1
|
Hou J, Hess JL, Zhang C, van Rooij JGJ, Hearn GC, Fan CC, Faraone SV, Fennema-Notestine C, Lin SJ, Escott-Price V, Seshadri S, Holmans P, Tsuang MT, Kremen WS, Gaiteri C, Glatt SJ. Meta-Analysis of Transcriptomic Studies of Blood and Six Brain Regions Identifies a Consensus of 15 Cross-Tissue Mechanisms in Alzheimer's Disease and Suggests an Origin of Cross-Study Heterogeneity. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33019. [PMID: 39679839 DOI: 10.1002/ajmg.b.33019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
The comprehensive genome-wide nature of transcriptome studies in Alzheimer's disease (AD) should provide a reliable description of disease molecular states. However, the genes and molecular systems nominated by transcriptomic studies do not always overlap. Even when results do align, it is not clear if those observations represent true consensus across many studies. A couple of sources of variation have been proposed to explain this variability, including tissue-of-origin and cohort type, but its basis remains uncertain. To address this variability and extract reliable results, we utilized all publicly available blood or brain transcriptomic datasets of AD, comprised of 24 brain studies with 4007 samples from six different brain regions, and eight blood studies with 1566 samples. We identified a consensus of AD-associated genes across brain regions and AD-associated gene-sets across blood and brain, generalizable machine learning and linear scoring classifiers, and significant contributors to biological diversity in AD datasets. While AD-associated genes did not significantly overlap between blood and brain, our findings highlighted 15 dysregulated processes shared across blood and brain in AD. The top five most significantly dysregulated processes were DNA replication, metabolism of proteins, protein localization, cell cycle, and programmed cell death. Conversely, addressing the discord across studies, we found that large-scale gene co-regulation patterns can account for a significant fraction of variability in AD datasets. Overall, this study ranked and characterized a compilation of genes and molecular systems consistently identified across a large assembly of AD transcriptome studies in blood and brain, providing potential candidate biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jiahui Hou
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jonathan L Hess
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Chunling Zhang
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jeroen G J van Rooij
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gentry C Hearn
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Chun Chieh Fan
- Department of Cognitive Science, University of California San Diego, La Jolla, California, USA
| | - Stephen V Faraone
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Shu-Ju Lin
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Valentina Escott-Price
- Dementia Research Institute, School of Medicine, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurology and Medical Research Council (MRC) Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Sudha Seshadri
- Department of Neurology, School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Peter Holmans
- Division of Psychological Medicine and Clinical Neurology and Medical Research Council (MRC) Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Ming T Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Chris Gaiteri
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Stephen J Glatt
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Public Health and Preventive Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
2
|
Naeem A, Waseem A, Khan MA, Robertson AA, Raza SS. Therapeutic Potential of MCC950 in Restoring Autophagy and Cognitive Function in STZ-Induced Rat Model of Alzheimer's Disease. Mol Neurobiol 2025; 62:6041-6058. [PMID: 39702834 DOI: 10.1007/s12035-024-04662-y] [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: 07/15/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024]
Abstract
Alzheimer's disease (AD) is currently the seventh leading cause of death worldwide. In this study, we explored the critical role of autophagy in AD pathology using a streptozotocin (STZ)-induced AD model in Wistar rats. The experimental groups included sham, STZ-induced AD, and STZ + MCC950-treated animals. Our findings revealed that administering two doses of STZ (3 mg/kg) intracerebroventricular at the interval of 48 h (on days 0 and 2), triggered autophagy, as evidenced by elevated levels of autophagy markers such as LC3II, ULK1, Beclin1, Ambra1, Cathepsin B, and a reduction in p62 levels. Behavioral assessments, including the water maze and novel object recognition tests, confirmed cognitive deficits and memory impairment, while the open-field test indicated increased anxiety in STZ-induced AD rats. In particular, treating the STZ-induced AD group with MCC950 (50 mg/kg) decreased the overexpression of autophagy-related proteins, which was consistent with better behavioral outcomes and lower anxiety. Overall, this study highlights new insights into AD pathophysiology and suggests potential therapeutic avenues.
