N. Sime
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - MaKayla F. Cox
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Erol D. Ikiz
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Caleigh A. Findley
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
- Department of Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kathleen Quinn
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Yimin Fang
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R. Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
- Department of Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N. Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Neurosciences Institute, Springfield, IL, USA
- Department of Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
4
|
Wang J, Huffman D, Ajwad A, McLouth CJ, Bachstetter A, Kohler K, Murphy MP, O'Hara BF, Duncan MJ, Sunderam S. Thermoneutral temperature exposure enhances slow-wave sleep with a correlated improvement in amyloid pathology in a triple-transgenic mouse model of Alzheimer's disease. Sleep 2024; 47:zsae078. [PMID: 38512801 DOI: 10.1093/sleep/zsae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/14/2024] [Indexed: 03/23/2024] Open
Abstract
Accumulation of amyloid-β (Aβ) plays an important role in Alzheimer's disease (AD) pathology. There is growing evidence that disordered sleep may accelerate AD pathology by impeding the physiological clearance of Aβ from the brain that occurs in normal sleep. Therapeutic strategies for improving sleep quality may therefore help slow disease progression. It is well documented that the composition and dynamics of sleep are sensitive to ambient temperature. We therefore compared Aβ pathology and sleep metrics derived from polysomnography in 12-month-old female 3xTg-AD mice (n = 8) exposed to thermoneutral temperatures during the light period over 4 weeks to those of age- and sex-matched controls (n = 8) that remained at normal housing temperature (22°C) during the same period. The treated group experienced greater proportions of slow wave sleep (SWS)-i.e. epochs of elevated 0.5-2 Hz EEG slow wave activity during non-rapid eye movement (NREM) sleep-compared to controls. Assays performed on mouse brain tissue harvested at the end of the experiment showed that exposure to thermoneutral temperatures significantly reduced levels of DEA-soluble (but not RIPA- or formic acid-soluble) Aβ40 and Aβ42 in the hippocampus, though not in the cortex. With both groups pooled together and without regard to treatment condition, NREM sleep continuity and any measure of SWS within NREM at the end of the treatment period were inversely correlated with DEA-soluble Aβ40 and Aβ42 levels, again in the hippocampus but not in the cortex. These findings suggest that experimental manipulation of SWS could offer useful clues into the mechanisms and treatment of AD.
Collapse
Affiliation(s)
- Jun Wang
- F. Joseph Halcomb III, MD, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| | - Dillon Huffman
- F. Joseph Halcomb III, MD, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| | - Asma'a Ajwad
- F. Joseph Halcomb III, MD, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Diyala College of Medicine, Diyala, Iraq
| | | | - Adam Bachstetter
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Katarina Kohler
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - M Paul Murphy
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Bruce F O'Hara
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | - Marilyn J Duncan
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Sridhar Sunderam
- F. Joseph Halcomb III, MD, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
5
|
Li T, Tian J, Wu M, Tian Y, Li Z. Electroacupuncture stimulation improves cognitive ability and regulates metabolic disorders in Alzheimer's disease model mice: new insights from brown adipose tissue thermogenesis. Front Endocrinol (Lausanne) 2024; 14:1330565. [PMID: 38283741 PMCID: PMC10811084 DOI: 10.3389/fendo.2023.1330565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/26/2023] [Indexed: 01/30/2024] Open
Abstract
Background Metabolic defects play a crucial role in Alzheimer's disease (AD) development. Brown adipose tissue (BAT) has been identified as a novel potential therapeutic target for AD due to its unique role in energy metabolism. Electroacupuncture (EA) shows promise in improving cognitive ability and brain glucose metabolism in AD, but its effects on peripheral and central metabolism are unclear. Methods In this study, SAMP8 mice (AD model) received EA stimulation at specific acupoints. Cognitive abilities were evaluated using the Morris water maze test, while neuronal morphology and tau pathology were assessed through Nissl staining and immunofluorescence staining, respectively. Metabolic variations and BAT thermogenesis were measured using ELISA, HE staining, Western blotting, and infrared thermal imaging. Results Compared to SAMR1 mice, SAMP8 mice showed impaired cognitive ability, neuronal damage, disrupted thermoregulation, and metabolic disorders with low BAT activity. Both the EA and DD groups improved cognitive ability and decreased tau phosphorylation (p<0.01 or p<0.05). However, only the EA group had a significant effect on metabolic disorders and BAT thermogenesis (p<0.01 or p<0.05), while the DD group did not. Conclusion These findings indicate that EA not only improves the cognitive ability of SAMP8 mice, but also effectively regulates peripheral and central metabolic disorders, with this effect being significantly related to the activation of BAT thermogenesis.
Collapse
Affiliation(s)
- Ting Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Junjian Tian
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Meng Wu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanshuo Tian
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhigang Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Landry O, François A, Oye Mintsa Mi-Mba MF, Traversy MT, Tremblay C, Emond V, Bennett DA, Gylys KH, Buxbaum JD, Calon F. Postsynaptic Protein Shank3a Deficiency Synergizes with Alzheimer's Disease Neuropathology to Impair Cognitive Performance in the 3xTg-AD Murine Model. J Neurosci 2023; 43:4941-4954. [PMID: 37253603 PMCID: PMC10312061 DOI: 10.1523/jneurosci.1945-22.2023] [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: 08/09/2022] [Revised: 04/17/2023] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Synaptic loss is intrinsically linked to Alzheimer's disease (AD) neuropathology and symptoms, but its direct impact on clinical symptoms remains elusive. The postsynaptic protein Shank3 (SH3 and multiple ankyrin repeat domains) is of particular interest, as the loss of a single allele of the SHANK3 gene is sufficient to cause profound cognitive symptoms in children. We thus sought to determine whether a SHANK3 deficiency could contribute to the emergence or worsening of AD symptoms and neuropathology. We first found a 30%-50% postmortem loss of SHANK3a associated with cognitive decline in the parietal cortex of individuals with AD. To further probe the role of SHANK3 in AD, we crossed male and female 3xTg-AD mice modelling Aβ and tau pathologies with Shank3a-deficient mice (Shank3Δex4-9). We observed synergistic deleterious effects of Shank3a deficiency and AD neuropathology on object recognition memory at 9, 12, and 18 months of age and on anxious behavior at 9 and 12 months of age in hemizygous Shank3Δex4-9-3xTg-AD mice. In addition to the expected 50% loss of Shank3a, levels of other synaptic proteins, such as PSD-95, drebrin, and homer1, remained unchanged in the parietotemporal cortex of hemizygous Shank3Δex4-9 animals. However, Shank3a deficiency increased the levels of soluble Aβ42 and human tau at 18 months of age compared with 3xTg-AD mice with normal Shank3 expression. The results of this study in human brain samples and in transgenic mice are consistent with the hypothesis that Shank3 deficiency makes a key contribution to cognitive impairment in AD.SIGNIFICANCE STATEMENT Although the loss of several synaptic proteins has been described in Alzheimer's disease (AD), it remains unclear whether their reduction contributes to clinical symptoms. The results of this study in human samples show lower levels of SHANK3a in AD brain, correlating with cognitive decline. Data gathered in a novel transgenic mouse suggest that Shank3a deficiency synergizes with AD neuropathology to induce cognitive impairment, consistent with a causal role in AD. Therefore, treatment aiming at preserving Shank3 in the aging brain may be beneficial to prevent AD.
Collapse
Affiliation(s)
- Olivier Landry
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Arnaud François
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Méryl-Farelle Oye Mintsa Mi-Mba
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Marie-Therese Traversy
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Vincent Emond
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - Karen H Gylys
- School of Nursing, University of California, Los Angeles, California 90095
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York 10029, New York
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| |
Collapse
|
7
|
Carús-Cadavieco M, Berenguer López I, Montoro Canelo A, Serrano-Lope MA, González-de la Fuente S, Aguado B, Fernández-Rodrigo A, Saido TC, Frank García A, Venero C, Esteban JA, Guix F, Dotti CG. Cognitive decline in diabetic mice predisposed to Alzheimer's disease is greater than in wild type. Life Sci Alliance 2023; 6:e202201789. [PMID: 37059474 PMCID: PMC10105330 DOI: 10.26508/lsa.202201789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/16/2023] Open
Abstract
In this work, we tested the hypothesis that the development of dementia in individuals with type 2 diabetes (T2DM) requires a genetic background of predisposition to neurodegenerative disease. As a proof of concept, we induced T2DM in middle-aged hAPP NL/F mice, a preclinical model of Alzheimer's disease. We show that T2DM produces more severe behavioral, electrophysiological, and structural alterations in these mice compared with wild-type mice. Mechanistically, the deficits are not paralleled by higher levels of toxic forms of Aβ or by neuroinflammation but by a reduction in γ-secretase activity, lower levels of synaptic proteins, and by increased phosphorylation of tau. RNA-seq analysis of the cerebral cortex of hAPP NL/F and wild-type mice suggests that the former could be more susceptible to T2DM because of defects in trans-membrane transport. The results of this work, on the one hand, confirm the importance of the genetic background in the severity of the cognitive disorders in individuals with T2DM and, on the other hand, suggest, among the involved mechanisms, the inhibition of γ-secretase activity.
Collapse
Affiliation(s)
- Marta Carús-Cadavieco
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa(CBM), CSIC-UAM, Madrid, Spain
| | - Inés Berenguer López
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa(CBM), CSIC-UAM, Madrid, Spain
| | - Alba Montoro Canelo
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa(CBM), CSIC-UAM, Madrid, Spain
- Escuela Técnica Superior (E.T.S) de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid, Madrid, Spain
| | - Miguel A Serrano-Lope
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa(CBM), CSIC-UAM, Madrid, Spain
| | | | - Begoña Aguado
- Genomics and NGS Facility, Centro de Biología Molecular Severo Ochoa(CBM) CSIC-UAM, Madrid, Spain
| | - Alba Fernández-Rodrigo
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa(CBM), CSIC-UAM, Madrid, Spain
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Ana Frank García
- Department of Neurology, Division Neurodegenerative Disease, University Hospital La Paz, Madrid, Spain
| | - César Venero
- Department of Psychobiology, Universidad Nacional de Educación a Distancia, Madrid, Spain
| | - José A Esteban
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa(CBM), CSIC-UAM, Madrid, Spain
| | - Francesc Guix
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa(CBM), CSIC-UAM, Madrid, Spain
- Department of Bioengineering, Institut Químic de Sarrià (IQS) - Universitat Ramón Llull (URL), Barcelona, Spain
| | - Carlos G Dotti
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa(CBM), CSIC-UAM, Madrid, Spain
| |
Collapse
|
8
|
Brain temperature in healthy and diseased conditions: A review on the special implications of MRS for monitoring brain temperature. Biomed Pharmacother 2023; 160:114287. [PMID: 36709597 DOI: 10.1016/j.biopha.2023.114287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/30/2023] Open
Abstract
Brain temperature determines not only an individual's cognitive functionality but also the prognosis and mortality rates of many brain diseases. More specifically, brain temperature not only changes in response to different physiological events like yawning and stretching, but also plays a significant pathophysiological role in a number of neurological and neuropsychiatric illnesses. Here, we have outlined the function of brain hyperthermia in both diseased and healthy states, focusing particularly on the amyloid beta aggregation in Alzheimer's disease.
