1
|
Ma J, Wu C, Zhang Z, Liu H, Zong K, Wang Y, Lin R, Li R, Zou C, Zuo Q, Xu Y, Liu J, Zhao R. Metabolic pathway and genetically causal links of 1,400 circulating metabolites on the risk of intracranial aneurysms and aneurysmal subarachnoid hemorrhage. Neuroscience 2025; 568:27-37. [PMID: 39800046 DOI: 10.1016/j.neuroscience.2025.01.017] [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: 09/29/2024] [Revised: 12/29/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND The rupture of intracranial aneurysms (IAs) leads to aneurysmal subarachnoid hemorrhage (aSAH), which is associated with significant disability and mortality rates. This study aims to identify metabolic markers causally linked to the occurrence of IAs and aSAH through Mendelian randomization (MR), thereby offering novel predictive and therapeutic targets. METHODS We conducted a genome-wide association study (GWAS) on IAs and aSAH, analyzing 1,400 metabolomic indices from the Canadian Longitudinal Study on Aging (CLSA) cohort (n = 8,299). Subsequently, we employed two-sample Mendelian randomization to ascertain potential causal relationships between each metabolite and the conditions IAs and aSAH by various MR methodologies, including MR Egger, Weighted median, Inverse variance weighted (IVW), MR-PRESSO, Simple mode, and Weighted mode. The heterogeneity of instrumental variables was assessed using Cochran's Q statistics, and metabolic pathway analyses were performed via the Metaconflict 5.0 platform. RESULTS Our analysis found that 87 metabolites/metabolic ratios were associated with IAs, and 85 metabolites/metabolic ratios were associated with aSAH. After false discovery rate (FDR) correction and sensitivity analyses, nine metabolites/metabolic ratios were significantly causally associated with aSAH. Conversely, while 87 metabolites and their ratios initially showed potential causal links with IA, none demonstrated significant causal associations post-FDR correction. The study also pinpointed eight significant metabolic pathways implicated in both IAs and aSAH. CONCLUSION This study found that nine circulating metabolites and their ratios with significant causal associations to aSAH, while no metabolites and their ratios were causally linked to IAs. These results suggest possible mechanisms and predictive molecular targets for IAs and aSAH.
Collapse
Affiliation(s)
- Junren Ma
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Congyan Wu
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Zhentao Zhang
- Department of Emergency, Changhai hospital, Naval Medical University, Shanghai, China
| | - Hanchen Liu
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Kang Zong
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Yonghui Wang
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Ruyue Lin
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Rui Li
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Chao Zou
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Qiao Zuo
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Yi Xu
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China
| | - Jianmin Liu
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China.
| | - Rui Zhao
- Neurovascular Center, Changhai hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
2
|
Mu Z, Sun Z, Wu S, Yang J, Wang P, Zhao X. Trehalose Inhibits ferroptosis Through Activating SIRT3/SOD2 Signaling Axis and Alleviates Brain Injury After Traumatic Brain Injury. Neurochem Res 2025; 50:78. [PMID: 39798057 DOI: 10.1007/s11064-024-04330-6] [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: 10/15/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
Trehalose has neuroprotective effects in neurodegenerative diseases. This study aimed to explore the impact of trehalose on traumatic brain injury (TBI) by investigating its role in neuroprotection. The TBI mice model was established utilizing the cortical impact technique followed by trehalose treatment. Traumatic neuronal injury induced by scratch followed by trehalose treatment was performed to mimic TBI in vitro. Memory function was assessed using the Water maze test. Brain damage was evaluated through various methods including brain water content analysis, Nissl staining, Evans blue exudation, and TUNEL staining. Biochemical and morphological changes related to ferroptosis post-TBI were also examined. The results showed that trehalose was found to enhance spatial memory, reduce brain injury, and inhibit ferroptosis in TBI mice, similar to ferroptosis inhibitors. The influence of trehalose on TBI was reversed by the SIRT3 inhibitor. Trehalose upregulated SIRT3 to increase SOD activity in TBI, which could also be counteracted by the SIRT3 inhibitor. Combining trehalose with a ferroptosis inhibitor had a more significant effect on reducing brain injury and inhibiting ferroptosis. Furthermore, in TBI mice treated with trehalose and SIRT3 inhibitors, the effect of trehalose was reversed by SIRT3 inhibitors, but the addition of ferroptosis inhibitors reversed the effect of SIRT3 inhibitors, as shown by decreased ferroptosis and neuronal apoptosis in damaged brain tissue. In summary, this study provides initial evidence that trehalose plays a crucial role in neuroprotection post-TBI through the SIRT3/SOD2 pathway-mediated ferroptosis.
Collapse
Affiliation(s)
- Zhenqian Mu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, China
| | - Zhenlie Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, China
| | - Shuai Wu
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong, 226019, China
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, 214002, China
| | - Jieqiong Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, China
| | - Peng Wang
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong, 226019, China
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, 214002, China
| | - Xudong Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong, 226019, China.
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, 214002, China.
- Wuxi Neurosurgical Institute, Wuxi, 214002, China.
- , 68 Zhongshan Road, Wuxi, 214000, China.
| |
Collapse
|
3
|
Licitra R, Della Vecchia S, Santucci L, Vivarelli R, Bernardi S, Santorelli FM, Marchese M. Trehalose Ameliorates Zebrafish Emotional and Social Deficits Caused by CLN8 Dysfunction. Cells 2025; 14:55. [PMID: 39791756 PMCID: PMC11720655 DOI: 10.3390/cells14010055] [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: 11/07/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
CLN8 and other neuronal ceroid lipofuscinoses (NCLs) often lead to cognitive decline, emotional disturbances, and social deficits, worsening with disease progression. Disrupted lysosomal pH, impaired autophagy, and defective dendritic arborization contribute to these symptoms. Using a cln8-/- zebrafish model, we identified significant impairments in locomotion, anxiety, and aggression, along with subtle deficits in social interactions, positioning zebrafish as a useful model for therapeutic studies in NCL. Our findings show that trehalose, an autophagy enhancer, ameliorates anxiety, and modestly improves social behavior and predator avoidance in mutant zebrafish. This finding aligns animal models with clinical reports suggestive of behavioral improvements in NCL patients. Trehalose holds promise as a therapeutic agent for CLN8, warranting further research into its neuroprotective mechanisms and clinical applications.
Collapse
Affiliation(s)
- Rosario Licitra
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Calambrone, Italy; (R.L.); (S.D.V.); (L.S.); (R.V.); (S.B.); (F.M.S.)
- Department of Veterinary Sciences, University of Pisa, 56124 Pisa, Italy
| | - Stefania Della Vecchia
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Calambrone, Italy; (R.L.); (S.D.V.); (L.S.); (R.V.); (S.B.); (F.M.S.)
| | - Lorenzo Santucci
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Calambrone, Italy; (R.L.); (S.D.V.); (L.S.); (R.V.); (S.B.); (F.M.S.)
| | - Rachele Vivarelli
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Calambrone, Italy; (R.L.); (S.D.V.); (L.S.); (R.V.); (S.B.); (F.M.S.)
| | - Sara Bernardi
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Calambrone, Italy; (R.L.); (S.D.V.); (L.S.); (R.V.); (S.B.); (F.M.S.)
| | - Filippo M. Santorelli
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Calambrone, Italy; (R.L.); (S.D.V.); (L.S.); (R.V.); (S.B.); (F.M.S.)
| | - Maria Marchese
- Neurobiology and Molecular Medicine Unit, IRCCS Fondazione Stella Maris, 56128 Calambrone, Italy; (R.L.); (S.D.V.); (L.S.); (R.V.); (S.B.); (F.M.S.)
| |
Collapse
|
4
|
Sevriev B, Dimitrova S, Kehayova G, Dragomanova S. Trehalose: Neuroprotective Effects and Mechanisms-An Updated Review. NEUROSCI 2024; 5:429-444. [PMID: 39484301 PMCID: PMC11503274 DOI: 10.3390/neurosci5040032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
Trehalose is a naturally occurring disaccharide that has recently gained significant attention for its neuroprotective properties in various models of neurodegeneration. This review provides an overview of available experimental data on the beneficial properties of trehalose for central nervous system pathological conditions. Trehalose's impact on neuronal cell survival and function was also examined. As a result, we identified that trehalose's neuroprotection includes autophagy modulation as well as its capability to stabilize proteins and inhibit the formation of misfolded ones. Moreover, trehalose mitigates oxidative stress-induced neuronal damage by stabilizing cellular membranes and modulating mitochondrial function. Furthermore, trehalose attenuates excitotoxicity-induced neuroinflammation by suppressing pro-inflammatory cytokine release and inhibiting inflammasome activation. A possible connection of trehalose with the gut-brain axis was also examined. These findings highlight the potential therapeutic effects of trehalose in neurodegenerative diseases. According to the conclusions drawn from this study, trehalose is a promising neuroprotective agent as a result of its distinct mechanism of action, which makes this compound a candidate for further research and the development of therapeutic strategies to combat neuronal damage and promote neuroprotection in various neurological diseases.