Collapse
Affiliation(s)
- Abdul Naeem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow, 226003, India
| | - Arshi Waseem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow, 226003, India
| | - Mohsin Ali Khan
- Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow, 226003, India
| | - Avril Ab Robertson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow, 226003, India.
| |
Collapse
|
3
|
Haghi M, Masoudi R, Ataellahi F, Yousefi R, Najibi SM. Role of Tau and Amyloid-beta in autophagy gene dysregulation through oxidative stress. Tissue Cell 2025; 93:102765. [PMID: 39923646 DOI: 10.1016/j.tice.2025.102765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/18/2025] [Accepted: 01/25/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by memory impairment and cognitive decline. Our previous research has demonstrated that pathological Tau and Amyloid-beta (Aβ) disrupt autophagy gene expression, independently. Other studies have shown that these pathological aggregates create a vicious cycle with oxidative stress. METHODS In the current research, the effect of Tau and Amyloid-beta was compared on behavioral function, autophagy gene dysfunction, and oxidative stress in the Drosophila model for AD. Thymoquinone (TQ), an antioxidant agent, was then tested to examine if it could ameliorate the adverse effects of Tau and Amyloid-beta. In addition, the impact of TQ on Tau aggregation was investigated in vitro. RESULTS Our data showed that Tau and Amyloid-beta induced behavioral disability, autophagy gene dysregulation, and oxidative stress. TQ treatment significantly improved conditions in both types of transgenic flies, with a more profound alleviation in Tau transgenic flies, despite tau having a greater impact on autophagy gene dysregulation. Furthermore, TQ prevented the aggregation of Tau in vitro. CONCLUSION To sum up, Tau may exert its toxic effect on autophagy and behavioral dysfunctions significantly through oxidative stress while Amyloid-beta may confer its toxicity through multiple pathways, including oxidative stress. Moreover, since TQ ameliorates the adverse effect of tau and amyloid beta, it could be considered a promising approach for treating AD, probably in combination with other medications against Aβ or Tau.
Collapse
Affiliation(s)
- Mehrnaz Haghi
- Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran.
| | - Raheleh Masoudi
- Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran.
| | - Fatemeh Ataellahi
- Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran.
| | - Reza Yousefi
- Protein Chemistry Laboratory (PCL), Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran.
| | - Seyed Morteza Najibi
- Department of Biochemistry and Structural Biology, Lund University, Box 124, Lund 22100, Sweden.
| |
Collapse
|
4
|
Armeli F, Mengoni B, Schifano E, Lenz T, Archer T, Uccelletti D, Businaro R. The Probiotic Yeast, Milmed, Promotes Autophagy and Antioxidant Pathways in BV-2 Microglia Cells and C. elegans. Antioxidants (Basel) 2025; 14:393. [PMID: 40298639 PMCID: PMC12023983 DOI: 10.3390/antiox14040393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Autophagy, a catabolic process essential for maintaining cellular homeostasis, declines with age and unhealthy lifestyles, contributing to neurodegenerative diseases. Probiotics, including Milmed yeast, have demonstrated anti-inflammatory and antioxidant properties. This study evaluated the activity of Milmed on BV-2 microglial cells in vitro and in the in vivo model of Caenorhabditis elegans (C. elegans) in restoring autophagic processes. Methods: BV-2 microglial cells were incubated with S. cerevisiae (Milmed treated yeast or untreated yeast) and then stimulated with lipopolysaccharide (LPS). mRNAs of the autophagic factors and antioxidant enzymes were assessed by qPCR; mTOR and NRF2 were evaluated by ELISA. pNRF2 compared with cytosolic NRF2 was evaluated by immunofluorescence. The longevity, body size, and reactive oxygen species (ROS) levels of C. elegans were measured by fluorescence microscopy. Results: Treatment with Milmed YPD cultured yeast or the dried powder obtained from it promoted autophagic flux, as shown by the increased expression of the Beclin-1, ATG7, LC3, and p62 mRNAs and the inhibition of mTOR, as evaluated by ELISA. It also enhanced the antioxidant response by increasing the expression of NRF2, SOD1, and GPX; moreover, pNRF2 expression compared with cytosolic NRF2 expression was enhanced, as shown by immunofluorescence. Milmed dietary supplementation prolonged the survival of C. elegans and reduced the age-related ROS accumulation without changing the expression of gst-4. The pro-longevity effect was found to be dependent on SKN-1/Nrf2 activation, as shown by the absence of benefit in skn-1 mutants. Conclusions: Milmed yeast demonstrates significant pro-autophagy and antioxidant activity with significant pro-longevity effects in C. elegans, thereby extending the lifespan and improving stress resistance, which, together with the previously demonstrated anti-inflammatory activity, highlights its role as a highly effective probiotic for its beneficial health effects. Activation of the SKN-1/NRF2 pathway and the modulation of autophagy support the therapeutic potential of Milmed in neuroprotection and healthy aging.