Collapse
|
9
|
Alagiakrishnan K, Dhami P, Senthilselvan A. Predictors of Conversion to Dementia in Patients With Mild Cognitive Impairment: The Role of Low Body Temperature. J Clin Med Res 2023; 15:216-224. [PMID: 37187716 PMCID: PMC10181356 DOI: 10.14740/jocmr4883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
Background Subjects with mild cognitive impairment (MCI) can progress to dementia. Studies have shown that neuropsychological tests, biological or radiological markers individually or in combination have helped to determine the risk of conversion from MCI to dementia. These techniques are complex and expensive, and clinical risk factors were not considered in these studies. This study examined demographic, lifestyle and clinical factors including low body temperature that may play a role in the conversion of MCI to dementia in elderly patients. Methods In this retrospective study, a chart review was conducted on patients aged 61 to 103 years who were seen at the University of Alberta Hospital. Information on onset of MCI and demographic, social, and lifestyle factors, family history of dementia and clinical factors, and current medications at baseline was collected from patient charts on an electronic database. The conversion from MCI to dementia within 5.5 years was also determined. Logistic regression analysis was conducted to identify the baseline factors associated with conversion from MCI to dementia. Results The prevalence of MCI at baseline was 25.6% (335/1,330). During the 5.5 years follow-up period, 43% (143/335) of the subjects converted to dementia from MCI. The factors that were significantly associated with conversion from MCI to dementia were family history of dementia (odds ratio (OR): 2.78, 95% confidence interval (CI): 1.56 - 4.95, P = 0.001), Montreal cognitive assessment (MoCA) score (OR: 0.91, 95% CI: 0.85 - 0.97, P = 0.01), and low body temperature (below 36 °C) (OR: 10.01, 95% CI: 3.59 - 27.88, P < 0.001). Conclusion In addition to family history of dementia and MoCA, low body temperature was shown to be associated with the conversion from MCI to dementia. This study would help clinicians to identify patients with MCI who are at highest risk of conversion to dementia.
Collapse
Affiliation(s)
- Kannayiram Alagiakrishnan
- Division of Geriatric Medicine, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Prabhpaul Dhami
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
10
|
Chen P, Zhao K, Zhang H, Wei Y, Wang P, Wang D, Song C, Yang H, Zhang Z, Yao H, Qu Y, Kang X, Du K, Fan L, Han T, Yu C, Zhou B, Jiang T, Zhou Y, Lu J, Han Y, Zhang X, Liu B, Liu Y. Altered global signal topography in Alzheimer's disease. EBioMedicine 2023; 89:104455. [PMID: 36758481 PMCID: PMC9941064 DOI: 10.1016/j.ebiom.2023.104455] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 12/31/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease associated with widespread disruptions in intrinsic local specialization and global integration in the functional system of the brain. These changes in integration may further disrupt the global signal (GS) distribution, which might represent the local relative contribution to global activity in functional magnetic resonance imaging (fMRI). METHODS fMRI scans from a discovery dataset (n = 809) and a validated dataset (n = 542) were used in the analysis. We investigated the alteration of GS topography using the GS correlation (GSCORR) in patients with mild cognitive impairment (MCI) and AD. The association between GS alterations and functional network properties was also investigated based on network theory. The underlying mechanism of GSCORR alterations was elucidated using imaging-transcriptomics. FINDINGS Significantly increased GS topography in the frontal lobe and decreased GS topography in the hippocampus, cingulate gyrus, caudate, and middle temporal gyrus were observed in patients with AD (Padj < 0.05). Notably, topographical GS changes in these regions correlated with cognitive ability (P < 0.05). The changes in GS topography also correlated with the changes in functional network segregation (ρ = 0.5). Moreover, the genes identified based on GS topographical changes were enriched in pathways associated with AD and neurodegenerative diseases. INTERPRETATION Our findings revealed significant changes in GS topography and its molecular basis, confirming the informative role of GS in AD and further contributing to the understanding of the relationship between global and local neuronal activities in patients with AD. FUNDING Beijing Natural Science Funds for Distinguished Young Scholars, China; Fundamental Research Funds for the Central Universities, China; National Natural Science Foundation, China.
Collapse
Affiliation(s)
- Pindong Chen
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Kun Zhao
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China; Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing, China
| | - Han Zhang
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Yongbin Wei
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital Tianjin University, Tianjin, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Chengyuan Song
- Department of Neurology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Hongwei Yang
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | | | - Hongxiang Yao
- Department of Radiology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yida Qu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaopeng Kang
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Kai Du
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Lingzhong Fan
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bo Zhou
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Tianzi Jiang
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Yuying Zhou
- Department of Neurology, Tianjin Huanhu Hospital Tianjin University, Tianjin, China
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Institute of Geriatrics, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Xi Zhang
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Bing Liu
- State Key Laboratory of Cognition Neuroscience & Learning, Beijing Normal University, Beijing, China
| | - Yong Liu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China.
| | | |
Collapse
|
11
|
Leclerc M, Bourassa P, Tremblay C, Caron V, Sugère C, Emond V, Bennett DA, Calon F. Cerebrovascular insulin receptors are defective in Alzheimer's disease. Brain 2023; 146:75-90. [PMID: 36280236 PMCID: PMC9897197 DOI: 10.1093/brain/awac309] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/24/2022] [Accepted: 08/12/2022] [Indexed: 01/11/2023] Open
Abstract
Central response to insulin is suspected to be defective in Alzheimer's disease. As most insulin is secreted in the bloodstream by the pancreas, its capacity to regulate brain functions must, at least partly, be mediated through the cerebral vasculature. However, how insulin interacts with the blood-brain barrier and whether alterations of this interaction could contribute to Alzheimer's disease pathophysiology both remain poorly defined. Here, we show that human and murine cerebral insulin receptors (INSRs), particularly the long isoform INSRα-B, are concentrated in microvessels rather than in the parenchyma. Vascular concentrations of INSRα-B were lower in the parietal cortex of subjects diagnosed with Alzheimer's disease, positively correlating with cognitive scores, leading to a shift towards a higher INSRα-A/B ratio, consistent with cerebrovascular insulin resistance in the Alzheimer's disease brain. Vascular INSRα was inversely correlated with amyloid-β plaques and β-site APP cleaving enzyme 1, but positively correlated with insulin-degrading enzyme, neprilysin and P-glycoprotein. Using brain cerebral intracarotid perfusion, we found that the transport rate of insulin across the blood-brain barrier remained very low (<0.03 µl/g·s) and was not inhibited by an insulin receptor antagonist. However, intracarotid perfusion of insulin induced the phosphorylation of INSRβ that was restricted to microvessels. Such an activation of vascular insulin receptor was blunted in 3xTg-AD mice, suggesting that Alzheimer's disease neuropathology induces insulin resistance at the level of the blood-brain barrier. Overall, the present data in post-mortem Alzheimer's disease brains and an animal model of Alzheimer's disease indicate that defects in the insulin receptor localized at the blood-brain barrier strongly contribute to brain insulin resistance in Alzheimer's disease, in association with β-amyloid pathology.
Collapse
Affiliation(s)
- Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC G1V 4G2, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada
| | - Philippe Bourassa
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC G1V 4G2, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC G1V 4G2, Canada
| | - Vicky Caron
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC G1V 4G2, Canada
| | - Camille Sugère
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC G1V 4G2, Canada
| | - Vincent Emond
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC G1V 4G2, Canada
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC G1V 4G2, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada
| |
Collapse
|
12
|
Tayanloo-Beik A, Nikkhah A, Alaei S, Goodarzi P, Rezaei-Tavirani M, Mafi AR, Larijani B, Shouroki FF, Arjmand B. Brown adipose tissue and alzheimer's disease. Metab Brain Dis 2023; 38:91-107. [PMID: 36322277 DOI: 10.1007/s11011-022-01097-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/01/2022] [Indexed: 01/12/2023]
Abstract
Alzheimer's disease (AD), the most common type of senile dementia, is a chronic neurodegenerative disease characterized by cognitive dysfunction and behavioral disability. The two histopathological hallmarks in this disease are the extraneuronal accumulation of amyloid-β (Aβ) and the intraneuronal deposition of neurofibrillary tangles (NFTs). Despite this, central and peripheral metabolic dysfunction, such as abnormal brain signaling, insulin resistance, inflammation, and impaired glucose utilization, have been indicated to be correlated with AD. There is solid evidence that the age-associated thermoregulatory deficit induces diverse metabolic changes associated with AD development. Brown adipose tissue (BAT) has been known as a thermoregulatory organ particularly vital during infancy. However, in recent years, BAT has been accepted as an endocrine organ, being involved in various functions that prevent AD, such as regulating energy metabolism, secreting hormones, improving insulin sensitivity, and increasing glucose utilization in adult humans. This review focuses on the mechanisms of BAT activation and the effect of aging on BAT production and signaling. Specifically, the evidence demonstrating the effect of BAT on pathological mechanisms influencing the development of AD, including insulin pathway, thermoregulation, and other hormonal pathways, are reviewed in this article.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirabbas Nikkhah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Setareh Alaei
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ahmad Rezazadeh Mafi
- Department of Radiation Oncology, Imam Hossein Hospital, Shaheed Beheshti Medical University, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran.
| | - Fatemeh Fazeli Shouroki
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Husain MA, Vachon A, Chouinard-Watkins R, Vandal M, Calon F, Plourde M. Investigating the plasma-liver-brain axis of omega-3 fatty acid metabolism in mouse knock-in for the human apolipoprotein E epsilon 4 allele. J Nutr Biochem 2023; 111:109181. [PMID: 36220526 DOI: 10.1016/j.jnutbio.2022.109181] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 06/30/2022] [Accepted: 09/13/2022] [Indexed: 11/06/2022]
Abstract
The metabolism of docosahexaenoic acid (DHA), an omega-3 fatty acid, is different in carriers of APOE4, the main genetic risk factor for late-onset Alzheimer's disease. The brain relies on the plasma DHA pool for its need, but the plasma-liver-brain axis in relation to cognition remains obscure. We hypothesized that this relationship is compromised in APOE4 mice considering the differences in fatty acid metabolism between APOE3 and APOE4 mice. Male and female APOE3 and APOE4 mice were fed either a diet enriched with DHA (0.7 g DHA/100 g diet) or a control diet for 8 months. There was a significant genotype × diet interaction for DHA concentration in the liver and adipose tissue. In the cortex, a genotype effect was found where APOE4 mice had a higher concentration of DHA than APOE3 mice fed the control diet. There was a significant genotype × diet interaction for the liver and hippocampal arachidonic acid (AA). APOE4 mice had 20-30% lower plasma DHA and AA concentrations than APOE3 mice, independent of diet. Plasma and liver DHA levels were significantly correlated in APOE3 and APOE4 mice. In APOE4 mice, there was a significant correlation between plasma, adipose tissues, cortex DHA and the Barnes maze and/or with a better recognition index. Moreover, higher AA levels in the liver and the hippocampus of APOE4 mice were correlated with lower cognitive performance. Our results suggest that there is a plasma-liver-brain axis of DHA that is modified in APOE4 mice. Moreover, our data support that APOE4 mice rely more on plasma DHA than APOE3 mice, especially in cognitive performance. Any disturbance in plasma DHA metabolism might have a greater impact on cognition in APOE4 carriers.