Collapse
Affiliation(s)
- Borislav Sevriev
- Faculty of Pharmacy, Medical University of Varna "Prof. Dr. Paraskev Stoyanov", 9000 Varna, Bulgaria;
| | - Simeonka Dimitrova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna "Prof. Dr. Paraskev Stoyanov", 9000 Varna, Bulgaria; (S.D.); (G.K.)
| | - Gabriela Kehayova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna "Prof. Dr. Paraskev Stoyanov", 9000 Varna, Bulgaria; (S.D.); (G.K.)
| | - Stela Dragomanova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna "Prof. Dr. Paraskev Stoyanov", 9000 Varna, Bulgaria; (S.D.); (G.K.)
| |
Collapse
|
5
|
Ghorbani M, Abouei Mehrizi M, Tajvidi M, Amin Habibi M, Mohammadi M, Esmaeilian S, Torabi P, Rahmanipour E, Daskareh M, Mohammadi A. Trehalose: A promising new treatment for traumatic brain injury? A systematic review of animal evidence. INTERDISCIPLINARY NEUROSURGERY 2024; 36:101947. [DOI: 10.1016/j.inat.2023.101947] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2024] Open
|
6
|
Gao S, Wang J, Wei L, Luo C, Qian F, Bo L, Mao C. Trehalosemodulates OVRAS to improve oxidative stress and apoptosis in KGN cells and ovaries of PCOS mice. J Ovarian Res 2024; 17:11. [PMID: 38195648 PMCID: PMC10775634 DOI: 10.1186/s13048-023-01337-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/28/2023] [Indexed: 01/11/2024] Open
Abstract
The etiology of polycystic ovary syndrome (PCOS) is complex and variable, and there is no exact cause or good treatment method. Most of the methods of hormones are used to temporarily meet the needs of patients. Experimental evidence has shown that trehalose has, anti-apoptotic, anti-oxidative, glucose-lowering, and insulin resistance effects. However, whether trehalose has a therapeutic effect on PCOS is unknown. It has been reported that the ovarian renin-angiotensin system (OVRAS) is involved in the development of PCOS, but it has not been fully elucidated. This study aims to explore the effect of trehalose on PCOS and elucidate the related OVRAS mechanism. We first observed that body weight, estrous cycle, ovarian follicles at all levels, glucose tolerance, serum hormones, and insulin resistance were improved by trehalose treatment in the PCOS mouse model. Moreover, trehalose treatment also ameliorated ovarian oxidative stress and apoptosis in PCOS mice, as determined by TUNNEL apoptosis staining, total SOD in ovarian homogenate, and WB assay. OVRAS mainly involves two classic pathways, namely the ACE/AngII/AT1R/AT2R, and ACE2 / Ang1-7/ MASR, Which play different functions. In PCOS mouse ovaries, we found that ACE/AngII/AT1R was up-regulated and ACE2/Ang1-7/MASR and AT2R were down-regulated by PCR and WB experiments, However, trehalose treatment changed its direction. In addition, we also found that trehalose ameliorated DHEA-induced oxidative stress and apoptosis in KGN by PCR and WB experiments, mainly by down-regulating ACE/AngII/AT1R. Our study shows that trehalose improves symptoms of PCOS mainly by down-regulating ACE/AngII/AT1R, revealing a potential therapeutic target for PCOS.
Collapse
Affiliation(s)
- Shasha Gao
- Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou, Jiangsu, 215000, China
| | - Juan Wang
- Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou, Jiangsu, 215000, China
| | - Lun Wei
- Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou, Jiangsu, 215000, China
| | - Chao Luo
- Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou, Jiangsu, 215000, China
| | - Fei Qian
- Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou, Jiangsu, 215000, China
| | - Le Bo
- Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou, Jiangsu, 215000, China
| | - Caiping Mao
- Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou, Jiangsu, 215000, China.
| |
Collapse
|
7
|
Jain MK, Bhat R. Modulation of the conformation, fibrillation, and fibril morphologies of human brain α-, β-, and γ-syn proteins by the disaccharide chemical chaperone trehalose. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2023:140932. [PMID: 37433401 DOI: 10.1016/j.bbapap.2023.140932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/13/2023]
Abstract
Human α-, β-, and γ-synuclein (syn) are natively unfolded proteins present in the brain. Deposition of aggregated α-syn in Lewy bodies is associated with Parkinson's disease (PD) and γ-syn is known to be involved in both neurodegeneration and breast cancer. At physiological pH, while α-syn has the highest propensity for fibrillation followed by γ-syn, β-syn does not form any fibrils. Fibril formation in these proteins could be modulated by protein structure stabilizing osmolytes such as trehalose which has an exceptional stabilizing effect for globular proteins. We present a comprehensive study of the effect of trehalose on the conformation, aggregation, and fibril morphology of α-, β-, and γ-syn proteins. Rather than stabilizing the intrinsically disordered state of the synucleins, trehalose accelerates the rate of fibril formation by forming aggregation-competent partially folded intermediate structures. Fibril morphologies are also strongly dependent on the concentration of trehalose with ≤ 0.4M favoring the formation of mature fibrils in α-, and γ-syn with no effect on the fibrillation of β-syn. At ≥ 0.8M, trehalose promotes the formation of smaller aggregates that are more cytotoxic. Live cell imaging of preformed aggregates of a labeled A90C α-syn shows their rapid internalization into neural cells which could be useful in reducing the load of aggregated species of α-syn. The findings throw light on the differential effect of trehalose on the conformation and aggregation of disordered synuclein proteins with respect to globular proteins and could help in understanding the effect of osmolytes on intrinsically disordered proteins under cellular stress conditions.
Collapse
Affiliation(s)
- Manish K Jain
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110 067, India; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rajiv Bhat
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110 067, India.
| |
Collapse
|
8
|
Hosseinpour Mashkani SM, Bishop DP, Raoufi-Rad N, Adlard PA, Shimoni O, Golzan SM. Distribution of Copper, Iron, and Zinc in the Retina, Hippocampus, and Cortex of the Transgenic APP/PS1 Mouse Model of Alzheimer's Disease. Cells 2023; 12:cells12081144. [PMID: 37190053 DOI: 10.3390/cells12081144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
A mis-metabolism of transition metals (i.e., copper, iron, and zinc) in the brain has been recognised as a precursor event for aggregation of Amyloid-β plaques, a pathological hallmark of Alzheimer's disease (AD). However, imaging cerebral transition metals in vivo can be extremely challenging. As the retina is a known accessible extension of the central nervous system, we examined whether changes in the hippocampus and cortex metal load are also mirrored in the retina. Laser ablation inductively coupled plasma-mass spectrometry (LA-ICP-MS) was used to visualise and quantify the anatomical distribution and load of Cu, Fe, and Zn in the hippocampus, cortex, and retina of 9-month-old Amyloid Precursor Protein/Presenilin 1 (APP/PS1, n = 10) and Wild Type (WT, n = 10) mice. Our results show a similar metal load trend between the retina and the brain, with the WT mice displaying significantly higher concentrations of Cu, Fe, and Zn in the hippocampus (p < 0.05, p < 0.0001, p < 0.01), cortex (p < 0.05, p = 0.18, p < 0.0001) and the retina (p < 0.001, p = 0.01, p < 0.01) compared with the APP/PS1 mice. Our findings demonstrate that dysfunction of the cerebral transition metals in AD is also extended to the retina. This could lay the groundwork for future studies on the assessment of transition metal load in the retina in the context of early AD.
Collapse
Affiliation(s)
- Seyed Mostafa Hosseinpour Mashkani
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - David P Bishop
- Hyphenated Mass Spectrometry Laboratory (HyMaS), School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - Newsha Raoufi-Rad
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Olga Shimoni
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - S Mojtaba Golzan
- Vision Science Group, Graduate School of Health (GSH), University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| |
Collapse
|
9
|
Dietary Trehalose as a Bioactive Nutrient. Nutrients 2023; 15:nu15061393. [PMID: 36986123 PMCID: PMC10054017 DOI: 10.3390/nu15061393] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/06/2023] [Accepted: 03/11/2023] [Indexed: 03/15/2023] Open
Abstract
Trehalose is a naturally occurring, non-reducing disaccharide comprising two covalently-linked glucose molecules. It possesses unique physiochemical properties, which account for multiple biological roles in a variety of prokaryotic and eukaryotic organisms. In the past few decades, intensive research on trehalose has uncovered its functions, and extended its uses as a sweetener and stabilizer in the food, medical, pharmaceutical, and cosmetic industries. Further, increased dietary trehalose consumption has sparked research on how trehalose affects the gut microbiome. In addition to its role as a dietary sugar, trehalose has gained attention for its ability to modulate glucose homeostasis, and potentially as a therapeutic agent for diabetes. This review discusses the bioactive effects of dietary trehalose, highlighting its promise in future industrial and scientific contributions.