Collapse
Affiliation(s)
- Federica Armeli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (F.A.); (B.M.)
- Department of Human Sciences, European University of Rome, 00163 Rome, Italy
| | - Beatrice Mengoni
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (F.A.); (B.M.)
| | - Emily Schifano
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (E.S.); (D.U.)
| | - Thomas Lenz
- Milmed Unico AB, 11139 Stockholm, Sweden; (T.L.); (T.A.)
| | - Trevor Archer
- Milmed Unico AB, 11139 Stockholm, Sweden; (T.L.); (T.A.)
| | - Daniela Uccelletti
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (E.S.); (D.U.)
- NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, 00185 Rome, Italy
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (F.A.); (B.M.)
| |
Collapse
|
5
|
Yang S, Wang L, Liang X, Pei T, Zeng Y, Xie B, Wang Y, Yang M, Wei D, Cheng W. Radix Hedysari Polysaccharides modulate the gut-brain axis and improve cognitive impairment in SAMP8 mice. Int J Biol Macromol 2025; 306:141715. [PMID: 40044002 DOI: 10.1016/j.ijbiomac.2025.141715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/09/2025]
Abstract
OBJECTIVE Radix Hedysari Polysaccharides (RHP) are the principal bioactive constituents of the traditional Chinese medicinal herb Radix Hedysari. This study aims to evaluate the neuroprotective effects of RHP in both cellular and animal models of Alzheimer's disease (AD) and to elucidate the underlying molecular mechanisms. METHODS HT22 cells subjected to Aβ25-35-induced cytotoxicity were pretreated with RHP, followed by assessments of reactive oxygen species (ROS) generation, mitochondrial superoxide (mSOX) levels, and mitochondrial membrane potential (ΔΨm). Senescence-accelerated mouse-prone 8 (SAMP8) mice were orally administered RHP for 12 weeks. Behavioral assays were conducted to assess cognitive function, while metabolomic and proteomic analyses were performed to examine serum metabolic alterations and hippocampal protein expression profiles. Additionally, neuronal autophagy and gut barrier integrity were evaluated using immunohistochemistry, transmission electron microscopy, and biomarker quantification. RESULTS RHP treatment significantly attenuated Aβ25-35-induced oxidative stress in HT22 cells by reducing ROS and mSOX production while preserving ΔΨm. In SAMP8 mice, RHP improved cognitive performance, preserved hippocampal mitochondrial ultrastructure, and enhanced neuronal autophagic activity. Moreover, RHP modulated serum metabolic pathways and alleviated gut barrier dysfunction, suggesting a role in gut-brain axis regulation. CONCLUSION RHP ameliorates cognitive impairment in SAMP8 mice, potentially through its modulation of systemic metabolism, mitigation of neuronal mitochondrial damage, and restoration of gut barrier integrity. These findings highlight the therapeutic potential of RHP in AD intervention and warrant further investigation into its mechanistic underpinnings.