Collapse
Affiliation(s)
- Mohammed Amir Husain
- Centre de Recherche sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada; Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Annick Vachon
- Centre de Recherche sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Milène Vandal
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Frédéric Calon
- Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, Quebec, Canada; Faculté de pharmacie et center de recherche du CHU de Québec-Université Laval, Quebec, Canada
| | - Mélanie Plourde
- Centre de Recherche sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada; Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada; Institut de la nutrition et des aliments fonctionnels, Université Laval, Québec, Quebec, Canada.
| |
Collapse
|
14
|
McFadden S, Sime LN, Cox MKF, Findley CA, Peck MR, Quinn K, Fang Y, Bartke A, Hascup ER, Hascup KN. Chronic, Mild Hypothermic Environmental Temperature does not Ameliorate Cognitive Deficits in an Alzheimer's Disease Mouse. J Gerontol A Biol Sci Med Sci 2022:6832816. [PMID: 36398842 DOI: 10.1093/gerona/glac223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Indexed: 11/19/2022] Open
Abstract
Metabolic dysfunction increases with age and is a contributing factor to Alzheimer's disease (AD) development. We have previously observed impaired insulin sensitivity and glucose homeostasis in the APP/PS1 model of AD. To improve these parameters, we chronically exposed male and female mice to mild hypothermic environmental temperature (eT), which positively modulates metabolism. Although a hypothermic eT normalized insulin sensitivity, glucose tolerance was still impaired in both sexes of AD mice. We observed increased plasma glucagon and BAFF in both sexes, but additional sexually dimorphic mechanisms may explain the impaired glucose homeostasis in AD mice. Hepatic Glut2 was decreased in female while visceral adipose tissue TNFα was increased in male APP/PS1 mice. A mild hypothermic eT did not improve spatial learning and memory in either sex and increased amyloid plaque burden in male APP/PS1 mice. Overall, plasma markers of glucose homeostasis and AD pathology were worse in female compared to male APP/PS1 mice suggesting a faster disease progression. This could affect therapeutic outcome if interventional strategies are administered at the same chronological age to male and female APP/PS1 mice. Furthermore, this data suggests a dichotomy exists between mechanisms to improve metabolic function and cognitive health that may be further impaired in AD.
Collapse
Affiliation(s)
- Samuel McFadden
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Lindsey N Sime
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Ma Kayla F Cox
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Caleigh A Findley
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neurosciences Institute, Springfield, IL, USA.,Department of Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Mackenzie R Peck
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Kathleen Quinn
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Yimin Fang
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neurosciences Institute, Springfield, IL, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neurosciences Institute, Springfield, IL, USA.,Department of Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neurosciences Institute, Springfield, IL, USA.,Department of Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
15
|
Blessing EM, Parekh A, Betensky RA, Babb J, Saba N, Debure L, Varga AW, Ayappa I, Rapoport DM, Butler TA, de Leon MJ, Wisniewski T, Lopresti BJ, Osorio RS. Association between lower body temperature and increased tau pathology in cognitively normal older adults. Neurobiol Dis 2022; 171:105748. [PMID: 35550158 PMCID: PMC9751849 DOI: 10.1016/j.nbd.2022.105748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/25/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Preclinical studies suggest body temperature (Tb) and consequently brain temperature has the potential to bidirectionally interact with tau pathology in Alzheimer's Disease (AD). Tau phosphorylation is substantially increased by a small (<1 °C) decrease in temperature within the human physiological range, and thermoregulatory nuclei are affected by tau pathology early in the AD continuum. In this study we evaluated whether Tb (as a proxy for brain temperature) is cross-sectionally associated with clinically utilized markers of tau pathology in cognitively normal older adults. METHODS Tb was continuously measured with ingestible telemetry sensors for 48 h. This period included two nights of nocturnal polysomnography to delineate whether Tb during waking vs sleep is differentially associated with tau pathology. Tau phosphorylation was assessed with plasma and cerebrospinal fluid (CSF) tau phosphorylated at threonine 181 (P-tau), sampled the day following Tb measurement. In addition, neurofibrillary tangle (NFT) burden in early Braak stage regions was imaged with PET-MR using the [18F]MK-6240 radiotracer on average one month later. RESULTS Lower Tb was associated with increased NFT burden, as well as increased plasma and CSF P-tau levels (p < 0.05). NFT burden was associated with lower Tb during waking (p < 0.05) but not during sleep intervals. Plasma and CSF P-tau levels were highly correlated with each other (p < 0.05), and both variables were correlated with tau tangle radiotracer uptake (p < 0.05). CONCLUSIONS These results, the first available for human, suggest that lower Tb in older adults may be associated with increased tau pathology. Our findings add to the substantial preclinical literature associating lower body and brain temperature with tau hyperphosphorylation. CLINICAL TRIAL NUMBER NCT03053908.
Collapse
Affiliation(s)
- Esther M Blessing
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Ankit Parekh
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States of America.
| | - Rebecca A Betensky
- Department of NYU School of Global Public Health, New York, NY 10016, United States of America.
| | - James Babb
- Alzheimer's Disease Research Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Natalie Saba
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Ludovic Debure
- Alzheimer's Disease Research Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Andrew W Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States of America.
| | - Indu Ayappa
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States of America.
| | - David M Rapoport
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States of America.
| | - Tracy A Butler
- Department of Neurology, Weill Cornell Medicine, New York, NY 10065, United States of America.
| | - Mony J de Leon
- Department of Neurology, Weill Cornell Medicine, New York, NY 10065, United States of America.
| | - Thomas Wisniewski
- Alzheimer's Disease Research Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15213, United States of America.
| | - Ricardo S Osorio
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, United States of America; Alzheimer's Disease Research Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| |
Collapse
|
16
|
Frankle L. Entropy, Amnesia, and Abnormal Déjà Experiences. Front Psychol 2022; 13:794683. [PMID: 35967717 PMCID: PMC9364811 DOI: 10.3389/fpsyg.2022.794683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Previous research has contrasted fleeting erroneous experiences of familiarity with equally convincing, and often more stubborn erroneous experiences of remembering. While a subset of the former category may present as nonpathological “déjà vu,” the latter, termed “déjà vécu” can categorize a delusion-like confabulatory phenomenon first described in elderly dementia patients. Leading explanations for this experience include the dual process view, in which erroneous familiarity and erroneous recollection are elicited by inappropriate activation of the parahippocampal cortex and the hippocampus, respectively, and the more popular encoding-as-retrieval explanation in which normal memory encoding processes are falsely flagged and interpreted as memory retrieval. This paper presents a novel understanding of this recollective confabulation that builds on the encoding-as-retrieval hypothesis but more adequately accounts for the co-occurrence of persistent déjà vécu with both perceptual novelty and memory impairment, the latter of which occurs not only in progressive dementia but also in transient epileptic amnesia (TEA) and psychosis. It makes use of the growing interdisciplinary understanding of the fluidity of time and posits that the functioning of memory and the perception of novelty, long known to influence the subjective experience of time, may have a more fundamental effect on the flow of time.
Collapse
|
17
|
Asfuroğlu BB, Topkan TA, Kaydu NE, Sakai K, Öner AY, Karaman Y, Yamada K, Tali ET. DWI-based MR thermometry: could it discriminate Alzheimer's disease from mild cognitive impairment and healthy subjects? Neuroradiology 2022; 64:1979-1987. [PMID: 35536331 DOI: 10.1007/s00234-022-02969-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE The aim of this study is to compare lateral ventricular cerebrospinal fluid (CSF) temperature of the patients with Alzheimer's disease (AD), mild cognitive impairment (MCI), and healthy subjects (HS) using diffusion-weighted imaging (DWI)-based magnetic resonance (MR) thermometry. METHODS Seventy-two patients (37 AD, 19 MCI, 16 HS) who underwent 3-T MR examination from September 2018 to August 2019 were included in this study. Smoking habits, education level, disease duration, and comorbidity status were recorded. Patients were assessed using Mini-Mental State Examination (MMSE) and the Clinical Dementia Rating (CDR) score. Brain temperatures were measured using DWI-based MR thermometry. Group comparisons of brain temperature were performed using the Pearson chi-square, Mann-Whitney, and Kruskal-Wallis tests. Further analysis was performed using the post hoc Bonferroni test. Receiver operating characteristic (ROC) analysis was also used. RESULTS A CDR score of 0.5, 1, and 2 was 2 (5.4%), 14 (37.8%), and 21 (56.8%) in AD, respectively. The median MMSE score had significant differences among groups and also in pairwise comparisons. The median CSF temperature (°C) values showed statistically significant difference among groups (HS: 38.5 °C, MCI: 38.17 °C, AD: 38.0 °C). The post hoc Mann-Whitney U test indicated a significant difference between AD patients and HS (p = 0.009). There were no significant CSF temperature differences in other pairwise comparisons. CONCLUSION Lower CSF temperatures were observed in AD patients than in HS, probably due to decreased brain metabolism in AD. DWI-based MR thermometry as a noninvasive imaging method enabling the measurement of CSF temperatures may contribute to the diagnosis of AD.
Collapse
Affiliation(s)
- Berrak Barutcu Asfuroğlu
- Department of Radiology, Faculty of Medicine, School of Medicine, Gazi University, 06500, Besevler, Ankara, Turkey.
| | - Tuğberk Andaç Topkan
- Department of Neurology, Faculty of Medicine, School of Medicine, Gazi University, Ankara, Turkey
| | - Nesrin Erdoğan Kaydu
- Department of Radiology, Faculty of Medicine, School of Medicine, Gazi University, 06500, Besevler, Ankara, Turkey
| | - Koji Sakai
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ali Yusuf Öner
- Department of Radiology, Faculty of Medicine, School of Medicine, Gazi University, 06500, Besevler, Ankara, Turkey
| | - Yahya Karaman
- Department of Neurology, Faculty of Medicine, School of Medicine, Gazi University, Ankara, Turkey
| | - Kei Yamada
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - E Turgut Tali
- Department of Radiology, Faculty of Medicine, School of Medicine, Gazi University, 06500, Besevler, Ankara, Turkey.,Department of Radiology, School of Medicine, Lokman Hekim University, Ankara, Turkey
| |
Collapse
|
18
|
Guzzardi MA, La Rosa F, Campani D, Collado MC, Monleon D, Cacciato Insilla A, Tripodi M, Zega A, Dattilo A, Brunetto MR, Maffei M, Bonino F, Iozzo P. Liver and White/Brown Fat Dystrophy Associates with Gut Microbiota and Metabolomic Alterations in 3xTg Alzheimer’s Disease Mouse Model. Metabolites 2022; 12:metabo12040278. [PMID: 35448465 PMCID: PMC9028874 DOI: 10.3390/metabo12040278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolic impairments and liver and adipose depots alterations were reported in subjects with Alzheimer’s disease (AD), highlighting the role of the liver–adipose–tissue–brain axis in AD pathophysiology. The gut microbiota might play a modulating role. We investigated the alterations to the liver and white/brown adipose tissues (W/BAT) and their relationships with serum and gut metabolites and gut bacteria in a 3xTg mouse model during AD onset (adulthood) and progression (aging) and the impact of high-fat diet (HFD) and intranasal insulin (INI). Glucose metabolism (18FDG-PET), tissue radiodensity (CT), liver and W/BAT histology, BAT-thermogenic markers were analyzed. 16S-RNA sequencing and mass-spectrometry were performed in adult (8 months) and aged (14 months) 3xTg-AD mice with a high-fat or control diet. Generalized and HFD resistant deficiency of lipid accumulation in both liver and W/BAT, hypermetabolism in WAT (adulthood) and BAT (aging), abnormal cytokine–hormone profiles, and liver inflammation were observed in 3xTg mice; INI could antagonize all these alterations. Specific gut microbiota–metabolome profiles correlated with a significant disruption of the gut–microbiota–liver–adipose axis in AD mice. In conclusion, fat dystrophy in liver and adipose depots contributes to AD progression, and associates with altered profiles of the gut microbiota, which candidates as an appealing early target for preventive intervention.