Collapse
|
10
|
Banerjee M, Shenoy RR. Emphasizing roles of BDNF promoters and inducers in Alzheimer's disease for improving impaired cognition and memory. J Basic Clin Physiol Pharmacol 2023; 34:125-136. [PMID: 34751526 DOI: 10.1515/jbcpp-2021-0182] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/11/2021] [Indexed: 12/13/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a crucial neurotrophic factor adding to neurons' development and endurance. The amount of BDNF present in the brain determines susceptibility to various neurodegenerative diseases. In Alzheimer's disease (AD), often it is seen that low levels of BDNF are present, which primarily contributes to cognition deficit by regulating long-term potentiation (LTP) and synaptic plasticity. Molecular mechanisms underlying the synthesis, storage and release of BDNF are widely studied. New molecules are found, which contribute to the signal transduction pathway. Two important receptors of BDNF are TrkB and p75NTR. When BDNF binds to the TrkB receptor, it activates three main signalling pathways-phospholipase C, MAPK/ERK, PI3/AKT. BDNF holds an imperative part in LTP and dendritic development, which are essential for memory formation. BDNF supports synaptic integrity by influencing LTP and LTD. This action is conducted by modulating the glutamate receptors; AMPA and NMDA. This review paper discusses the aforesaid points along with inducers of BDNF. Drugs and herbals promote neuroprotection by increasing the hippocampus' BDNF level in various disease-induced animal models for neurodegeneration. Advancement in finding pertinent molecules contributing to the BDNF signalling pathway has been discussed, along with the areas that require further research and study.
Collapse
Affiliation(s)
- Madhuparna Banerjee
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Udupi District, Karnataka, India
| | - Rekha R Shenoy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Udupi District, Karnataka, India
| |
Collapse
|
11
|
Daglas M, Truong PH, Miles LQ, Juan SMA, Rao SS, Adlard PA. Deferiprone attenuates neuropathology and improves outcome following traumatic brain injury. Br J Pharmacol 2023; 180:214-234. [PMID: 36102035 DOI: 10.1111/bph.15950] [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: 02/10/2022] [Revised: 08/27/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Traumatic brain injury (TBI) remains a leading cause of mortality and morbidity in young adults. The role of iron in potentiating neurodegeneration following TBI has gained recent interest as iron deposition has been detected in the injured brain in the weeks to months post-TBI, in both the preclinical and clinical setting. A failure in iron homeostasis can lead to oxidative stress, inflammation and excitotoxicity; and whether this is a cause or consequence of the long-term effects of TBI remains unknown. EXPERIMENTAL APPROACH We investigated the role of iron and the effect of therapeutic intervention using a brain-permeable iron chelator, deferiprone, in a controlled cortical impact mouse model of TBI. An extensive assessment of cognitive, motor and anxiety/depressive outcome measures were examined, and neuropathological and biochemical changes, over a 3-month period post-TBI. KEY RESULTS Lesion volume was significantly reduced at 3 months, which was preceded by a reduction in astrogliosis, microglia/macrophages and preservation of neurons in the injured brain at 2 weeks and/or 1 month post-TBI in mice receiving oral deferiprone. Deferiprone treatment showed significant improvements in neurological severity scores, locomotor/gait performance and cognitive function, and attenuated anxiety-like symptoms post-TBI. Deferiprone reduced iron levels, lipid peroxidation/oxidative stress and altered expression of neurotrophins in the injured brain over this period. CONCLUSION AND IMPLICATIONS Our findings support a detrimental role of iron in the injured brain and suggest that deferiprone (or similar iron chelators) may be promising therapeutic approaches to improve survival, functional outcomes and quality of life following TBI.
Collapse
Affiliation(s)
- Maria Daglas
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Phan H Truong
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Linh Q Miles
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Sydney M A Juan
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Shalini S Rao
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
12
|
Sarkar A, Kumari N, Mukherjee P. The curious case of SARM1: Dr. Jekyll and Mr. Hyde in cell death and immunity? FEBS J 2023; 290:340-358. [PMID: 34710262 DOI: 10.1111/febs.16256] [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: 07/26/2021] [Revised: 09/21/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
Sterile alpha and toll/interleukin-1 receptor motif-containing protein 1 (SARM1) was first identified as a novel ortholog of Drosophila protein CG7915 and was subsequently placed as the fifth member of the human TIR-containing adaptor protein. SARM1 holds a unique position in this family where, unlike other members, it downregulates NFκB activity in response to immunogenic stimulation, interacts with another member of the family, TRIF, to negatively regulate its function, and it also mediates cell death responses. Over the past decade, SARM1 has emerged as one of the primary mediators of programmed axonal degeneration and this robust regulation of axonal degeneration-especially in models of peripheral neuropathy and traumatic injury-makes it an attractive target for therapeutic intervention. The TIR domain of SARM1 possesses an intrinsic NADase activity resulting in cellular energy deficits within the axons, a striking deviation from its other family members of human TLR adaptors. Interestingly, the TIR NADase activity, as seen in SARM1, is also observed in several prokaryotic TIR-containing proteins where they are involved in immune evasion once within the host. Although the immune function of SARM1 is yet to be conclusively discerned, this closeness in function with the prokaryotic TIR-domain containing proteins, places it at an interesting juncture of evolution raising questions about its origin and function in cell death and immunity. In this review, we discuss how a conserved immune adaptor protein like SARM1 switches to a pro-neurodegenerative function and the evolutionarily significance of the process.
Collapse
Affiliation(s)
- Ankita Sarkar
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| | - Nripa Kumari
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| | - Piyali Mukherjee
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
13
|
Potential Properties of Natural Nutraceuticals and Antioxidants in Age-Related Eye Disorders. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010077. [PMID: 36676026 PMCID: PMC9863869 DOI: 10.3390/life13010077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022]
Abstract
Eye health is crucial, and the onset of diseases can reduce vision and affect the quality of life of patients. The main causes of progressive and irreversible vision loss include various pathologies, such as cataracts, ocular atrophy, corneal opacity, age-related macular degeneration, uncorrected refractive error, posterior capsular opacification, uveitis, glaucoma, diabetic retinopathy, retinal detachment, undetermined disease and other disorders involving oxidative stress and inflammation. The eyes are constantly exposed to the external environment and, for this reason, must be protected from damage from the outside. Many drugs, including cortisonics and antinflammatory drugs have widely been used to counteract eye disorders. However, recent advances have been obtained via supplementation with natural antioxidants and nutraceuticals for patients. In particular, evidence has accumulated that polyphenols (mostly deriving from Citrus Bergamia) represent a reliable source of antioxidants able to counteract oxidative stress accompanying early stages of eye diseases. Luteolin in particular has been found to protect photoreceptors, thereby improving vision in many disease states. Moreover, a consistent anti-inflammatory response was found to occur when curcumin is used alone or in combination with other nutraceuticals. Additionally, Coenzyme Q10 has been demonstrated to produce a consistent effect in reducing ocular pressure, thereby leading to protection in patients undergoing glaucoma. Finally, both grape seed extract, rich in anthocyanosides, and polynsatured fatty acids seem to contribute to the prevention of retinal disorders. Thus, a combination of nutraceuticals and antioxidants may represent the right solution for a multi-action activity in eye protection, in association with current drug therapies, and this will be of potential interest in early stages of eye disorders.
Collapse
|
14
|
Cano GH, Dean J, Abreu SP, Rodríguez AH, Abbasi C, Hinson M, Lucke-Wold B. Key Characteristics and Development of Psychoceuticals: A Review. Int J Mol Sci 2022; 23:15777. [PMID: 36555419 PMCID: PMC9779201 DOI: 10.3390/ijms232415777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
Psychoceuticals have brought benefits to the pharmacotherapeutic management of central nervous system (CNS) illnesses since the 19th century. However, these drugs have potential side effects or lack high response rates. This review covers twenty drugs' biochemical mechanisms, benefits, risks, and clinical trial reports. For this study, medications from seven psychoceutical organizations were reviewed and evaluated. Nineteen drugs were chosen from the organizations, and one was selected from the literature. The databases used for the search were Pubmed, Google Scholar, and NIH clinical trials. In addition, information from the organizations' websites and other sources, such as news reports, were also used. From the list of drugs, the most common targets were serotonergic, opioid, and N-methyl-D-aspartate (NMDA) receptors. These drugs have shown promise in psychiatric illnesses such as substance abuse, post-traumatic stress disorder (PTSD), anxiety, depression, and neurological conditions, such as Parkinson's disease, traumatic brain injury, and neuroinflammation. Some of these drugs, however, are still early in development, so their therapeutic significance cannot be determined. These twenty drugs have promising benefits, but their clinical usage and efficacy must still be explored.