Collapse
Affiliation(s)
- Sixia Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Linshuang Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaotong Liang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Tingting Pei
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou 510145, China
| | - Yi Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Bicen Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yuhua Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Min Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Dongfeng Wei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Weidong Cheng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
6
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial Autophagic Dysregulation in Traumatic Brain Injury: Molecular Insights and Therapeutic Avenues. ACS Chem Neurosci 2025; 16:543-562. [PMID: 39920904 DOI: 10.1021/acschemneuro.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2025] Open
Abstract
Traumatic brain injury (TBI) is a complex and multifaceted condition that can result in cognitive and behavioral impairments. One aspect of TBI that has received increasing attention in recent years is the role of microglia, the brain-resident immune cells, in the pathophysiology of the injury. Specifically, increasing evidence suggests that dysfunction in microglial autophagy, the process by which cells degrade and recycle their own damaged components, may contribute to the development and progression of TBI-related impairments. Here, we unravel the pathways by which microglia autophagic dysregulation predisposes the brain to secondary damage and neurological deficits following TBI. An overview of the role of autophagic dysregulation in perpetuation and worsening of the inflammatory response, neuroinflammation, and neuronal cell death in TBI follows. Further, we have evaluated several signaling pathways and processes that contribute to autophagy dysfunction-mediated inflammation, neurodegeneration, and poor outcome in TBI. Additionally, a discussion on the small molecule therapeutics employed to modulate these pathways and mechanisms to treat TBI have been presented. However, additional research is required to fully understand the processes behind these underlying pathways and uncover potential therapeutic targets for restoring microglial autophagic failure in TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, California 92037, United States
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University, Sakaka 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research, New Delhi 201002, India
| | | |
Collapse
|
7
|
Shilatifard A, Ben-Sahra I. Purifying and profiling lysosomes to expand understanding of lysosomal dysfunction-associated diseases. J Clin Invest 2025; 135:e188507. [PMID: 39959975 PMCID: PMC11827833 DOI: 10.1172/jci188507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
Lysosome storage dysfunction plays a central role in numerous human diseases, but a lack of appropriate tools has hindered lysosomal content profiling in clinical settings. In this issue of the JCI, Saarela et al. introduce a method called tagless LysoIP that enabled rapid isolation of intact lysosomes from blood and brain cells via immunoprecipitation of the endogenous protein TMEM192. Applied to the neurodegenerative lysosomal storage disorder known as Batten disease (caused by mutations in the CLN3 gene), tagless LysoIP revealed substantial accumulation of glycerophosphodiesters (GPDs) in patient lysosomes. These findings highlight the role of CLN3 in GPD clearance and present an innovative method that will enable biomarker discovery and therapeutic advancement in lysosomal diseases.
Collapse
|
8
|
Fernandes SM, Mayer J, Nilsson P, Shimozawa M. How close is autophagy-targeting therapy for Alzheimer's disease to clinical use? A summary of autophagy modulators in clinical studies. Front Cell Dev Biol 2025; 12:1520949. [PMID: 39845082 PMCID: PMC11750832 DOI: 10.3389/fcell.2024.1520949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder clinically characterized by progressive decline of memory and cognitive functions, and it is the leading cause of dementia accounting for 60%-80% of dementia patients. A pathological hallmark of AD is the accumulation of aberrant protein/peptide aggregates such as extracellular amyloid plaques containing amyloid-beta peptides and intracellular neurofibrillary tangles composed of hyperphosphorylated tau. These aggregates result from the failure of the proteostasis network, which encompasses protein synthesis, folding, and degradation processes. Autophagy is an intracellular self-digesting system responsible for the degradation of protein aggregates and damaged organelles. Impaired autophagy is observed in most neurodegenerative disorders, indicating the link between autophagy dysfunction and these diseases. A massive accumulation of autophagic vacuoles in neurons in Alzheimer's brains evidences autophagy impairment in AD. Modulating autophagy has been proposed as a therapeutic strategy for AD because of its potential to clear aggregated proteins. However, autophagy modulation therapy for AD is not yet clinically available. This mini-review aims to summarize clinical studies testing potential autophagy modulators for AD and to evaluate their proximity to clinical use. We accessed clinicaltrials.gov provided by the United States National Institutes of Health to identify completed and ongoing clinical trials. Additionally, we discuss the limitations and challenges of these therapies.
Collapse
Affiliation(s)
| | | | | | - Makoto Shimozawa
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Bathla S, Datta D, Bolat D, Woo E, Duque A, Arellano J, Arnsten A, Nairn AC. Dysregulated calcium signaling in the aged macaque entorhinal cortex associated with tau hyperphosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.626721. [PMID: 39713378 PMCID: PMC11661118 DOI: 10.1101/2024.12.05.626721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Tau pathology in sporadic Alzheimer's disease (AD) follows a distinct pattern, beginning in the entorhinal cortex (ERC) and spreading to interconnected brain regions. Early-stage tau pathology, characterized by soluble phosphorylated tau, is difficult to study in human brains post-mortem due to rapid dephosphorylation. Rhesus macaques, which naturally develop age-related tau pathology resembling human AD, provide an ideal model for investigating early tau etiology. This study examines the molecular processes underlying tau pathology in the macaque ERC, focusing on calcium and inflammatory signaling pathways. Our findings reveal age-related decreases in PDE4 phosphodiesterases that hydrolyze cAMP and increases in calpain-2 and GCPII that occur in parallel with early-stage tau hyperphosphorylation at multiple epitopes (pS214-tau, pT181-tau, pT217-tau). These findings suggest that dysregulated calcium signaling in ERC, beginning in middle-age, primes tau for hyperphosphorylation, potentially driving the early stages of AD, advancing our understanding of how ERC vulnerabilities contribute to neurodegeneration in AD.