Collapse
Affiliation(s)
- Maria Angela Guzzardi
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
- Correspondence: ; Tel.: +39-050-3152722
| | - Federica La Rosa
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| | - Daniela Campani
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, Division of Pathology, Pisa University Hospital, 56124 Pisa, Italy; (D.C.); (A.C.I.)
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain;
| | - Daniel Monleon
- Faculty of Medicine, Health Research Institute INCLIVA/CIBERFES for Frailty and Healthy Aging, University of Valencia, 46003 Valencia, Spain;
| | - Andrea Cacciato Insilla
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, Division of Pathology, Pisa University Hospital, 56124 Pisa, Italy; (D.C.); (A.C.I.)
| | - Maria Tripodi
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| | - Alessandro Zega
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| | | | - Maurizia Rossana Brunetto
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy;
- Hepatology Unit, Department of Medical Specialties, Laboratory of Molecular Genetics and Pathology of Hepatitis Viruses, Pisa University Hospital, 56124 Pisa, Italy
- Institute of Biostructure and Bioimaging (IBB), National Research Council (CNR), 80145 Napoli, Italy;
| | - Margherita Maffei
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| | - Ferruccio Bonino
- Institute of Biostructure and Bioimaging (IBB), National Research Council (CNR), 80145 Napoli, Italy;
| | - Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy; (F.L.R.); (M.T.); (A.Z.); (M.M.); (P.I.)
| |
Collapse
|
19
|
Paul PS, Cho JY, Wu Q, Karthivashan G, Grabovac E, Wille H, Kulka M, Kar S. Unconjugated PLGA nanoparticles attenuate temperature-dependent β-amyloid aggregation and protect neurons against toxicity: implications for Alzheimer's disease pathology. J Nanobiotechnology 2022; 20:67. [PMID: 35120558 PMCID: PMC8817552 DOI: 10.1186/s12951-022-01269-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 01/16/2022] [Indexed: 12/26/2022] Open
Abstract
Conversion of β-amyloid (Aβ) peptides from soluble random-coil to aggregated protein enriched with β-sheet-rich intermediates has been suggested to play a role in the degeneration of neurons and development of Alzheimer's disease (AD) pathology. Aggregation of Aβ peptide can be prompted by a variety of environmental factors including temperature which can influence disease pathogenesis. Recently, we reported that FDA-approved unconjugated poly (D,L-lactide-co-glycolide) (PLGA) nanoparticles can have beneficial effects in cellular and animal models of AD by targeting different facets of the Aβ axis. In this study, using biochemical, structural and spectroscopic analyses, we evaluated the effects of native PLGA on temperature-dependent Aβ aggregation and its ability to protect cultured neurons from degeneration. Our results show that the rate of spontaneous Aβ1-42 aggregation increases with a rise in temperature from 27 to 40 °C and PLGA with 50:50 resomer potently inhibits Aβ aggregation at all temperatures, but the effect is more profound at 27 °C than at 40 °C. It appears that native PLGA, by interacting with the hydrophobic domain of Aβ1-42, prevents a conformational shift towards β-sheet structure, thus precluding the formation of Aβ aggregates. Additionally, PLGA triggers disassembly of matured Aβ1-42 fibers at a faster rate at 40 °C than at 27 °C. PLGA-treated Aβ samples can significantly enhance viability of cortical cultured neurons compared to neurons treated with Aβ alone by attenuating phosphorylation of tau protein. Injection of native PLGA is found to influence the breakdown/clearance of Aβ peptide in the brain. Collectively, these results suggest that PLGA nanoparticles can inhibit Aβ aggregation and trigger disassembly of Aβ aggregates at temperatures outside the physiological range and can protect neurons against Aβ-mediated toxicity thus validating its unique therapeutic potential in the treatment of AD pathology.
Collapse
Affiliation(s)
- Pallabi Sil Paul
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| | - Jae-Young Cho
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9 Canada
| | - Qi Wu
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| | - Govindarajan Karthivashan
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| | - Emily Grabovac
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9 Canada
| | - Holger Wille
- Department of Biochemistry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| | - Mariana Kulka
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9 Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1 Canada
| | - Satyabrata Kar
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
- Departments of Medicine (Neurology) and Psychiatry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| |
Collapse
|
20
|
Sauna-like conditions or menthol treatment reduce tau phosphorylation through mild hyperthermia. Neurobiol Aging 2022; 113:118-130. [DOI: 10.1016/j.neurobiolaging.2022.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 02/02/2022] [Accepted: 02/20/2022] [Indexed: 02/08/2023]
|
21
|
Tournissac M, Leclerc M, Valentin-Escalera J, Vandal M, Bosoi CR, Planel E, Calon F. Metabolic determinants of Alzheimer's disease: A focus on thermoregulation. Ageing Res Rev 2021; 72:101462. [PMID: 34534683 DOI: 10.1016/j.arr.2021.101462] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/09/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a complex age-related neurodegenerative disease, associated with central and peripheral metabolic anomalies, such as impaired glucose utilization and insulin resistance. These observations led to a considerable interest not only in lifestyle-related interventions, but also in repurposing insulin and other anti-diabetic drugs to prevent or treat dementia. Body temperature is the oldest known metabolic readout and mechanisms underlying its maintenance fail in the elderly, when the incidence of AD rises. This raises the possibility that an age-associated thermoregulatory deficit contributes to energy failure underlying AD pathogenesis. Brown adipose tissue (BAT) plays a central role in thermogenesis and maintenance of body temperature. In recent years, the modulation of BAT activity has been increasingly demonstrated to regulate energy expenditure, insulin sensitivity and glucose utilization, which could also provide benefits for AD. Here, we review the evidence linking thermoregulation, BAT and insulin-related metabolic defects with AD, and we propose mechanisms through which correcting thermoregulatory impairments could slow the progression and delay the onset of AD.
Collapse
|
22
|
Bongioanni P, Del Carratore R, Corbianco S, Diana A, Cavallini G, Masciandaro SM, Dini M, Buizza R. Climate change and neurodegenerative diseases. ENVIRONMENTAL RESEARCH 2021; 201:111511. [PMID: 34126048 DOI: 10.1016/j.envres.2021.111511] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/25/2021] [Accepted: 06/08/2021] [Indexed: 06/12/2023]
Abstract
The climate change induced global warming, and in particular the increased frequency and intensity of heat waves, have been linked to health problems. Among them, scientific works have been reporting an increased incidence of neurological diseases, encompassing also neurodegenerative ones, such as Dementia of Alzheimer's type, Parkinson's Disease, and Motor Neuron Diseases. Although the increase in prevalence of neurodegenerative diseases is well documented by literature reports, the link between global warming and the enhanced prevalence of such diseases remains elusive. This is the main theme of our work, which aims to examine the connection between high temperature exposure and neurodegenerative diseases. Firstly, we evaluate the influence of high temperatures exposure on the pathophysiology of these disorders. Secondly, we discuss its effects on the thermoregulation, already compromised in affected patients, and its interference with processes of excitotoxicity, oxidative stress and neuroinflammation, all of them related with neurodegeneration. Finally, we investigate chronic versus acute stressors on body warming, and put forward a possible interpretation of the beneficial or detrimental effects on the brain, which is responsible for the incidence or progression of neurological disorders.
Collapse
Affiliation(s)
- Paolo Bongioanni
- Severe Acquired Brain Injuries Dpt Section, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; NeuroCare Onlus, Pisa, Italy
| | | | - Silvia Corbianco
- Interdepartmental Research Centre on Biology and Pathology of Aging, University of Pisa, Italy; Human Movement and Rehabilitation Research Laboratory, Pisa, Italy
| | - Andrea Diana
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Gabriella Cavallini
- Interdepartmental Research Centre on Biology and Pathology of Aging, University of Pisa, Italy
| | - Silvia M Masciandaro
- NeuroCare Onlus, Pisa, Italy; Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Marco Dini
- Interdepartmental Research Centre on Biology and Pathology of Aging, University of Pisa, Italy; Human Movement and Rehabilitation Research Laboratory, Pisa, Italy
| | - Roberto Buizza
- Scuola Superiore Sant'Anna and Centre for Climate Change Studies and Sustainable Actions (3CSA), Pisa, Italy
| |
Collapse
|
23
|
Kim E, Nohara K, Wirianto M, Escobedo G, Lim JY, Morales R, Yoo SH, Chen Z. Effects of the Clock Modulator Nobiletin on Circadian Rhythms and Pathophysiology in Female Mice of an Alzheimer's Disease Model. Biomolecules 2021; 11:biom11071004. [PMID: 34356628 PMCID: PMC8301787 DOI: 10.3390/biom11071004] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder and the most common cause of dementia. Various pathogenic mechanisms have been proposed to contribute to disease progression, and recent research provided evidence linking dysregulated circadian rhythms/sleep and energy metabolism with AD. Previously, we found that the natural compound Nobiletin (NOB) can directly activate circadian cellular oscillators to promote metabolic health in disease models and healthy aging in naturally aged mice. In the current study, using the amyloid-β AD model APP/PS1, we investigated circadian, metabolic and amyloid characteristics of female mice and the effects of NOB. Female APP/PS1 mice showed reduced sleep bout duration, and NOB treatment exhibited a trend to improve it. While glucose tolerance was unchanged, female APP/PS1 mice displayed exaggerated oxygen consumption and CO2 production, which was mitigated by NOB. Likewise, cold tolerance in APP/PS1 was impaired relative to WT, and interestingly was markedly enhanced in NOB-treated APP/PS1 mice. Although circadian behavioral rhythms were largely unchanged, real-time qPCR analysis revealed altered expression of several core clock genes by NOB in the cerebral cortex, notably Bmal1, Npas2, and Rora. Moreover, NOB was also able to activate various clock-controlled metabolic genes involved in insulin signaling and mitochondrial function, including Igf1, Glut1, Insr, Irs1, Ucp2, and Ucp4. Finally, we observed that NOB attenuated the expression of several AD related genes including App, Bace1, and ApoE, reduced APP protein levels, and strongly ameliorated Aβ pathology in the cortex. Collectively, these results reveal novel genotype differences and importantly beneficial effects of a natural clock-enhancing compound in biological rhythms and related pathophysiology, suggesting the circadian clock as a modifiable target for AD.