Collapse
Affiliation(s)
- Genaro Herrera Cano
- University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Jordan Dean
- University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Samuel Padilla Abreu
- University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | | | - Cyrena Abbasi
- University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Madison Hinson
- Wake Forest University School of Medicine, 475 Vine St, Winston-Salem, NC 27101, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
15
|
Chen A, Tapia H, Goddard JM, Gibney PA. Trehalose and its applications in the food industry. Compr Rev Food Sci Food Saf 2022; 21:5004-5037. [PMID: 36201393 DOI: 10.1111/1541-4337.13048] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/29/2022] [Accepted: 08/31/2022] [Indexed: 01/28/2023]
Abstract
Trehalose is a nonreducing disaccharide composed of two glucose molecules linked by α, α-1,1-glycosidic bond. It is present in a wide variety of organisms, including bacteria, fungi, insects, plants, and invertebrate animals. Trehalose has distinct physical and chemical properties that have been investigated for their biological importance in a range of prokaryotic and eukaryotic species. Emerging research on trehalose has identified untapped opportunities for its application in the food, medical, pharmaceutical, and cosmetics industries. This review summarizes the chemical and biological properties of trehalose, its occurrence and metabolism in living organisms, its protective role in molecule stabilization, and natural and commercial production methods. Utilization of trehalose in the food industry, in particular how it stabilizes protein, fat, carbohydrate, and volatile compounds, is also discussed in depth. Challenges and opportunities of its application in specific applications (e.g., diagnostics, bioprocessing, ingredient technology) are described. We conclude with a discussion on the potential of leveraging the unique molecular properties of trehalose in molecular stabilization for improving the safety, quality, and sustainability of our food systems.
Collapse
Affiliation(s)
- Anqi Chen
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Hugo Tapia
- Biology Program, California State University - Channel Islands, Camarillo, California, USA
| | - Julie M Goddard
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Patrick A Gibney
- Department of Food Science, Cornell University, Ithaca, New York, USA
| |
Collapse
|
16
|
Pupyshev AB, Klyushnik TP, Akopyan AA, Singh SK, Tikhonova MA. Disaccharide Trehalose in Experimental Therapies for Neurodegenerative Disorders: Molecular Targets and Translational Potential. Pharmacol Res 2022; 183:106373. [PMID: 35907433 DOI: 10.1016/j.phrs.2022.106373] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 10/16/2022]
Abstract
Induction of autophagy is a prospective approach to the treatment of neurodegeneration. In the recent decade, trehalose attracted special attention. It is an autophagy inducer with negligible adverse effects and is approved for use in humans according to FDA requirements. Trehalose has a therapeutic effect in various experimental models of diseases. This glucose disaccharide with a flexible α-1-1'-glycosidic bond has unique properties: induction of mTOR-independent autophagy (with kinase AMPK as the main target) and a chaperone-like effect on proteins imparting them natural spatial structure. Thus, it can reduce the accumulation of neurotoxic aberrant/misfolded proteins. Trehalose has an anti-inflammatory effect and inhibits detrimental oxidative stress partially owing to the enhancement of endogenous antioxidant defense represented by the Nrf2 protein. The disaccharide activates lysosome and autophagosome biogenesis pathways through the protein factors TFEB and FOXO1. Here we review various mechanisms of the neuroprotective action of trehalose and touch on the possibility of pleiotropic effects. Current knowledge about specific features of trehalose pharmacodynamics is discussed. The neuroprotective effects of trehalose in animal models of major neurodegenerative disorders such as Alzheimer's, Parkinson's, and Huntington's diseases are examined too. Attention is given to translational transition to clinical trials of this drug, especially oral and parenteral routes of administration. Besides, the possibility of enhancing the therapeutic benefit via a combination of mTOR-dependent and mTOR-independent autophagy inducers is analyzed. In general, trehalose appears to be a promising multitarget tool for the inhibition of experimental neurodegeneration and requires thorough investigation of its clinical capabilities.
Collapse
Affiliation(s)
- Alexander B Pupyshev
- Scientific Research Institute of Neurosciences and Medicine (SRINM); Timakova Str. 4, Novosibirsk 630117, Russia.
| | - Tatyana P Klyushnik
- Mental Health Research Center, Kashirskoye shosse 34, Moscow 115522, Russia.
| | - Anna A Akopyan
- Scientific Research Institute of Neurosciences and Medicine (SRINM); Timakova Str. 4, Novosibirsk 630117, Russia.
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, Krishna Bhawan, 594 Kha/123, Shahinoor Colony, Nilmatha, Uttar Pradesh, Lucknow 226002, India.
| | - Maria A Tikhonova
- Scientific Research Institute of Neurosciences and Medicine (SRINM); Timakova Str. 4, Novosibirsk 630117, Russia.
| |
Collapse
|
17
|
Li Y, Lei Z, Ritzel RM, He J, Li H, Choi HMC, Lipinski MM, Wu J. Impairment of autophagy after spinal cord injury potentiates neuroinflammation and motor function deficit in mice. Theranostics 2022; 12:5364-5388. [PMID: 35910787 PMCID: PMC9330534 DOI: 10.7150/thno.72713] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/24/2022] [Indexed: 01/25/2023] Open
Abstract
Autophagy is a catabolic process that degrades cytoplasmic constituents and organelles in the lysosome, thus serving an important role in cellular homeostasis and protection against insults. We previously reported that defects in autophagy contribute to neuronal cell damage in traumatic spinal cord injury (SCI). Recent data from other inflammatory models implicate autophagy in regulation of immune and inflammatory responses, with low levels of autophagic flux associated with pro-inflammatory phenotypes. In the present study, we examined the effects of genetically or pharmacologically manipulating autophagy on posttraumatic neuroinflammation and motor function after SCI in mice. Methods: Young adult male C57BL/6, CX3CR1-GFP, autophagy hypomorph Becn1+/- mice, and their wildtype (WT) littermates were subjected to moderate thoracic spinal cord contusion. Neuroinflammation and autophagic flux in the injured spinal cord were assessed using flow cytometry, immunohistochemistry, and NanoString gene expression analysis. Motor function was evaluated with the Basso Mouse Scale and horizontal ladder test. Lesion volume and spared white matter were evaluated by unbiased stereology. To stimulate autophagy, disaccharide trehalose, or sucrose control, was administered in the drinking water immediately after injury and for up to 6 weeks after SCI. Results: Flow cytometry demonstrated dysregulation of autophagic function in both microglia and infiltrating myeloid cells from the injured spinal cord at 3 days post-injury. Transgenic CX3CR1-GFP mice revealed increased autophagosome formation and inhibition of autophagic flux specifically in activated microglia/macrophages. NanoString analysis using the neuroinflammation panel demonstrated increased expression of proinflammatory genes and decreased expression of genes related to neuroprotection in Becn1+/- mice as compared to WT controls at 3 days post-SCI. These findings were further validated by qPCR, wherein we observed significantly higher expression of proinflammatory cytokines. Western blot analysis confirmed higher protein expression of the microglia/macrophage marker IBA-1, inflammasome marker, NLRP3, and innate immune response markers cGAS and STING in Becn1+/- mice at 3 day after SCI. Flow cytometry demonstrated that autophagy deficit did not affect either microglial or myeloid counts at 3 days post-injury, instead resulting in increased microglial production of proinflammatory cytokines. Finally, locomotor function showed significantly worse impairments in Becn1+/- mice up to 6 weeks after SCI, which was accompanied by worsening tissue damage. Conversely, treatment with a naturally occurring autophagy inducer trehalose, reduced protein levels of p62, an adaptor protein targeting cargo to autophagosomes as well as the NLRP3, STING, and IBA-1 at 3 days post-injury. Six weeks of trehalose treatment after SCI led to improved motor function recovery as compared to control group, which was accompanied by reduced tissue damage. Conclusions: Our data indicate that inhibition of autophagy after SCI potentiates pro-inflammatory activation in microglia and is associated with worse functional outcomes. Conversely, increasing autophagy with trehalose, decreased inflammation and improved outcomes. These findings highlight the importance of autophagy in spinal cord microglia and its role in secondary injury after SCI.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Zhuofan Lei
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junyun He
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Hui Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Harry M C Choi
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Marta M Lipinski
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| |
Collapse
|
18
|
Ritzel RM, Li Y, Lei Z, Carter J, He J, Choi HMC, Khan N, Li H, Allen S, Lipinski MM, Faden AI, Wu J. Functional and transcriptional profiling of microglial activation during the chronic phase of TBI identifies an age-related driver of poor outcome in old mice. GeroScience 2022; 44:1407-1440. [PMID: 35451674 PMCID: PMC9213636 DOI: 10.1007/s11357-022-00562-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/01/2022] [Indexed: 12/14/2022] Open
Abstract
Elderly patients with traumatic brain injury (TBI) have greater mortality and poorer outcomes than younger individuals. The extent to which old age alters long-term recovery and chronic microglial activation after TBI is unknown, and evidence for therapeutic efficacy in aged mice is sorely lacking. The present study sought to identify potential inflammatory mechanisms underlying age-related outcomes late after TBI. Controlled cortical impact was used to induce moderate TBI in young and old male C57BL/6 mice. At 12 weeks post-injury, aged mice exhibited higher mortality, poorer functional outcomes, larger lesion volumes, and increased microglial activation. Transcriptomic analysis identified age- and TBI-specific gene changes consistent with a disease-associated microglial signature in the chronically injured brain, including those involved with complement, phagocytosis, and autophagy pathways. Dysregulation of phagocytic and autophagic function in microglia was accompanied by increased neuroinflammation in old mice. As proof-of-principle that these pathways have functional importance, we administered an autophagic enhancer, trehalose, in drinking water continuously for 8 weeks after TBI. Old mice treated with trehalose showed enhanced functional recovery and reduced microglial activation late after TBI compared to the sucrose control group. Our data indicate that microglia undergo chronic changes in autophagic regulation with both normal aging and TBI that are associated with poorer functional outcome. Enhancing autophagy may therefore be a promising clinical therapeutic strategy for TBI, especially in older patients.