Collapse
|
10
|
García Porta C, Mahfooz K, Komorowska J, Garcia-Rates S, Greenfield S. A Novel 14mer Peptide Inhibits Autophagic Flux via Selective Activation of the mTORC1 Signalling Pathway: Implications for Alzheimer's Disease. Int J Mol Sci 2024; 25:12837. [PMID: 39684549 DOI: 10.3390/ijms252312837] [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: 10/27/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
During development, a 14mer peptide, T14, modulates cell growth via the α-7 nicotinic acetylcholine receptor (α7 nAChR). However, this process could become excitotoxic in the context of the adult brain, leading to pathologies such as Alzheimer's disease (AD). Recent work shows that T14 acts selectively via the mammalian target of rapamycin complex 1 (mTORC1). This pathway is essential for normal development but is overactive in AD. The triggering of mTORC1 has also been associated with the suppression of autophagy, commonly observed in ageing and neurodegeneration. We therefore investigated the relationship between T14 and autophagic flux in tissue cultures, mouse brain slices, and human Alzheimer's disease hippocampus. Here, we demonstrate that T14 and p-mTOR s2448 expression significantly increases in AD human hippocampus, which was associated with the gradual decrease in the autophagosome number across Braak stages. During development, the reduction in T14 positively correlated with pTau (Ser202, Thr205) and two selective autophagy receptors: p62 and optineurin. In vitro studies also indicated that T14 increases p-mTOR s2448 expression, resulting in the aggregation of polyubiquinated substances. The effective blockade of T14 via its cyclic variant, NBP14, has been validated in vitro, in vivo, and ex vivo. In this study, NBP14 significantly attenuated p-mTOR s2448 expression and restored normal autophagic flux, as seen with rapamycin. We conclude that T14 acts at the α-7 receptor to selectively activate the mTORC1 pathway and consequently inhibit autophagic flux. Hence, this study describes a further step in the process by which T14 could drive neurodegeneration.
Collapse
Affiliation(s)
- Cloe García Porta
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| | - Kashif Mahfooz
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| | - Joanna Komorowska
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| | - Sara Garcia-Rates
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| | - Susan Greenfield
- Neuro-Bio Ltd., Building F5, Culham Science Centre, Abingdon OX14 3DB, UK
| |
Collapse
|
11
|
Barmaki H, Nourazarian A, Shademan B, Khaki-Khatibi F. The autophagy paradox: A new hypothesis in neurodegenerative disorders. Neurochem Int 2024; 179:105827. [PMID: 39111406 DOI: 10.1016/j.neuint.2024.105827] [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: 05/22/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
A recent study showed that while autophagy is usually tied to protein and organelle turnover, it can also play dual roles in neurodegenerative diseases. Traditionally, autophagy was seen as protective since it removes damaged proteins and organelles. but new data suggests autophagy can sometimes promote neuron death. and This review tackles autophagy's seemingly contradictory effects in neurodegeneration, or the "autophagy paradox. " It offers a framework for understanding autophagy in neurodegenerative research and the cellular processes involved. In short, our data uncovers a harmful autophagy role in certain situations, conflicting the view that it's always beneficial. We describe the distinct, disease-specific autophagy pathways functioning in various neurodegenerative diseases. Part two concerns potential therapeutic implications of manipulating autophagy and current strategies targeting the autophagic system, suggesting interesting areas for future research into tailored modulators. This could eventually enable activating or controlling specific autophagy pathways and aid in developing more effective treatments. Researchers believe more molecular-level research is needed so patient-tailored autophagy-modulating therapeutics can be developed given this dilemma. Moreover, research must translate faster into effective neurodegenerative disease treatment options. This article aims to provide a wholly new perspective on autophagy's classically described role in these severe diseases, challenging current dogma and opening new therapeutic avenue options.