Collapse
Affiliation(s)
- Eunju Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Kazunari Nohara
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Marvin Wirianto
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Gabriel Escobedo
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (G.E.J.); (R.M.)
| | - Ji Ye Lim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (G.E.J.); (R.M.)
- Centro Integrativo de Biologia y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370993, Chile
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.K.); (K.N.); (M.W.); (J.Y.L.); (S.-H.Y.)
- Correspondence:
| |
Collapse
|
24
|
Tournissac M, Vu TM, Vrabic N, Hozer C, Tremblay C, Mélançon K, Planel E, Pifferi F, Calon F. Repurposing beta-3 adrenergic receptor agonists for Alzheimer's disease: beneficial effects in a mouse model. ALZHEIMERS RESEARCH & THERAPY 2021; 13:103. [PMID: 34020681 PMCID: PMC8140479 DOI: 10.1186/s13195-021-00842-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/04/2021] [Indexed: 12/14/2022]
Abstract
Background Old age, the most important risk factor for Alzheimer’s disease (AD), is associated with thermoregulatory deficits. Brown adipose tissue (BAT) is the main thermogenic driver in mammals and its stimulation, through β3 adrenergic receptor (β3AR) agonists or cold acclimation, counteracts metabolic deficits in rodents and humans. Studies in animal models show that AD neuropathology leads to thermoregulatory deficits, and cold-induced tau hyperphosphorylation is prevented by BAT stimulation through cold acclimation. Since metabolic disorders and AD share strong pathogenic links, we hypothesized that BAT stimulation through a β3AR agonist could exert benefits in AD as well. Methods CL-316,243, a specific β3AR agonist, was administered to the triple transgenic mouse model of AD (3xTg-AD) and non-transgenic controls from 15 to 16 months of age at a dose of 1 mg/kg/day i.p. Results Here, we show that β3AR agonist administration decreased body weight and improved peripheral glucose metabolism and BAT thermogenesis in both non-transgenic and 3xTg-AD mice. One-month treatment with a β3AR agonist increased recognition index by 19% in 16-month-old 3xTg-AD mice compared to pre-treatment (14-month-old). Locomotion, anxiety, and tau pathology were not modified. Finally, insoluble Aβ42/Aβ40 ratio was decreased by 27% in the hippocampus of CL-316,243-injected 3xTg-AD mice. Conclusions Overall, our results indicate that β3AR stimulation reverses memory deficits and shifts downward the insoluble Aβ42/Aβ40 ratio in 16-month-old 3xTg-AD mice. As β3AR agonists are being clinically developed for metabolic disorders, repurposing them in AD could be a valuable therapeutic strategy. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00842-3.
Collapse
Affiliation(s)
- Marine Tournissac
- Faculté de pharmacie, Université Laval, 1050 Avenue de la Médecine, Quebec, QC, G1V 0A6, Canada.,Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, G1V 4G2, Canada
| | - Tra-My Vu
- Faculté de pharmacie, Université Laval, 1050 Avenue de la Médecine, Quebec, QC, G1V 0A6, Canada.,Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, G1V 4G2, Canada
| | - Nika Vrabic
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, G1V 4G2, Canada
| | - Clara Hozer
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Évolution, 1 Avenue du Petit Château, 91800, Brunoy, France
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, G1V 4G2, Canada
| | - Koralie Mélançon
- Faculté de pharmacie, Université Laval, 1050 Avenue de la Médecine, Quebec, QC, G1V 0A6, Canada.,Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, G1V 4G2, Canada
| | - Emmanuel Planel
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, G1V 4G2, Canada.,Département de psychiatrie et neurosciences, Faculté de médecine, Université Laval, 1050 Avenue de la Médecine, Quebec, QC, G1V 0A6, Canada
| | - Fabien Pifferi
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Évolution, 1 Avenue du Petit Château, 91800, Brunoy, France
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, 1050 Avenue de la Médecine, Quebec, QC, G1V 0A6, Canada. .,Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval (Pavillon CHUL), 2705 Boulevard Laurier, Quebec, QC, G1V 4G2, Canada.
| |
Collapse
|
25
|
Fanet H, Tournissac M, Leclerc M, Caron V, Tremblay C, Vancassel S, Calon F. Tetrahydrobiopterin Improves Recognition Memory in the Triple-Transgenic Mouse Model of Alzheimer's Disease, Without Altering Amyloid-β and Tau Pathologies. J Alzheimers Dis 2021; 79:709-727. [PMID: 33337360 PMCID: PMC7902975 DOI: 10.3233/jad-200637] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a multifactorial disease, implying that multi-target treatments may be necessary to effectively cure AD. Tetrahydrobiopterin (BH4) is an enzymatic cofactor required for the synthesis of monoamines and nitric oxide that also exerts antioxidant and anti-inflammatory effects. Despite its crucial role in the CNS, the potential of BH4 as a treatment in AD has never been scrutinized. OBJECTIVE Here, we investigated whether BH4 peripheral administration improves cognitive symptoms and AD neuropathology in the triple-transgenic mouse model of AD (3xTg-AD), a model of age-related tau and amyloid-β (Aβ) neuropathologies associated with behavior impairment. METHODS Non-transgenic (NonTg) and 3xTg-AD mice were subjected to a control diet (5% fat - CD) or to a high-fat diet (35% fat - HFD) from 6 to 13 months to exacerbate metabolic disorders. Then, mice received either BH4 (15 mg/kg/day, i.p.) or vehicle for ten consecutive days. RESULTS This sub-chronic administration of BH4 rescued memory impairment in 13-month-old 3xTg-AD mice, as determined using the novel object recognition test. Moreover, the HFD-induced glucose intolerance was completely reversed by the BH4 treatment in 3xTg-AD mice. However, the HFD or BH4 treatment had no significant impact on Aβ and tau neuropathologies. CONCLUSION Overall, our data suggest a potential benefit from BH4 administration against AD cognitive and metabolic deficits accentuated by HFD consumption in 3xTg-AD mice, without altering classical neuropathology. Therefore, BH4 should be considered as a candidate for drug repurposing, at least in subtypes of cognitively impaired patients experiencing metabolic disorders.
Collapse
Affiliation(s)
- Hortense Fanet
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- INRA, Nutrition et Neurobiologie Intégrée, UMR, Bordeaux, France
- Université de Bordeaux, Nutrition et Neurobiologie Intégrée, UMR, Bordeaux, France
- International Associated Laboratory OptiNutriBrain, Pavillon des Services, Québec, Canada
| | - Marine Tournissac
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- International Associated Laboratory OptiNutriBrain, Pavillon des Services, Québec, Canada
| | - Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
| | - Vicky Caron
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
| | - Sylvie Vancassel
- INRA, Nutrition et Neurobiologie Intégrée, UMR, Bordeaux, France
- Université de Bordeaux, Nutrition et Neurobiologie Intégrée, UMR, Bordeaux, France
- International Associated Laboratory OptiNutriBrain, Pavillon des Services, Québec, Canada
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- International Associated Laboratory OptiNutriBrain, Pavillon des Services, Québec, Canada
| |
Collapse
|
26
|
Vialard F, Olivier M. Thermoneutrality and Immunity: How Does Cold Stress Affect Disease? Front Immunol 2020; 11:588387. [PMID: 33329571 PMCID: PMC7714907 DOI: 10.3389/fimmu.2020.588387] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
One of the major challenges the scientific community faces today is the lack of translational data generated from mouse trials for human health application. Housing temperature-dependent chronic cold stress in laboratory rodents is one of the key factors contributing to lack of translatability because it reveals major metabolic differences between humans and rodents. While humans tend to operate at temperatures within their thermoneutral zone, most laboratory rodents are housed at temperatures below this zone and have an increased energy demand to generate heat. This has an impact on the immune system of mice and thus affects results obtained using murine models of human diseases. A limited number of studies and reviews have shown that results obtained on mice housed at thermoneutrality were different from those obtained from mice housed in traditional housing conditions. Most of those studies, focused on obesity and cancer, found that housing mice at thermoneutrality changed the outcomes of the diseases negatively and positively, respectively. In this review, we describe how thermoneutrality impacts the immune system of rodents generally and in the context of different disease models. We show that thermoneutrality exacerbates cardiovascular and auto-immune diseases; alleviates asthma and Alzheimer’s disease; and, changes gut microbiome populations. We also show that thermoneutrality can have exacerbating or alleviating effects on the outcome of infectious diseases. Thus, we join the call of others in this field to urge researchers to refine murine models of disease and increase their translational capacity by considering housing at thermoneutrality for trials involving rodents.
Collapse
Affiliation(s)
- Fiorella Vialard
- Department of Microbiology and Immunology, Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
Tsai YJ, Jhong YC, Ching SH, Liao YC, Ching CH, Chuang JI. Cold Exposure After Exercise Impedes the Neuroprotective Effects of Exercise on Thermoregulation and UCP4 Expression in an MPTP-Induced Parkinsonian Mouse Model. Front Neurosci 2020; 14:573509. [PMID: 33041765 PMCID: PMC7522410 DOI: 10.3389/fnins.2020.573509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/17/2020] [Indexed: 11/26/2022] Open
Abstract
Moderate exercise and mild hypothermia have protective effects against brain injury and neurodegeneration. Running in a cold environment alters exercise-induced hyperthermia and outcomes; however, evaluations of post-exercise cold exposure related to exercise benefits for the brain are relatively rare. We investigated the effects of 4°C cold exposure after exercise on exercise-induced thermal responses and neuroprotection in an MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced Parkinsonian mouse model. Male C57BL/6J mice were pretreated with MPTP for five consecutive days and follow-up treadmill exercise for 4 weeks. After 1-h running at a 22°C temperature, the mice were exposed to a 4°C environment for 2 h. An MPTP injection induced a transient drop in body and brain temperatures, while mild brain hypothermia was found to last for 4 weeks after MPTP treatment. Preventing brain hypothermia by exercise or 4°C exposure was associated with an improvement in MPTP-induced striatal uncoupling protein 4 (UCP4) downregulation and nigrostriatal dopaminergic neurodegeneration. However, 4°C exposure after exercise abrogated the exercise-induced beneficial effects and thermal responses in MPTP-treated mice, including a low amplitude of exercise-induced brain hyperthermia and body temperature while at rest after exercise. Our findings elucidate that post-exercise thermoregulation and UCP4 expression are important in the neuroprotective effects of exercise against MPTP toxicity.