Collapse
Affiliation(s)
- Rodney M. Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Yun Li
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Zhuofan Lei
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Jordan Carter
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Junyun He
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Harry M. C. Choi
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Niaz Khan
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Hui Li
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Samantha Allen
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Marta M. Lipinski
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Alan I. Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| |
Collapse
|
19
|
Mobini M, Radbakhsh S, Kubaski F, Eshraghi P, Vakili S, Vakili R, Khalili M, Varesvazirian M, Jamialahmadi T, Alamdaran SA, Sayedi SJ, Rajabi O, Emami SA, Reiner Ž, Sebkar A. Impact of Intravenous Trehalose Administration in Patients with Niemann-Pick Disease Types A and B. J Clin Med 2022; 11:247. [PMID: 35011993 PMCID: PMC8745869 DOI: 10.3390/jcm11010247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/21/2021] [Accepted: 01/01/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND AIMS Niemann-Pick disease (NPD) types A (NPA) and B (NPB) are caused by deficiency of the acid sphingomyelinase enzyme, which is encoded by the SMPD1 gene, resulting in progressive pathogenic accumulation of lipids in tissues. Trehalose has been suggested as an autophagy inducer with therapeutic neuroprotective effects. We performed a single-arm, open-label pilot study to assess the potential efficacy of trehalose treatment in patients with NPA and NPB patients. METHODS Five patients with NPD type A and B were enrolled in an open-label, single-arm clinical trial. Trehalose was administrated intravenously (IV) (15 g/week) for three months. The efficacy of trehalose in the management of clinical symptoms was evaluated in patients by assessing the quality of life, serum biomarkers, and high-resolution computed tomography (HRCT) of the lungs at the baseline and end of the interventional trial (day 0 and week 12). RESULTS The mean of TNO-AZL Preschool children Quality of Life (TAPQOL) scores increased in all patients after intervention at W12 compared to the baseline W0, although the difference was not statistically significant. The serum levels of lyso-SM-509 and lyso-SM were decreased in three and four patients out of five, respectively, compared with baseline. Elevated ALT and AST levels were decreased in all patients after 12 weeks of treatment; however, changes were not statistically significant. Pro-oxidant antioxidant balance (PAB) was also decreased and glutathione peroxidase (GPX) activity was increased in serum of patients at the end of the study. Imaging studies of spleen and lung HRCT showed improvement of symptoms in two patients. CONCLUSIONS Positive trends in health-related quality of life (HRQoL), serum biomarkers, and organomegaly were observed after 3 months of treatment with trehalose in patients with NPA and NPB. Although not statistically significant, due to the small number of patients enrolled, these results are encouraging and should be further explored.
Collapse
Affiliation(s)
- Moein Mobini
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
| | - Shabnam Radbakhsh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
- Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Francyne Kubaski
- Department of Genetics, UFRGS, Porto Alegre 91501970, Brazil;
- Medical Genetics Service, HCPA, Porto Alegre 90035903, Brazil
- Biodiscovery Lab, HCPA, Porto Alegre 90035903, Brazil
| | - Peyman Eshraghi
- Department of Pediatric Diseases, Akbar Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177897157, Iran;
| | - Saba Vakili
- Medical Genetic Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran; (S.V.); (R.V.)
| | - Rahim Vakili
- Medical Genetic Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran; (S.V.); (R.V.)
| | - Manijeh Khalili
- Children and Adolescents Health Research Center, Research Institute of cellular and Molecular Science in Infectious Diseases, Zahedan University of Medical Science, Zahedan 9816743463, Iran;
| | - Majid Varesvazirian
- Shafa Hospital, Kerman University of Medical Sciences, Kerman 7618751151, Iran;
| | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
| | - Seyed Ali Alamdaran
- Pediatric Radiology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
| | - Seyed Javad Sayedi
- Department of Pediatrics, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
| | - Omid Rajabi
- Department of Pharmaceutical and Food Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran;
| | - Seyed Ahmad Emami
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran;
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Center Zagreb, University of Zagreb, Kišpatićeva 12, 1000 Zagreb, Croatia;
| | - Amirhossein Sebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
20
|
Lin PH, Kuo LT, Luh HT. The Roles of Neurotrophins in Traumatic Brain Injury. LIFE (BASEL, SWITZERLAND) 2021; 12:life12010026. [PMID: 35054419 PMCID: PMC8780368 DOI: 10.3390/life12010026] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 02/08/2023]
Abstract
Neurotrophins are a collection of structurally and functionally related proteins. They play important roles in many aspects of neural development, survival, and plasticity. Traumatic brain injury (TBI) leads to different levels of central nervous tissue destruction and cellular repair through various compensatory mechanisms promoted by the injured brain. Many studies have shown that neurotrophins are key modulators of neuroinflammation, apoptosis, blood–brain barrier permeability, memory capacity, and neurite regeneration. The expression of neurotrophins following TBI is affected by the severity of injury, genetic polymorphism, and different post-traumatic time points. Emerging research is focused on the potential therapeutic applications of neurotrophins in managing TBI. We conducted a comprehensive review by organizing the studies that demonstrate the role of neurotrophins in the management of TBI.
Collapse
Affiliation(s)
- Ping-Hung Lin
- Department of Medical Education, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Lu-Ting Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan;
| | - Hui-Tzung Luh
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City 235, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei 100, Taiwan
- Correspondence: ; Tel.: +886-956279587
| |
Collapse
|
21
|
Zhou HH, Luo L, Zhai XD, Chen L, Wang G, Qin LQ, Yu Z, Xin LL, Wan Z. Sex-Specific Neurotoxicity of Dietary Advanced Glycation End Products in APP/PS1 Mice and Protective Roles of Trehalose by Inhibiting Tau Phosphorylation via GSK-3β-TFEB. Mol Nutr Food Res 2021; 65:e2100464. [PMID: 34669246 DOI: 10.1002/mnfr.202100464] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/04/2021] [Indexed: 11/11/2022]
Abstract
SCOPE It remains unclear whether dietary advanced glycation end products (dAGEs)-induced cognitive impairment is sex-dependent. Trehalose may antagonize dAGEs-induced neurotoxicity via glycogen synthase kinase-3 beta (GSK3β)-transcription factor EB (TFEB) signaling. METHODS AND RESULTS The sex-specific neurotoxicity of dAGEs and the protective role of trehalose are investigated both in vivo and in vitro. Both sexes of APP/PS1 mice are divided into three groups: that is, control, dAGEs, and dAGEs supplemented with trehalose. SHSY-5Y cells incubated with AGE-BSA and trehalose are also utilized. Dietary AGEs impair cognitive function only in female mice, which is restored by trehalose. Trehalose upregulates phosphorylated-GSK3β serine9 (p-GSK3β ser9), TFEB and transient receptor potential mucolipin 1, ADAM10, oligosaccharyl transferase-48, estrogen receptor α and induces TFEB nuclear translocation in hippocampus, elevates IDE and ERβ in cortex, while reduces p-tau ser396&404, CDK5, cathepsin B, and glial fibrillary acidic protein in hippocampus. Trehalose elevates p-GSK3β ser9, induces TFEB nuclear translocation, consequently reverses AGE-BSA-induced tau phosphorylation in vitro. CONCLUSIONS Female mice are more susceptible to the deleterious effects of dAGEs on cognitive function, which may be owing to its regulation on ERβ. Trehalose can strongly reverse dAGEs-induced tau phosphorylation by potentiating TFEB nuclear translocation via inhibiting GSK-3β.
Collapse
Affiliation(s)
- Huan-Huan Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Lan Luo
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Xue-Di Zhai
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guiping Wang
- School of Physical Education, Soochow University, No. 50, Donghuan Road, Suzhou, 215006, China
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Zengli Yu
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Li-Li Xin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou, 215123, China.,Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China
| |
Collapse
|
22
|
Trehalose Reduces Nerve Injury Induced Nociception in Mice but Negatively Affects Alertness. Nutrients 2021; 13:nu13092953. [PMID: 34578829 PMCID: PMC8469914 DOI: 10.3390/nu13092953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 07/31/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
Trehalose, a sugar from fungi, mimics starvation due to a block of glucose transport and induces Transcription Factor EB- mediated autophagy, likely supported by the upregulation of progranulin. The pro-autophagy effects help to remove pathological proteins and thereby prevent neurodegenerative diseases such as Alzheimer’s disease. Enhancing autophagy also contributes to the resolution of neuropathic pain in mice. Therefore, we here assessed the effects of continuous trehalose administration via drinking water using the mouse Spared Nerve Injury model of neuropathic pain. Trehalose had no effect on drinking, feeding, voluntary wheel running, motor coordination, locomotion, and open field, elevated plus maze, and Barnes Maze behavior, showing that it was well tolerated. However, trehalose reduced nerve injury-evoked nociceptive mechanical and thermal hypersensitivity as compared to vehicle. Trehalose had no effect on calcium currents in primary somatosensory neurons, pointing to central mechanisms of the antinociceptive effects. In IntelliCages, trehalose-treated mice showed reduced activity, in particular, a low frequency of nosepokes, which was associated with a reduced proportion of correct trials and flat learning curves in place preference learning tasks. Mice failed to switch corner preferences and stuck to spontaneously preferred corners. The behavior in IntelliCages is suggestive of sedative effects as a “side effect” of a continuous protracted trehalose treatment, leading to impairment of learning flexibility. Hence, trehalose diet supplements might reduce chronic pain but likely at the expense of alertness.