Collapse
Affiliation(s)
- Haleh Barmaki
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran; Student Research Committee, Khoy University of Medical Sciences, Khoy, Iran.
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Khaki-Khatibi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
12
|
Althobaiti NA, Al-Abbas NS, Alsharif I, Albalawi AE, Almars AI, Basabrain AA, Jafer A, Ellatif SA, Bauthman NM, Almohaimeed HM, Soliman MH. Gadd45A-mediated autophagy regulation and its impact on Alzheimer's disease pathogenesis: Deciphering the molecular Nexus. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167353. [PMID: 39004381 DOI: 10.1016/j.bbadis.2024.167353] [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: 12/30/2023] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND The growth arrest and DNA damage-inducible 45 (Gadd45) gene has been implicated in various central nervous system (CNS) functions, both normal and pathological, including aging, memory, and neurodegenerative diseases. In this study, we examined whether Gadd45A deletion triggers pathways associated with neurodegenerative diseases including Alzheimer's disease (AD). METHODS Utilizing transcriptome data from AD-associated hippocampus samples, we identified Gadd45A as a pivotal regulator of autophagy. Comprehensive analyses, including Gene Ontology enrichment and protein-protein interaction network assessments, highlighted Cdkn1A as a significant downstream target of Gadd45A. Experimental validation confirmed Gadd45A's role in modulating Cdkn1A expression and autophagy levels in hippocampal cells. We also examined the effects of autophagy on hippocampal functions and proinflammatory cytokine secretion. Additionally, a murine model was employed to validate the importance of Gadd45A in neuroinflammation and AD pathology. RESULTS Our study identified 20 autophagy regulatory factors associated with AD, with Gadd45A emerging as a critical regulator. Experimental findings demonstrated that Gadd45A influences hippocampal cell fate by reducing Cdkn1A expression and suppressing autophagic activity. Comparisons between wild-type (WT) and Gadd45A knockout (Gadd45A-/-) mice revealed that Gadd45A-/- mice exhibited significant cognitive impairments, including deficits in working and spatial memory, increased Tau hyperphosphorylation, and elevated levels of kinases involved in Tau phosphorylation in the hippocampus. Additionally, Gadd45A-/- mice showed significant increases in pro-inflammatory cytokines and decreases autophagy markers in the brain. Neurotrophin levels and dendritic spine length were also reduced in Gadd45A-/- mice, likely contributing to the observed cognitive deficits. CONCLUSIONS These findings support the direct involvement of the Gadd45A gene in AD pathogenesis, and enhancing the expression of Gadd45A may represent a promising therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Norah A Althobaiti
- Biology Department, College of Science and Humanities, Shaqra University, Saudi Arabia
| | - Nouf S Al-Abbas
- Department of Biology, Jamoum University College, Umm Al-Qura University, 21955 Makkah, Saudi Arabia
| | - Ifat Alsharif
- Department of Biology, Jamoum University College, Umm Al-Qura University, 21955 Makkah, Saudi Arabia
| | - Aishah E Albalawi
- Faculty of Science, Department of Biology, University of Tabuk, Tabuk 47913, Saudi Arabia
| | - Amany I Almars
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ammar A Basabrain
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ayman Jafer
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sawsan Abd Ellatif
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), Alexandria 21934, Egypt
| | - Nuha M Bauthman
- Department of Obstetric & Gynecology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hailah M Almohaimeed
- Department of Basic Science, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mona H Soliman
- Botany and Microbiology Department, Faculty of Science, Cairo University, Giza 12613, Egypt; Biology Department, Faculty of Science, Taibah University, Al-Sharm, Yanbu El-Bahr, Yanbu 46429, Saudi Arabia.