Collapse
Affiliation(s)
- Yi-Ju Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yue-Cih Jhong
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Hong Ching
- The Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ching Liao
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Hsin Ching
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jih-Ing Chuang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,The Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
28
|
Pražienková V, Schirmer C, Holubová M, Železná B, Kuneš J, Galas MC, Maletínská L. Lipidized Prolactin-Releasing Peptide Agonist Attenuates Hypothermia-Induced Tau Hyperphosphorylation in Neurons. J Alzheimers Dis 2020; 67:1187-1200. [PMID: 30689580 DOI: 10.3233/jad-180837] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases, characterized by the accumulation of extracellular amyloid plaques and intraneuronal neurofibrillary tangles. These tangles mainly consist of hyperphosphorylated tau protein. As it induces tau hyperphosphorylation in vitro and in vivo, hypothermia is a useful tool for screening potential neuroprotective compounds that ameliorate tau pathology. In this study, we examined the effect of prolactin-releasing peptide (PrRP), its lipidized analog palm11-PrRP31 and glucagon-like-peptide-1 agonist liraglutide, substances with anorexigenic and antidiabetic properties, on tau phosphorylation and on the main kinases and phosphatases involved in AD development. Our study was conducted in a neuroblastoma cell line SH-SY5Y and rat primary neuronal cultures under normothermic and hypothermic conditions. Hypothermia induced a significant increase in tau phosphorylation at the pThr212 and pSer396/pSer404 epitopes. The palmitoylated analogs liraglutide and palm11-PrRP31 attenuated tau hyperphosphorylation, suggesting their potential use in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Claire Schirmer
- Université Lille, INSERM, CHU Lille, UMR - S 1172 - Jean Pierre Aubert Research Centre, Alzheimer and Tauopathies, Lille, France
| | - Martina Holubová
- Institute of Organic Chemistry and Biochemistry, AS CR, Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry, AS CR, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, AS CR, Prague, Czech Republic.,Institute of Physiology, AS CR, Prague, Czech Republic
| | - Marie-Christine Galas
- Université Lille, INSERM, CHU Lille, UMR - S 1172 - Jean Pierre Aubert Research Centre, Alzheimer and Tauopathies, Lille, France
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, AS CR, Prague, Czech Republic
| |
Collapse
|
29
|
Abstract
Tau protein which was discovered in 1975 [310] became of great interest when it was identified as the main component of neurofibrillary tangles (NFT), a pathological feature in the brain of patients with Alzheimer's disease (AD) [39, 110, 232]. Tau protein is expressed mainly in the brain as six isoforms generated by alternative splicing [46, 97]. Tau is a microtubule associated proteins (MAPs) and plays a role in microtubules assembly and stability, as well as diverse cellular processes such as cell morphogenesis, cell division, and intracellular trafficking [49]. Additionally, Tau is involved in much larger neuronal functions particularly at the level of synapses and nuclei [11, 133, 280]. Tau is also physiologically released by neurons [233] even if the natural function of extracellular Tau remains to be uncovered (see other chapters of the present book).
Collapse
|
30
|
Hylander BL, Gordon CJ, Repasky EA. Manipulation of Ambient Housing Temperature To Study the Impact of Chronic Stress on Immunity and Cancer in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 202:631-636. [PMID: 30670578 DOI: 10.4049/jimmunol.1800621] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022]
Abstract
Mice are the preeminent research organism in which to model human diseases and study the involvement of the immune response. Rapidly accumulating evidence indicates a significant involvement of stress hormones in cancer progression, resistance to therapies, and suppression of immune responses. As a result, there has been a concerted effort to model human stress in mice. In this article, we discuss recent literature showing how mice in research facilities are chronically stressed at baseline because of environmental factors. Focusing on housing temperature, we suggest that the stress of cool housing temperatures contributes to the impact of other imposed experimental stressors and therefore has a confounding effect on mouse stress models. Furthermore, we propose that manipulation of housing temperature is a useful approach for studying the impact of chronic stress on disease and the immune response and for testing therapeutic methods of reducing the negative effects of chronic stress.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - Christopher J Gordon
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| |
Collapse
|
31
|
Wei Y, Wang Y, Lin CK, Yin K, Yang J, Shi L, Li L, Zanobetti A, Schwartz JD. Associations between seasonal temperature and dementia-associated hospitalizations in New England. ENVIRONMENT INTERNATIONAL 2019; 126:228-233. [PMID: 30822651 PMCID: PMC8491247 DOI: 10.1016/j.envint.2018.12.054] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/17/2018] [Accepted: 12/24/2018] [Indexed: 05/22/2023]
Abstract
Human-induced climate change has accelerated in recent decades, causing adverse health effects. However, the impact of the changing climate on neurological disorders in the older population is not well understood. We applied time-varying Cox proportional hazards models to estimate the associations between hospital admissions for dementia and the mean and variability of summer and winter temperatures in New England. We estimated seasonal temperatures for each New England zip code using a satellite-based prediction model. By characterizing spatial differences and temporal fluctuations in seasonal temperatures, we observed a lower risk of dementia-associated hospital admissions in years when local temperatures in either summer (hazard ration [HR] = 0.98; 95% confidence interval [CI]: 0.96, 1.00) or winter (HR = 0.97; 95% CI: 0.94, 0.99) were higher than average, and a greater risk of dementia-associated admissions for older adults living in zip codes with higher temperature variations. Effect modifications by sex, race, age, and dual eligibility were considered to examine vulnerability of population subgroups. Our results suggest that cooler-than-average temperatures and higher temperature variability increase the risk of dementia-associated hospital admissions. Thus, climate change may affect progression of dementia and associated hospitalization costs.
Collapse
Affiliation(s)
- Yaguang Wei
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, United States of America.
| | - Yan Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, United States of America; Department of Biostatistics, Harvard T.H. Chan School of Public Health, United States of America
| | - Cheng-Kuan Lin
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, United States of America
| | - Kanhua Yin
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, United States of America
| | - Jiabei Yang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, United States of America
| | - Liuhua Shi
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, United States of America
| | - Longxiang Li
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, United States of America
| | - Antonella Zanobetti
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, United States of America
| | - Joel D Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, United States of America
| |
Collapse
|
32
|
Perpetuini D, Cardone D, Chiarelli AM, Filippini C, Croce P, Zappasodi F, Rotunno L, Anzoletti N, Zito M, Merla A. Autonomic impairment in Alzheimer's disease is revealed by complexity analysis of functional thermal imaging signals during cognitive tasks. Physiol Meas 2019; 40:034002. [PMID: 30736015 DOI: 10.1088/1361-6579/ab057d] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Alzheimer's disease (AD) is characterized by progressive memory failures and visuospatial impairment. Moreover, AD can be accompanied by autonomic system alterations, which, among other impacts, affect thermoregulatory activity. We here investigate differences in autonomic activity between AD patients and healthy controls (HC), employing a complexity analysis of functional infrared imaging (fIRI) data acquired at rest and during the execution of clinical cognitive and mnemonic tests. APPROACH fIRI allows for contactless monitoring of autonomic activity and its thermoregulatory expression without interfering with the psychophysiological state of the subject, preserving free interaction with the doctor. The signal complexity analysis, based on the sample entropy, was compared to a standard frequency-based analysis of autonomic-related signals. MAIN RESULTS AD patients exhibited lower complexity of fIRI signals during the tests, which could be indicative of a stronger sympathetic activity with respect to HC. No significant effects were found at rest. No differences were found on employing frequency-based analysis. SIGNIFICANCE This study confirms that AD patients may exhibit peculiar autonomic responses associated with the execution of cognitive tasks that can be measured through fIRI. Moreover, these responses could be highlighted by a nonlinear metric of signal predictability such as the sample entropy establishing autonomic impairment of AD patients.
Collapse
Affiliation(s)
- David Perpetuini
- Infrared Imaging Lab, Centro ITAB-Institute for Advanced Biomedical Technologies, University G. d'Annunzio, Chieti, 66100, Italy. Department of Neurosciences, Imaging and Clinical Sciences, University G. d'Annunzio, Chieti-Pescara, 66100, Italy. Author to whom any correspondence should be addressed
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Repeated cold exposures protect a mouse model of Alzheimer's disease against cold-induced tau phosphorylation. Mol Metab 2019; 22:110-120. [PMID: 30770297 PMCID: PMC6437631 DOI: 10.1016/j.molmet.2019.01.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 02/06/2023] Open
Abstract
Objective Old age is associated with a rise in the incidence of Alzheimer's disease (AD) but also with thermoregulatory deficits. Indicative of a link between the two, hypothermia induces tau hyperphosphorylation. The 3xTg-AD mouse model not only develops tau and amyloid pathologies in the brain but also metabolic and thermoregulatory deficits. Brown adipose tissue (BAT) is the main thermogenic driver in mammals, and its stimulation counteracts metabolic deficits in rodents and humans. We thus investigated whether BAT stimulation impedes AD neuropathology. Methods 15-month-old 3xTg-AD mice were subjected to repeated short cold exposures (RSCE), consisting of 4-hour sessions of cold exposure (4 °C), five times per week for four weeks, compared to animals kept at housing temperature. Results First, we confirmed that 3xTg-AD RSCE-trained mice exhibited BAT thermogenesis and improved glucose tolerance. RSCE-trained mice were completely resistant to tau hyperphosphorylation in the hippocampus induced by a 24-hour cold challenge. Finally, RSCE increased plasma levels of fibroblast growth factor 21 (FGF21), a batokine, which inversely correlated with hippocampal tau phosphorylation. Conclusions Overall, BAT stimulation through RSCE improved metabolic deficits and completely blocked cold-induced tau hyperphosphorylation in the 3xTg-AD mouse model of AD neuropathology. These results suggest that improving thermogenesis could exert a therapeutic effect in AD. Cold acclimation increases brown adipose tissue thermogenesis in old 3xTg-AD mice. Cold acclimation improved glucose tolerance in old 3xTg-AD mice. Enhanced thermogenesis protects against cold-induced brain tau phosphorylation. Repeated cold exposures increased plasmatic levels of fibroblast growth factor 21. Peripheral fibroblast growth factor 21 levels correlate with tau phosphorylation.
Collapse
|
34
|
Motzko-Soares ACP, Vizin RCL, Martins TMS, Hungaro ARO, Sato JR, Almeida MC, Carrettiero DC. Thermoregulatory profile of neurodegeneration-induced dementia of the Alzheimer's type using intracerebroventricular streptozotocin in rats. Acta Physiol (Oxf) 2018; 224:e13084. [PMID: 29719119 DOI: 10.1111/apha.13084] [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] [Received: 01/23/2018] [Revised: 04/21/2018] [Accepted: 04/24/2018] [Indexed: 01/18/2023]
Abstract
AIM Here, we have extensively investigated the relationship between thermoregulation and neurodegeneration-induced dementia of the Alzheimer's type using intracerebroventricular injections of streptozotocin (icv-STZ). METHODS Male Wistar rats were treated with bilateral injections of icv-STZ, and their thermoregulatory profiles (core body temperature, tail-skin temperature, cold and heat defence responses and thermal place preference) were evaluated. Spatial memory, locomotor activity, social interaction, brain ventricular volume, and Aβ1-42 and tau protein levels in the brain were analysed to characterize the effects of STZ on the brain and behaviour. RESULTS In addition to deficits in spatial memory, reduced social interaction and an increased brain ventricular volume, icv-STZ rats presented a pattern of hyperthermia, as demonstrated by an increased core body temperature. Hyperthermia was due to the activation of both autonomic heat conservation and behavioural cold avoidance, as STZ-treated rats presented tail-cutaneous vasoconstriction and an altered thermal preference. They also showed a distinct cold defence response when exposed to cold. CONCLUSION Our data bring evidence that icv-STZ in rats causes hyperthermia, with activation of both autonomic and behavioural thermoregulatory defence responses when challenged at colder temperatures, leading us to hypothesize that they are more efficient in preventing hypothermia. These data are relevant for a better understanding of neurodegenerative disease mechanisms.