Collapse
|
23
|
The Role of BDNF in Experimental and Clinical Traumatic Brain Injury. Int J Mol Sci 2021; 22:ijms22073582. [PMID: 33808272 PMCID: PMC8037220 DOI: 10.3390/ijms22073582] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury is one of the leading causes of mortality and morbidity in the world with no current pharmacological treatment. The role of BDNF in neural repair and regeneration is well established and has also been the focus of TBI research. Here, we review experimental animal models assessing BDNF expression following injury as well as clinical studies in humans including the role of BDNF polymorphism in TBI. There is a large heterogeneity in experimental setups and hence the results with different regional and temporal changes in BDNF expression. Several studies have also assessed different interventions to affect the BDNF expression following injury. Clinical studies highlight the importance of BDNF polymorphism in the outcome and indicate a protective role of BDNF polymorphism following injury. Considering the possibility of affecting the BDNF pathway with available substances, we discuss future studies using transgenic mice as well as iPSC in order to understand the underlying mechanism of BDNF polymorphism in TBI and develop a possible pharmacological treatment.
Collapse
|
24
|
Huang J, Zhang H, Zhang J, Yu H, Lin Z, Cai Y. Spermidine Exhibits Protective Effects Against Traumatic Brain Injury. Cell Mol Neurobiol 2020; 40:927-937. [PMID: 31916070 PMCID: PMC11448793 DOI: 10.1007/s10571-019-00783-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/28/2019] [Indexed: 01/28/2023]
Abstract
Traumatic brain injury (TBI) causes permanent neurological and cognitive impairments. Effective pharmacological interventions remain elusive. Spermidine is a polyamine compound found in our body that may play a role in brain development and congenital function. In this study, we aimed to investigate the therapeutic potential of spermidine for TBI. We employed experimental closed head injury (CHI) model to evaluate the protective function of spermidine on brain injury. We assessed the neurobehavioral function recovery using Neurologic Severity Score (NSS) and Morris water maze test. At histological level, we evaluated the improvement on brain edema, brain-blood barrier integrity, and cell apoptosis. We also measured inflammatory cytokines and brain injury biomarkers to monitor the treatment outcomes. Last, we correlated the level of spermidine with CHI animal model and TBI patients with different levels of severity. Spermidine administration post-CHI was found effectively to accelerate NSS improvement and shorten latency in maze test. We observed consistent improvements in brain edema, BBB function, and cell death in spermidine-treated group. Inflammatory cytokines and TBI biomarkers, e.g., S100B, MBP and CFAP were reduced significantly in treatment group. Interestingly, inhibiting spermidine synthesis influenced the neurobehavioral recovery in CHI mice. ODC1, a rate-limiting enzyme for spermidine synthesis, was found lower in CHI mice. Serum level of spermidine was significantly lower in TBI patients with severe pathological scores. Spermidine pathway may carry an endogenous role in pathophysiological process of CHI. For the first time, we demonstrated that administrating spermidine may provide a new treatment for TBI.
Collapse
Affiliation(s)
- Jianxing Huang
- Department of Neurosurgery, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 Dong Road, Quanzhou, 362000, Fujian, China
| | - Heping Zhang
- Department of Neurosurgery, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 Dong Road, Quanzhou, 362000, Fujian, China.
| | - Jinning Zhang
- Department of Neurosurgery, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 Dong Road, Quanzhou, 362000, Fujian, China.
| | - Huiping Yu
- Department of Neurosurgery, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 Dong Road, Quanzhou, 362000, Fujian, China
| | - Zhizhong Lin
- Department of Neurosurgery, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 Dong Road, Quanzhou, 362000, Fujian, China
| | - Yonghui Cai
- Department of Neurosurgery, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 248-252 Dong Road, Quanzhou, 362000, Fujian, China
| |
Collapse
|
25
|
Trehalose for Ocular Surface Health. Biomolecules 2020; 10:biom10050809. [PMID: 32466265 PMCID: PMC7277924 DOI: 10.3390/biom10050809] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/14/2020] [Accepted: 05/21/2020] [Indexed: 12/14/2022] Open
Abstract
Trehalose is a natural disaccharide synthesized in various life forms, but not found in vertebrates. An increasing body of evidence demonstrates exceptional bioprotective characteristics of trehalose. This review discusses the scientific findings on potential functions of trehalose in oxidative stress, protein clearance, and inflammation, with an emphasis on animal models and clinical trials in ophthalmology. The main objective is to help understand the beneficial effects of trehalose in clinical trials and practice, especially in patients suffering from ocular surface disease. The discussion is supplemented with an overview of patents for the use of trehalose in dry eye and with prospects for the 2020s.
Collapse
|
26
|
Nosaka N, Martinon D, Moreira D, Crother TR, Arditi M, Shimada K. Autophagy Protects Against Developing Increased Lung Permeability and Hypoxemia by Down Regulating Inflammasome Activity and IL-1β in LPS Plus Mechanical Ventilation-Induced Acute Lung Injury. Front Immunol 2020; 11:207. [PMID: 32117318 PMCID: PMC7033480 DOI: 10.3389/fimmu.2020.00207] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/27/2020] [Indexed: 01/04/2023] Open
Abstract
Targeting inflammasome activation to modulate interleukin (IL)-1β is a promising treatment strategy against acute respiratory distress syndrome and ventilator-induced lung injury (VILI). Autophagy is a key regulator of inflammasome activation in macrophages. Here, we investigated the role of autophagy in the development of acute lung injury (ALI) induced by lipopolysaccharide (LPS) and mechanical ventilation (MV). Two hours before starting MV, 0.2 mg/kg LPS was administered to mice intratracheally. Mice were then placed on high-volume MV (30 ml/kg with 3 cmH2O positive end-expiratory pressure for 2.5 h without additional oxygen application). Mice with myeloid-specific deletion of the autophagic protein ATG16L1 (Atg16l1fl/flLysMCre) suffered severe hypoxemia (adjusted p < 0.05) and increased lung permeability (p < 0.05, albumin level in bronchoalveolar lavage fluid) with significantly higher IL-1β release into alveolar space (p < 0.05). Induction of autophagy by fasting-induced starvation led to improved arterial oxygenation (adjusted p < 0.0001) and lung permeability (p < 0.05), as well as significantly suppressed IL-1β production (p < 0.01). Intratracheal treatment with anti-mouse IL-1β monoclonal antibody (mAb; 2.5 mg/kg) significantly improved arterial oxygenation (adjusted p < 0.01) as well as lung permeability (p < 0.05). On the other hand, deletion of IL-1α gene or use of anti-mouse IL-1α mAb (2.5 mg/kg) provided no significant protection, suggesting that the LPS and MV-induced ALI is primarily dependent on IL-1β, but independent of IL-1α. These observations suggest that autophagy has a protective role in controlling inflammasome activation and production of IL-1β, which plays a critical role in developing hypoxemia and increased lung permeability in LPS plus MV-induced acute lung injury.
Collapse
Affiliation(s)
- Nobuyuki Nosaka
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Daisy Martinon
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Debbie Moreira
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Timothy R Crother
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Moshe Arditi
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Kenichi Shimada
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
27
|
Lu F, Sun X, Xu X, Jiang X. SILAC-based proteomic profiling of the suppression of TGF-β1-induced lung fibroblast-to-myofibroblast differentiation by trehalose. Toxicol Appl Pharmacol 2020; 391:114916. [PMID: 32035996 DOI: 10.1016/j.taap.2020.114916] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/05/2020] [Indexed: 12/16/2022]
Abstract
Fibroblast-to-myofibroblast differentiation is one of the most important characteristics of pulmonary fibrosis, and screening natural compounds targeting fibroblast differentiation is always a promising approach to discover drug candidates for treatment of pulmonary fibrosis. Trehalose reportedly has many potential medical applications, especially in treating neurodegeneration diseases. However, it remains unclear whether trehalose suppresses lung fibroblast differentiation. In this work, we found that trehalose decreased the expression levels of α-smooth muscle actin (α-SMA) following the induction of transforming growth factor β1 (TGF-β1) in pretreatment, co-treatment, and post-treatment groups. Trehalose also reduced the production of type I collagen, lung fibroblast-containing gel contractility and cell filament formation in TGF-β1-stimulated MRC-5 cells. Although trehalose is a known autophagy inducer, our results showed that its suppressive effect on fibroblast differentiation was not via trehalose-induced autophagy. And it did not affect canonical TGFβ/Smad2/3 pathway. By applying proteomic profiling technology, we demonstrated that the downregulation of β-catenin was involved in the trehalose-repressive action on fibroblast differentiation. The β-catenin agonist, SKL2001, reversed the suppressive effect of trehalose on fibroblast differentiation. Overall, these experiments demonstrated that trehalose suppressed fibroblast differentiation via the downregulation of β-catenin, but not through canonical autophagy and TGFβ/Smad2/3 pathway, which is not only a novel understanding of trehalose, but also quite helpful for in vivo research of trehalose on pulmonary fibrosis in future.