| |
Collapse
|
13
|
Sun F, Wang J, Meng L, Zhou Z, Xu Y, Yang M, Li Y, Jiang T, Liu B, Yan H. AdipoRon promotes amyloid-β clearance through enhancing autophagy via nuclear GAPDH-induced sirtuin 1 activation in Alzheimer's disease. Br J Pharmacol 2024; 181:3039-3063. [PMID: 38679474 DOI: 10.1111/bph.16400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/28/2024] [Accepted: 03/21/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND AND PURPOSE Amyloid-β (Aβ) peptide is one of the more important pathological markers in Alzheimer's disease (AD). The development of AD impairs autophagy, which results in an imbalanced clearance of Aβ. Our previous research demonstrated that AdipoRon, an agonist of adiponectin receptors, decreased the deposition of Aβ and enhanced cognitive function in AD. However, the exact mechanisms by which AdipoRon affects Aβ clearance remain unclear. EXPERIMENTAL APPROACH We studied how AdipoRon affects autophagy in HT22 cells and APP/PS1 transgenic mice. We also investigated the signalling pathway involved and used pharmacological inhibitors to examine the role of autophagy in this process. KEY RESULTS AdipoRon promotes Aβ clearance by activating neuronal autophagy in the APP/PS1 transgenic mice. Interestingly, we found that AdipoRon induces the nuclear translocation of GAPDH, where it interacts with the SIRT1/DBC1 complex. This interaction then leads to the release of DBC1 and the activation of SIRT1, which in turn activates autophagy. Importantly, we found that inhibiting either GAPDH or SIRT1 to suppress the activity of SIRT1 counteracts the elevated autophagy and decreased Aβ deposition caused by AdipoRon. This suggests that SIRT1 plays a critical role in the effect of AdipoRon on autophagic induction in AD. CONCLUSION AND IMPLICATIONS AdipoRon promotes the clearance of Aβ by enhancing autophagy through the AdipoR1/AMPK-dependent nuclear translocation of GAPDH and subsequent activation of SIRT1. This novel molecular pathway sheds light on the modulation of autophagy in AD and may lead to the development of new therapeutic strategies targeting this pathway.
Collapse
Affiliation(s)
- Fengjiao Sun
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Department of Pharmacology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Jiangong Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Department of Pharmacology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Lingbin Meng
- Department of Pharmacology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Zhenyu Zhou
- Department of Pharmacology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Yong Xu
- Department of Pharmacology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Meizi Yang
- Department of Pharmacology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Yixin Li
- Department of Pharmacology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Tianrui Jiang
- Department of Pharmacology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Bin Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
| | - Haijing Yan
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Department of Pharmacology, School of Basic Medicine, Binzhou Medical University, Yantai, China
| |
Collapse
|
14
|
Zhou X, Zhu Z, Kuang S, Huang K, Li Y, Wang Y, Chen H, Hoi MPM, Xu B, Yang X, Zhang Z. Tetramethylpyrazine Nitrone (TBN) Reduces Amyloid β Deposition in Alzheimer's Disease Models by Modulating APP Expression, BACE1 Activity, and Autophagy Pathways. Pharmaceuticals (Basel) 2024; 17:1005. [PMID: 39204110 PMCID: PMC11357250 DOI: 10.3390/ph17081005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with age. A wealth of evidence indicates that the amyloid β (Aβ) aggregates result from dyshomeostasis between Aβ production and clearance, which plays a pivotal role in the pathogenesis of AD. Consequently, therapies targeting Aβ reduction represent a promising strategy for AD intervention. Tetramethylpyrazine nitrone (TBN) is a novel tetramethylpyrazine derivative with potential for the treatment of AD. Previously, we demonstrated that TBN markedly enhanced cognitive functions and decreased the levels of Aβ, APP, BACE 1, and hyperphosphorylated tau in 3×Tg-AD mice. However, the mechanism by which TBN inhibits Aβ deposition is still unclear. In this study, we employed APP/PS1 mice treated with TBN (60 mg/kg, ig, bid) for six months, and N2a/APP695swe cells treated with TBN (300 μM) to explore the mechanism of TBN in Aβ reduction. Our results indicate that TBN significantly alleviated cognitive impairment and reduced Aβ deposition in APP/PS1 mice. Further investigation of the underlying mechanisms revealed that TBN decreased the expression of APP and BACE1, activated the AMPK/mTOR/ULK1 autophagy pathway, inhibited the PI3K/AKT/mTOR/ULK1 autophagy pathway, and decreased the phosphorylation levels of JNK and ERK in APP/PS1 mice. Moreover, TBN was found to significantly reduce the mRNA levels of APP and BACE1, as well as those of SP1, CTCF, TGF-β, and NF-κB, transcription factors involved in regulating gene expression. Additionally, TBN was observed to decrease the level of miR-346 and increase the levels of miR-147 and miR-106a in the N2a/APP695swe cells. These findings indicate that TBN may reduce Aβ levels likely by reducing APP expression by regulating APP gene transcriptional factors and miRNAs, reducing BACE1 expression, and promoting autophagy activities.