Collapse
Affiliation(s)
- A. C. P. Motzko-Soares
- Graduate Program in Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - R. C. L. Vizin
- Graduate Program in Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - T. M. S. Martins
- Undergraduate Program in Science and Technology; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - A. R. O. Hungaro
- Undergraduate Program in Science and Technology; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - J. R. Sato
- Graduate Program in Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
- Center for Mathematics Computation and Cognition (CMCC); Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - M. C. Almeida
- Graduate Program in Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
- Center for Natural and Human Sciences (CCNH); Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - D. C. Carrettiero
- Graduate Program in Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
- Center for Natural and Human Sciences (CCNH); Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| |
Collapse
|
35
|
|
36
|
Dal-Pan A, Dudonné S, Bourassa P, Bourdoulous M, Tremblay C, Desjardins Y, Calon F. Cognitive-Enhancing Effects of a Polyphenols-Rich Extract from Fruits without Changes in Neuropathology in an Animal Model of Alzheimer's Disease. J Alzheimers Dis 2018; 55:115-135. [PMID: 27662290 DOI: 10.3233/jad-160281] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
No effective preventive treatment is available for age-related cognitive decline and Alzheimer's disease (AD). Epidemiological studies indicate that a diet rich in fruit is associated with cognitive improvement. It was thus proposed that high polyphenol concentrations found in berries can prevent cognitive impairment associated with aging and AD. Therefore, the Neurophenols project aimed at investigating the effects of a polyphenolic extract from blueberries and grapes (PEBG) in the triple-transgenic (3xTg-AD) mouse model of AD, which develops AD neuropathological markers, including amyloid-β plaques and neurofibrillary tangles, leading to memory deficits. In this study, 12-month-old 3xTg-AD and NonTg mice were fed a diet supplemented with standardized PEBG (500 or 2500 mg/kg) for 4 months (n = 15-20/group). A cognitive evaluation with the novel object recognition test was performed at 15 months of age and mice were sacrificed at 16 months of age. We observed that PEBG supplementation with doses of 500 or 2500 mg/kg prevented the decrease in novel object recognition observed in both 15-month-old 3xTg-AD mice and NonTg mice fed a control diet. Although PEBG treatment did not reduce Aβ and tau pathologies, it prevented the decrease in mature BDNF observed in 16-month-old 3xTg-AD mice. Finally, plasma concentrations of phenolic metabolites, such as dihydroxyphenyl valerolactone, a microbial metabolite of epicatechin, positively correlated with memory performances in supplemented mice. The improvement in object recognition observed in 3xTg-AD mice after PEBG administration supports the consumption of polyphenols-rich extracts to prevent memory impairment associated with age-related disease, without significant effects on classical AD neuropathology.
Collapse
Affiliation(s)
- Alexandre Dal-Pan
- CHU de Québec Research Center, Quebec, QC, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec, QC, Canada.,OptiNutriBrain International Associated Laboratory (NutriNeuro France-INAF Canada).,http://www.neurophenols.org
| | - Stéphanie Dudonné
- Institute of Nutrition and Functional Foods, Laval University, Quebec, QC, Canada.,http://www.neurophenols.org
| | - Philippe Bourassa
- CHU de Québec Research Center, Quebec, QC, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec, QC, Canada.,Faculty of Pharmacy, Laval University, Quebec, QC, Canada.,OptiNutriBrain International Associated Laboratory (NutriNeuro France-INAF Canada)
| | | | | | - Yves Desjardins
- Institute of Nutrition and Functional Foods, Laval University, Quebec, QC, Canada.,http://www.neurophenols.org
| | - Frédéric Calon
- CHU de Québec Research Center, Quebec, QC, Canada.,Institute of Nutrition and Functional Foods, Laval University, Quebec, QC, Canada.,Faculty of Pharmacy, Laval University, Quebec, QC, Canada.,OptiNutriBrain International Associated Laboratory (NutriNeuro France-INAF Canada)
| | | |
Collapse
|
37
|
Qiao G, Chen M, Bucsek MJ, Repasky EA, Hylander BL. Adrenergic Signaling: A Targetable Checkpoint Limiting Development of the Antitumor Immune Response. Front Immunol 2018; 9:164. [PMID: 29479349 PMCID: PMC5812031 DOI: 10.3389/fimmu.2018.00164] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/18/2018] [Indexed: 12/15/2022] Open
Abstract
An immune response must be tightly controlled so that it will be commensurate with the level of response needed to protect the organism without damaging normal tissue. The roles of cytokines and chemokines in orchestrating these processes are well known, but although stress has long been thought to also affect immune responses, the underlying mechanisms were not as well understood. Recently, the role of nerves and, specifically, the sympathetic nervous system, in regulating immune responses is being revealed. Generally, an acute stress response is beneficial but chronic stress is detrimental because it suppresses the activities of effector immune cells while increasing the activities of immunosuppressive cells. In this review, we first discuss the underlying biology of adrenergic signaling in cells of both the innate and adaptive immune system. We then focus on the effects of chronic adrenergic stress in promoting tumor growth, giving examples of effects on tumor cells and immune cells, explaining the methods commonly used to induce stress in preclinical mouse models. We highlight how this relates to our observations that mandated housing conditions impose baseline chronic stress on mouse models, which is sufficient to cause chronic immunosuppression. This problem is not commonly recognized, but it has been shown to impact conclusions of several studies of mouse physiology and mouse models of disease. Moreover, the fact that preclinical mouse models are chronically immunosuppressed has critical ramifications for analysis of any experiments with an immune component. Our group has found that reducing adrenergic stress by housing mice at thermoneutrality or treating mice housed at cooler temperatures with β-blockers reverses immunosuppression and significantly improves responses to checkpoint inhibitor immunotherapy. These observations are clinically relevant because there are numerous retrospective epidemiological studies concluding that cancer patients who were taking β-blockers have better outcomes. Clinical trials testing whether β-blockers can be repurposed to improve the efficacy of traditional and immunotherapies in patients are on the horizon.
Collapse
Affiliation(s)
- Guanxi Qiao
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Minhui Chen
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Mark J. Bucsek
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Elizabeth A. Repasky
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Bonnie L. Hylander
- Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
38
|
The contribution of transgenic and nontransgenic animal models in Alzheimer's disease drug research and development. Behav Pharmacol 2018; 28:95-111. [PMID: 28177983 DOI: 10.1097/fbp.0000000000000296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Over the last few years, several papers have become available in the literature on both the main hallmarks of Alzheimer's disease (AD) and the several intracellular pathways whose alteration is responsible for its onset and progression. The use of transgenic and nontransgenic animal models has played a key role in achieving such a remarkable amount of preclinical data, allowing researchers to dissect the cellular changes occurring in the AD brain. In addition, the huge amount of preclinical evidence arising from these animal models was necessary for the further clinical development of pharmacological agents capable of interfering with most of the impaired neural pathways in AD patients. In this respect, a significant role is played by the dysfunction of excitatory and inhibitory neurotransmission responsible for the cognitive and behavioral symptoms described in AD patients. The aim of this review is to summarize the main animal models that contributed toward unraveling the pathological changes in neurotransmitter synthesis, release, and receptor binding in AD preclinical studies. The review also provides an updated description of the current pharmacological agents - still under clinical development - acting on the neurotransmitter systems.
Collapse
|
39
|
Almeida MC, Carrettiero DC. Hypothermia as a risk factor for Alzheimer disease. HANDBOOK OF CLINICAL NEUROLOGY 2018; 157:727-735. [PMID: 30459036 DOI: 10.1016/b978-0-444-64074-1.00044-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Alzheimer disease (AD), which is associated with chronic and progressive neurodegeneration, is the most prevalent cause of dementia linked to aging. Among the risk factors for AD, age stands as the greatest one, with the vast majority of people with AD being 65 years of age or older. Nevertheless, the pathophysiologic mechanisms underlying the link between aging and the development of AD, although not completely understood, might reveal important aspects for the understanding of this pathology. Thus, there is significant evidence that the impaired thermal homeostasis associated with normal aging leads to a variety of metabolic changes that could be associated with AD development. In this chapter, we assess the clinical and biochemical evidence implicating hypothermia as a risk factor for the development of AD and the impact of hypothermia on the two pathologic hallmarks of AD: accumulation of senile plaques of amyloid-beta and neurofibrillary tangles of aberrant hyperphosphorylated tau protein.
Collapse
Affiliation(s)
- Maria Camila Almeida
- Natural and Human Sciences Center, Federal University of ABC, São Bernardo do Campo, SP, Brazil.
| | | |
Collapse
|
40
|
Gratuze M, Joly-Amado A, Vieau D, Buée L, Blum D. Mutual Relationship between Tau and Central Insulin Signalling: Consequences for AD and Tauopathies? Neuroendocrinology 2018; 107:181-195. [PMID: 29439247 DOI: 10.1159/000487641] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/13/2018] [Indexed: 12/30/2022]
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disorder mainly characterized by cognitive deficits and neuropathological changes such as Tau lesions and amyloid plaques, but also associated with non-cognitive symptomatology. Metabolic and neuroendocrine abnormalities, such as alterations in body weight, brain insulin impairments, and lower brain glucose metabolism, which often precede clinical diagnosis, have been extensively reported in AD patients. However, the origin of these symptoms and their relation to pathology and cognitive impairments remain misunderstood. Insulin is a hormone involved in the control of energy homeostasis both peripherally and centrally, and insulin-resistant state has been linked to increased risk of dementia. It is now well established that insulin resistance can exacerbate Tau lesions, mainly by disrupting the balance between Tau kinases and phosphatases. On the other hand, the emerging literature indicates that Tau protein can also modulate insulin signalling in the brain, thus creating a detrimental vicious circle. The following review will highlight our current understanding of the role of insulin in the brain and its relation to Tau protein in the context of AD and tauopathies. Considering that insulin signalling is prone to be pharmacologically targeted at multiple levels, it constitutes an appealing approach to improve both insulin brain sensitivity and mitigate brain pathology with expected positive outcome in terms of cognition.