Collapse
Affiliation(s)
- Fanqing Lu
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Xionghua Sun
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Xiafang Xu
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Xiaogang Jiang
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China.
| |
Collapse
|
28
|
Wei L, Zhang J, Zhang B, Geng J, Tan Q, Wang L, Chen Z, Feng H, Zhu G. Complement C3 participates in the function and mechanism of traumatic brain injury at simulated high altitude. Brain Res 2019; 1726:146423. [PMID: 31654641 DOI: 10.1016/j.brainres.2019.146423] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) leads to severe mortality and disability, in which secondary injury induced by complement activation plays an important role. TBI tends to be associated with more severe cerebral edema and worse neurological functional recovery if it occurs in high-altitude areas than in low-altitude areas. However, the underlying mechanism of this difference is unknown. Thus, we used cobra venom factor (CVF) to deplete complement C3 in simulated high-altitude areas to explore whether the differences in outcome at different altitudes are related to secondary injury caused by complement C3. METHODS The weight-drop model was adopted to induce TBI in rats. Rats were randomly divided into the following groups: sham + saline (sham), high altitude + TBI + saline (HAT), high altitude + TBI + CVF (H-CVF), low altitude + TBI + saline (LAT), and low altitude + TBI + CVF (L-CVF). Brain contusion and edema volumes, brain water content, myelin basic protein (MBP) expression, tumor necrosis factor alpha (TNF-a) expression, interleukin 1 beta (IL1B) expression, mortality rate, neurological function, and complement component 3 (C3) mRNA expression were measured by techniques such as Evans blue fluorescence, Perls staining, TUNEL staining, ELISA, immunohistochemistry and Western blotting to evaluate correlations between complement activation and secondary injury. RESULTS The activation of complement after TBI was significantly higher at high altitude than at low altitude. High-altitude TBI resulted in a leakier blood-brain barrier, more severe cerebral edema and higher mortality than low-altitude TBI did. In addition, high-altitude TBI tended to be associated with more MBP degradation, ferric iron deposition, neuronal apoptosis, and inflammatory factor deposition than low-altitude TBI. All of these effects of TBI were partially reversed by inhibiting complement activation using CVF. CONCLUSION Our study provided evidence that TBI at high altitude leads to severe edema and high mortality and disability rates. Complement C3 activation is one of the important factors contributing to secondary brain injury.
Collapse
Affiliation(s)
- Linjie Wei
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Jianbo Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Bo Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Junjun Geng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Ling Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China.
| |
Collapse
|
29
|
Liu K, Jing M, Liu C, Yan D, Ma Z, Wang C, Deng Y, Liu W, Xu B. Effect of trehalose on manganese‐induced mitochondrial dysfunction and neuronal cell damage in mice. Basic Clin Pharmacol Toxicol 2019; 125:536-547. [DOI: 10.1111/bcpt.13316] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/27/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Kuan Liu
- Department of Environmental Health School of Public Health China Medical University Shenyang China
| | - Meng‐Jiao Jing
- Department of Environmental Health School of Public Health China Medical University Shenyang China
| | - Chang Liu
- Department of Environmental Health School of Public Health China Medical University Shenyang China
| | - Dong‐Ying Yan
- Department of Environmental Health School of Public Health China Medical University Shenyang China
| | - Zhuo Ma
- Department of Environmental Health School of Public Health China Medical University Shenyang China
| | - Can Wang
- Department of Environmental Health School of Public Health China Medical University Shenyang China
| | - Yu Deng
- Department of Environmental Health School of Public Health China Medical University Shenyang China
| | - Wei Liu
- Department of Environmental Health School of Public Health China Medical University Shenyang China
| | - Bin Xu
- Department of Environmental Health School of Public Health China Medical University Shenyang China
| |
Collapse
|
30
|
Howson PA, Johnston TH, Ravenscroft P, Hill MP, Su J, Brotchie JM, Koprich JB. Beneficial Effects of Trehalose on Striatal Dopaminergic Deficits in Rodent and Primate Models of Synucleinopathy in Parkinson's Disease. J Pharmacol Exp Ther 2019; 369:364-374. [PMID: 30918068 DOI: 10.1124/jpet.118.255695] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/18/2019] [Indexed: 12/26/2022] Open
Abstract
Disease modification in Parkinson's disease (PD) is an unmet medical need. In the current study, we evaluated trehalose, a safe and well-tolerated disaccharide that has previously demonstrated efficacy in rodent models of neurodegenerative diseases, including PD. In a rat model of PD, based on delivery of adeno-associated virus serotype 1/2 containing the mutated human A53T α-synuclein gene (AAV1/2-hourA53T-aSyn) to the substantia nigra (SN), we showed that rats administered trehalose (2.67 g/kg per day, by mouth) for 6 weeks had less forelimb asymmetry (93% reduction) and higher striatal dopamine (54% increase) compared with rats receiving vehicle. In a pharmacokinetic study, we determined that efficacy was associated with plasma C max of 8900 ng/ml and area under the curve from time 0 to infinity (AUC0-inf) of 11,136 hour⋅ng/ml. We then showed, in macaques, that oral administration of trehalose (2.67 g/kg per day) produced plasma exposures of similar magnitude, with plasma C max of 10,918 ng/ml and AUC0-inf of 27,445 hour⋅ng/ml. In a macaque model of PD, also based on delivery of AAV1/2-hourA53T-aSyn to the SN, trehalose (2.67 g/kg per day, by mouth), administered for 142 days, produced higher striatal dopamine (by 39%) and dopamine transporter levels (by 50%), compared with macaques receiving vehicle. In neither model did trehalose treatment prevent loss of tyrosine hydroxylase (TH) positive (TH+ve) cells in the SN or alter α-synuclein levels in the striatum. These studies demonstrated that trehalose reduces striatal dopaminergic deficits in a rodent and macaque model of synucleinopathy in PD. Furthermore, we have determined the pharmacokinetic parameters associated with efficacy, and thus defined exposures to target in future clinical trials.
Collapse
Affiliation(s)
- Patrick A Howson
- Atuka Inc., Toronto, Ontario, Canada (T.H.J., P.R., M.P.H., J.S., J.M.B., J.B.K.); Junaxo Inc., Toronto, Ontario, Canada (P.A.H.); and Krembil Research Institute, University Health Network, Toronto, Ontario, Canada (P.A.H., T.H.J., P.R., M.P.H., J.M.B., J.B.K.)
| | - Tom H Johnston
- Atuka Inc., Toronto, Ontario, Canada (T.H.J., P.R., M.P.H., J.S., J.M.B., J.B.K.); Junaxo Inc., Toronto, Ontario, Canada (P.A.H.); and Krembil Research Institute, University Health Network, Toronto, Ontario, Canada (P.A.H., T.H.J., P.R., M.P.H., J.M.B., J.B.K.)
| | - Paula Ravenscroft
- Atuka Inc., Toronto, Ontario, Canada (T.H.J., P.R., M.P.H., J.S., J.M.B., J.B.K.); Junaxo Inc., Toronto, Ontario, Canada (P.A.H.); and Krembil Research Institute, University Health Network, Toronto, Ontario, Canada (P.A.H., T.H.J., P.R., M.P.H., J.M.B., J.B.K.)
| | - Michael P Hill
- Atuka Inc., Toronto, Ontario, Canada (T.H.J., P.R., M.P.H., J.S., J.M.B., J.B.K.); Junaxo Inc., Toronto, Ontario, Canada (P.A.H.); and Krembil Research Institute, University Health Network, Toronto, Ontario, Canada (P.A.H., T.H.J., P.R., M.P.H., J.M.B., J.B.K.)
| | - Jin Su
- Atuka Inc., Toronto, Ontario, Canada (T.H.J., P.R., M.P.H., J.S., J.M.B., J.B.K.); Junaxo Inc., Toronto, Ontario, Canada (P.A.H.); and Krembil Research Institute, University Health Network, Toronto, Ontario, Canada (P.A.H., T.H.J., P.R., M.P.H., J.M.B., J.B.K.)
| | - Jonathan M Brotchie
- Atuka Inc., Toronto, Ontario, Canada (T.H.J., P.R., M.P.H., J.S., J.M.B., J.B.K.); Junaxo Inc., Toronto, Ontario, Canada (P.A.H.); and Krembil Research Institute, University Health Network, Toronto, Ontario, Canada (P.A.H., T.H.J., P.R., M.P.H., J.M.B., J.B.K.)
| | - James B Koprich
- Atuka Inc., Toronto, Ontario, Canada (T.H.J., P.R., M.P.H., J.S., J.M.B., J.B.K.); Junaxo Inc., Toronto, Ontario, Canada (P.A.H.); and Krembil Research Institute, University Health Network, Toronto, Ontario, Canada (P.A.H., T.H.J., P.R., M.P.H., J.M.B., J.B.K.)
| |
Collapse
|
31
|
Portbury SD, Hare DJ, Bishop DP, Finkelstein DI, Doble PA, Adlard PA. Trehalose elevates brain zinc levels following controlled cortical impact in a mouse model of traumatic brain injury. Metallomics 2019; 10:846-853. [PMID: 29872801 DOI: 10.1039/c8mt00068a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Zinc (Zn) deficiency is a clinical consequence of brain injury that can result in neuropathological outcomes that are exacerbated with age. Here, we present laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) imaging data showing modulation of brain Zn levels by the disaccharide trehalose in aged mice following a controlled cortical impact model of traumatic brain injury. In this proof-of-concept study, trehalose induced an increase in brain zinc levels, providing important preliminary data for larger studies using this simple carbohydrate as a modulator of this essential micronutrient in traumatic brain injury. Our results may have further implications for the treatment of a variety of neurodegenerative diseases and other disorders of the nervous system.
Collapse
Affiliation(s)
- Stuart D Portbury
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
32
|
Pupyshev AB, Tikhonova MA, Akopyan AA, Tenditnik MV, Dubrovina NI, Korolenko TA. Therapeutic activation of autophagy by combined treatment with rapamycin and trehalose in a mouse MPTP-induced model of Parkinson's disease. Pharmacol Biochem Behav 2019; 177:1-11. [DOI: 10.1016/j.pbb.2018.12.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/06/2018] [Accepted: 12/20/2018] [Indexed: 10/27/2022]
|
33
|
Portbury SD, Sedjahtera A, Perrones K, Sgambelloni C, Zhang M, Crack PJ, Finkelstein DI, Adlard PA. Metal chaperones: a novel therapeutic strategy for brain injury? Brain Inj 2018; 33:305-312. [PMID: 30507321 DOI: 10.1080/02699052.2018.1552988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE This study sought to assess the potential efficacy of a novel class of metal chaperone on the outcomes in an animal model of a controlled cortical impact. This work was predicated on previous observations that this class of compound has exhibited neuroprotective potential in other models of aging and neurodegeneration. RESEARCH DESIGN The study employed a controlled cortical impact traumatic brain injury in three month old mice with subsequent behavioral and cellular assessments to determine therapeutic efficacy. METHODS Cognitive (Y-maze) and motor assessments (Rotarod and Open Field) were employed to determine behavioral end points. Histological-based methods were utilized to assess neuronal integrity, astrocytosis, and lesion volume. OUTCOMES We demonstrate here that acute post-injury treatment with PBT2 (Prana Biotechnology) is sufficient to maintain neuronal integrity (evidenced by decreased lesion area and increased numbers of neurons; decreased astrocytosis was also present) and to normalize performance in cognitive testing (Y-maze). These effects occurred within days and were maintained for the entire duration of the study (26 days post-injury). These data support the further interrogation of the utility of metal chaperones for the treatment and/or prevention of the neuroanatomical, biochemical, and behavioral deficits that occur following brain injuries of different etiologies.
Collapse
Affiliation(s)
- S D Portbury
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - A Sedjahtera
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - K Perrones
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - C Sgambelloni
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - M Zhang
- b Department of Pharmacology and Therapeutics , The University of Melbourne , Parkville, Victoria , Australia
| | - P J Crack
- b Department of Pharmacology and Therapeutics , The University of Melbourne , Parkville, Victoria , Australia
| | - D I Finkelstein
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia
| | - P A Adlard
- a The Florey Institute of Neuroscience and Mental Health, Dementia Theme , Parkville, Victoria , Australia.,c The University of Melbourne , Melbourne , Australia
| |
Collapse
|
34
|
Zhang L, Wang H. Autophagy in Traumatic Brain Injury: A New Target for Therapeutic Intervention. Front Mol Neurosci 2018; 11:190. [PMID: 29922127 PMCID: PMC5996030 DOI: 10.3389/fnmol.2018.00190] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 05/15/2018] [Indexed: 11/23/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most devastating forms of brain injury. Many pathological mechanisms such as oxidative stress, apoptosis and inflammation all contribute to the secondary brain damage and poor outcomes of TBI. Current therapies are often ineffective and poorly tolerated, which drive the explore of new therapeutic targets for TBI. Autophagy is a highly conserved intracellular mechanism during evolution. It plays an important role in elimination abnormal intracellular proteins or organelles to maintain cell stability. Besides, autophagy has been researched in various models including TBI. Previous studies have deciphered that regulation of autophagy by different molecules and pathways could exhibit anti-oxidative stress, anti-apoptosis and anti-inflammation effects in TBI. Hence, autophagy is a promising target for further therapeutic development in TBI. The present review provides an overview of current knowledge about the mechanism of autophagy, the frequently used methods to monitor autophagy, the functions of autophagy in TBI as well as its potential molecular mechanisms based on the pharmacological regulation of autophagy.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
35
|
Mirzaie M, Karimi M, Fallah H, Khaksari M, Nazari-Robati M. Downregulation of Matrix Metalloproteinases 2 and 9 is Involved in the Protective Effect of Trehalose on Spinal Cord Injury. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2018; 7:8-16. [PMID: 30234068 PMCID: PMC6134419 DOI: 10.22088/ijmcm.bums.7.1.8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/17/2018] [Indexed: 12/02/2022]
Abstract
Upregulation of matrix metalloproteinases (MMPs), in particular MMP-2 and MMP-9 contributes to secondary pathogenesis of spinal cord injury (SCI) via promoting inflammation. Recently, we have reported that trehalose suppresses inflammatory responses following SCI. Therefore, we investigated the effect of trehalose on MMP-2 and MMP-9 expression in SCI. A weight-drop contusion SCI was induced in male rats. Then, the animals received trehalose at three doses of 10 (T10), 100 (T100) and 1000 (T1000) mM intrathecally. MMP-2 and MMP-9 transcripts were then measured in damaged spinal cord at 1, 3 and 7 days after trauma, and compared with vehicle and sham groups. Additionally, behavioral analysis was conducted for 1 week using Basso-Beattie-Bresnahan (BBB) locomotor rating scale. Our data showed an early upregulation of MMP-9 at 1 day post-SCI. However, MMP-2 expression was increased at 3 days after trauma. Treatment with 10 mM trehalose significantly reduced MMP-2 expression in 3 and 7 days (P< 0.01) and MMP-9 expression in 1, 3, and 7 days (P< 0.05) post-damage compared with vehicle. Nonetheless, downregulation of both MMPs was not observed in T100 and T1000 groups. In addition, T10 group showed more rapid recovery of hind limb strength compared with T100 and T1000 groups. We propose that the neuroprotective effect of low dose trehalose is mediated by attenuation of MMP-2 and MMP-9 expression.
Collapse
Affiliation(s)
- Masoumeh Mirzaie
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman , Iran.,Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehrnaz Karimi
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman , Iran
| | - Hossein Fallah
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman , Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Nazari-Robati
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman , Iran.,Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
36
|
Metabolic shift from glycogen to trehalose promotes lifespan and healthspan in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2018; 115:E2791-E2800. [PMID: 29511104 DOI: 10.1073/pnas.1714178115] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
As Western diets continue to include an ever-increasing amount of sugar, there has been a rise in obesity and type 2 diabetes. To avoid metabolic diseases, the body must maintain proper metabolism, even on a high-sugar diet. In both humans and Caenorhabditis elegans, excess sugar (glucose) is stored as glycogen. Here, we find that animals increased stored glycogen as they aged, whereas even young adult animals had increased stored glycogen on a high-sugar diet. Decreasing the amount of glycogen storage by modulating the C. elegans glycogen synthase, gsy-1, a key enzyme in glycogen synthesis, can extend lifespan, prolong healthspan, and limit the detrimental effects of a high-sugar diet. Importantly, limiting glycogen storage leads to a metabolic shift whereby glucose is now stored as trehalose. Two additional means to increase trehalose show similar longevity extension. Increased trehalose is entirely dependent on a functional FOXO transcription factor DAF-16 and autophagy to promote lifespan and healthspan extension. Our results reveal that when glucose is stored as glycogen, it is detrimental, whereas, when stored as trehalose, animals live a longer, healthier life if DAF-16 is functional. Taken together, these results demonstrate that trehalose modulation may be an avenue for combatting high-sugar-diet pathology.
Collapse
|