Collapse
Affiliation(s)
- Maud Gratuze
- Centre de Recherche du Centre Hospitalier de l'Université Laval de Québec, Axe Neurosciences, Université Laval, Québec, Québec, Canada
| | - Aurélie Joly-Amado
- Byrd Alzheimer's Institute, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Didier Vieau
- Université de Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, "Alzheimer and Tauopathies,", Lille, France
| | - Luc Buée
- Université de Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, "Alzheimer and Tauopathies,", Lille, France
| | - David Blum
- Université de Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, "Alzheimer and Tauopathies,", Lille, France
| |
Collapse
|
41
|
Fischer AW, Cannon B, Nedergaard J. Optimal housing temperatures for mice to mimic the thermal environment of humans: An experimental study. Mol Metab 2017; 7:161-170. [PMID: 29122558 PMCID: PMC5784327 DOI: 10.1016/j.molmet.2017.10.009] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 10/12/2017] [Accepted: 10/21/2017] [Indexed: 11/05/2022] Open
Abstract
Objectives The laboratory mouse is presently the most common model for examining mechanisms of human physiology and disease. Housing temperatures can have a large impact on the outcome of such experiments and on their translatability to the human situation. Humans usually create for themselves a thermoneutral environment without cold stress, while laboratory mice under standard conditions (≈20° C) are under constant cold stress. In a well-cited, theoretical paper by Speakman and Keijer in Molecular Metabolism, it was argued that housing mice under close to standard conditions is the optimal way of modeling the human metabolic situation. This tenet was mainly based on the observation that humans usually display average metabolic rates of about 1.6 times basal metabolic rate. The extra heat thereby produced would also be expected to lead to a shift in the ‘lower critical temperature’ towards lower temperatures. Methods To examine these tenets experimentally, we performed high time-resolution indirect calorimetry at different environmental temperatures on mice acclimated to different housing temperatures. Results Based on the high time-resolution calorimetry analysis, we found that mice already under thermoneutral conditions display mean diurnal energy expenditure rates 1.8 times higher than basal metabolism, remarkably closely resembling the human situation. At any temperature below thermoneutrality, mice metabolism therefore exceeds the human equivalent: Mice under standard conditions display energy expenditure 3.1 times basal metabolism. The discrepancy to previous conclusions is probably attributable to earlier limitations in establishing true mouse basal metabolic rate, due to low time resolution. We also found that the fact that mean energy expenditure exceeds resting metabolic rate does not move the apparent thermoneutral zone (the lower critical temperature) downwards. Conclusions We show that housing mice at thermoneutrality is an advantageous step towards aligning mouse energy metabolism to human energy metabolism. High time-resolution indirect calorimetry reveals true resting metabolic rate in mice. Mice at thermoneutrality display average energy expenditure 1.8 times their RMR (BMR). This relationship very closely resembles the human situation. Lower critical temperature is not influenced by enhanced energy expenditure. To model human metabolism, 30 °C remains the optimal housing temperature for mice.
Collapse
Affiliation(s)
- Alexander W Fischer
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91 Stockholm, Sweden; Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, DE-20246 Hamburg, Germany
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91 Stockholm, Sweden.
| |
Collapse
|
42
|
Chouinard-Watkins R, Vandal M, Léveillé P, Pinçon A, Calon F, Plourde M. Docosahexaenoic acid prevents cognitive deficits in human apolipoprotein E epsilon 4-targeted replacement mice. Neurobiol Aging 2017; 57:28-35. [PMID: 28595105 DOI: 10.1016/j.neurobiolaging.2017.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 04/25/2017] [Accepted: 05/02/2017] [Indexed: 11/26/2022]
Abstract
At a population level, dietary consumption of fish rich in docosahexaenoic acid (DHA) is associated with prevention of cognitive decline but this association is not clear in carriers of the apolipoprotein E epsilon 4 allele (E4). Plasma and liver DHA concentrations show significant alterations in E4 carriers, in part corrected by DHA supplementation. However, whether DHA sufficiency in E4 carriers has consequences on cognition is unknown. Mice expressing human E4 or apolipoprotein E epsilon 3 allele (E3) were fed either a control diet or a diet containing DHA for 8 months and cognitive performance was tested using the object recognition test and the Barnes maze test. In E4 mice fed the control diet, impaired memory was detected and arachidonic acid concentrations were elevated in the hippocampus compared to E3 mice fed the control diet. DHA consumption prevented memory decline and restored arachidonic acid concentrations in the hippocampus of E4 mice. Our results suggest that long-term high-dose DHA intake may prevent cognitive decline in E4 carriers.
Collapse
Affiliation(s)
- Raphaël Chouinard-Watkins
- Research Center on Aging, Centre intégré universitaire de santé et de services sociaux de l'estrie-Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Canada; Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Canada; Department of Physiology, Université de Sherbrooke, Sherbrooke, Canada; Institute of Nutrition and Functional Foods, Quebec City, Canada
| | - Milène Vandal
- Institute of Nutrition and Functional Foods, Quebec City, Canada; Faculté de Pharmacie, Université Laval, Quebec City, Canada; Neurosciences Axis, Centre hospitalier universitaire de Québec Research Center, Quebec City, Canada
| | - Pauline Léveillé
- Research Center on Aging, Centre intégré universitaire de santé et de services sociaux de l'estrie-Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Canada; Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Canada; Institute of Nutrition and Functional Foods, Quebec City, Canada
| | - Anthony Pinçon
- Research Center on Aging, Centre intégré universitaire de santé et de services sociaux de l'estrie-Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Canada; Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Canada; Institute of Nutrition and Functional Foods, Quebec City, Canada
| | - Frédéric Calon
- Institute of Nutrition and Functional Foods, Quebec City, Canada; Faculté de Pharmacie, Université Laval, Quebec City, Canada; Neurosciences Axis, Centre hospitalier universitaire de Québec Research Center, Quebec City, Canada
| | - Mélanie Plourde
- Research Center on Aging, Centre intégré universitaire de santé et de services sociaux de l'estrie-Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Canada; Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Canada; Institute of Nutrition and Functional Foods, Quebec City, Canada; Department of Medicine, Université de Sherbrooke, Sherbrooke, Canada.
| |
Collapse
|
43
|
Sparacia G, Sakai K, Yamada K, Giordano G, Coppola R, Midiri M, Grimaldi LM. Assessment of brain core temperature using MR DWI-thermometry in Alzheimer disease patients compared to healthy subjects. Jpn J Radiol 2017; 35:168-171. [DOI: 10.1007/s11604-017-0616-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/23/2017] [Indexed: 01/24/2023]
|
44
|
Jan A, Jansonius B, Delaidelli A, Somasekharan SP, Bhanshali F, Vandal M, Negri GL, Moerman D, MacKenzie I, Calon F, Hayden MR, Taubert S, Sorensen PH. eEF2K inhibition blocks Aβ42 neurotoxicity by promoting an NRF2 antioxidant response. Acta Neuropathol 2017; 133:101-119. [PMID: 27752775 DOI: 10.1007/s00401-016-1634-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/11/2016] [Accepted: 10/11/2016] [Indexed: 01/01/2023]
Abstract
Soluble oligomers of amyloid-β (Aβ) impair synaptic plasticity, perturb neuronal energy homeostasis, and are implicated in Alzheimer's disease (AD) pathogenesis. Therefore, significant efforts in AD drug discovery research aim to prevent the formation of Aβ oligomers or block their neurotoxicity. The eukaryotic elongation factor-2 kinase (eEF2K) plays a critical role in synaptic plasticity, and couples neurotransmission to local dendritic mRNA translation. Recent evidence indicates that Aβ oligomers activate neuronal eEF2K, suggesting a potential link to Aβ induced synaptic dysfunction. However, a detailed understanding of the role of eEF2K in AD pathogenesis, and therapeutic potential of eEF2K inhibition in AD, remain to be determined. Here, we show that eEF2K activity is increased in postmortem AD patient cortex and hippocampus, and in the hippocampus of aged transgenic AD mice. Furthermore, eEF2K inhibition using pharmacological or genetic approaches prevented the toxic effects of Aβ42 oligomers on neuronal viability and dendrite formation in vitro. We also report that eEF2K inhibition promotes the nuclear factor erythroid 2-related factor (NRF2) antioxidant response in neuronal cells, which was crucial for the beneficial effects of eEF2K inhibition in neurons exposed to Aβ42 oligomers. Accordingly, NRF2 knockdown or overexpression of the NRF2 inhibitor, Kelch-Like ECH-Associated Protein-1 (Keap1), significantly attenuated the neuroprotection associated with eEF2K inhibition. Finally, genetic deletion of the eEF2K ortholog efk-1 reduced oxidative stress, and improved chemotaxis and serotonin sensitivity in C. elegans expressing human Aβ42 in neurons. Taken together, these findings highlight the potential utility of eEF2K inhibition to reduce Aβ-mediated oxidative stress in AD.
Collapse
Affiliation(s)
- Asad Jan
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- British Columbia Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Brandon Jansonius
- British Columbia Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Alberto Delaidelli
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- British Columbia Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | | | - Forum Bhanshali
- Department of Medical Genetics, Child and Family Research Institute, University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - Milène Vandal
- Faculté de Pharmacie, Université Laval, Pavillon Ferdinand-Vandry 1050, Avenue de la Médecine, Quebec, QC, G1V 0A6, Canada
| | - Gian Luca Negri
- British Columbia Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Don Moerman
- Department of Zoology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Ian MacKenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Pavillon Ferdinand-Vandry 1050, Avenue de la Médecine, Quebec, QC, G1V 0A6, Canada
| | - Michael R Hayden
- Department of Medical Genetics, Child and Family Research Institute, University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - Stefan Taubert
- Department of Medical Genetics, Child and Family Research Institute, University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada
| | - Poul H Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.
- British Columbia Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| |
Collapse
|
45
|
Tournissac M, Vandal M, François A, Planel E, Calon F. Old age potentiates cold-induced tau phosphorylation: linking thermoregulatory deficit with Alzheimer's disease. Neurobiol Aging 2016; 50:25-29. [PMID: 27838492 DOI: 10.1016/j.neurobiolaging.2016.09.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 09/09/2016] [Accepted: 09/29/2016] [Indexed: 02/08/2023]
Abstract
Thermoregulatory deficits coincide with a rise in the incidence of Alzheimer's disease (AD) in old age. Lower body temperature increases tau phosphorylation, a neuropathological hallmark of AD. To determine whether old age potentiates cold-induced tau phosphorylation, we compared the effects of cold exposure (4 °C, 24 hours) in 6- and 18-month-old mice. Cold-induced changes in body temperature, brown adipose tissue activity, and phosphorylation of tau at Ser202 were not different between 6- and 18-month-old mice. However, following cold exposure, only old mice displayed a significant rise in soluble tau pThr181 and pThr231, which was correlated with body temperature. Inactivation of glycogen synthase kinase 3β was more prominent in young mice, suggesting a protective mechanism against cold-induced tau phosphorylation. These results suggest that old age confers higher susceptibility to tau hyperphosphorylation following a change in body temperature, thereby contributing to an enhanced risk of developing AD.
Collapse
Affiliation(s)
- Marine Tournissac
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Québec, Canada
| | - Milène Vandal
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Québec, Canada
| | - Arnaud François
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Québec, Canada
| | - Emmanuel Planel
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Québec, Canada; Faculté de médecine, Département de psychiatrie et de neurosciences, Université Laval, Québec, Québec, Canada
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Québec, Canada.
| |
Collapse
|