Collapse
Affiliation(s)
- Xinhua Zhou
- Guangzhou Medical Research Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou 510440, China; (X.Z.); (S.K.); (K.H.); (Y.L.)
| | - Zeyu Zhu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, Institute of New Drug Research, Jinan University, Guangzhou 511436, China; (Z.Z.); (Y.W.)
| | - Shaoming Kuang
- Guangzhou Medical Research Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou 510440, China; (X.Z.); (S.K.); (K.H.); (Y.L.)
| | - Kaipeng Huang
- Guangzhou Medical Research Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou 510440, China; (X.Z.); (S.K.); (K.H.); (Y.L.)
| | - Yueping Li
- Guangzhou Medical Research Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou 510440, China; (X.Z.); (S.K.); (K.H.); (Y.L.)
| | - Yuqiang Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, Institute of New Drug Research, Jinan University, Guangzhou 511436, China; (Z.Z.); (Y.W.)
- Guangdong-Hong Kong-Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University College of Pharmacy, Guangzhou 511436, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University College of Pharmacy, Guangzhou 511436, China
| | - Haiyun Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China;
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinse Medical Sciences, University of Macau, Macau, China;
| | - Benhong Xu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China;
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China;
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, Institute of New Drug Research, Jinan University, Guangzhou 511436, China; (Z.Z.); (Y.W.)
- Guangdong-Hong Kong-Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University College of Pharmacy, Guangzhou 511436, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University College of Pharmacy, Guangzhou 511436, China
| |
Collapse
|
15
|
Chau DDL, Yu Z, Chan WWR, Yuqi Z, Chang RCC, Ngo JCK, Chan HYE, Lau KF. The cellular adaptor GULP1 interacts with ATG14 to potentiate autophagy and APP processing. Cell Mol Life Sci 2024; 81:323. [PMID: 39080084 PMCID: PMC11335243 DOI: 10.1007/s00018-024-05351-8] [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: 01/15/2024] [Revised: 06/06/2024] [Accepted: 07/05/2024] [Indexed: 08/22/2024]
Abstract
Autophagy is a highly conserved catabolic mechanism by which unnecessary or dysfunctional cellular components are removed. The dysregulation of autophagy has been implicated in various neurodegenerative diseases, including Alzheimer's disease (AD). Understanding the molecular mechanism(s)/molecules that influence autophagy may provide important insights into developing therapeutic strategies against AD and other neurodegenerative disorders. Engulfment adaptor phosphotyrosine-binding domain-containing protein 1 (GULP1) is an adaptor that interacts with amyloid precursor protein (APP) to promote amyloid-β peptide production via an unidentified mechanism. Emerging evidence suggests that GULP1 has a role in autophagy. Here, we show that GULP1 is involved in autophagy through an interaction with autophagy-related 14 (ATG14), which is a regulator of autophagosome formation. GULP1 potentiated the stimulatory effect of ATG14 on autophagy by modulating class III phosphatidylinositol 3-kinase complex 1 (PI3KC3-C1) activity. The effect of GULP1 is attenuated by a GULP1 mutation (GULP1m) that disrupts the GULP1-ATG14 interaction. Conversely, PI3KC3-C1 activity is enhanced in cells expressing APP but not in those expressing an APP mutant that does not bind GULP1, which suggests a role of GULP1-APP in regulating PI3KC3-C1 activity. Notably, GULP1 facilitates the targeting of ATG14 to the endoplasmic reticulum (ER). Moreover, the levels of both ATG14 and APP are elevated in the autophagic vacuoles (AVs) of cells expressing GULP1, but not in those expressing GULP1m. APP processing is markedly enhanced in cells co-expressing GULP1 and ATG14. Hence, GULP1 alters APP processing by promoting the entry of APP into AVs. In summary, we unveil a novel role of GULP1 in enhancing the targeting of ATG14 to the ER to stimulate autophagy and, consequently, APP processing.
Collapse
Affiliation(s)
- Dennis Dik-Long Chau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhicheng Yu
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wai Wa Ray Chan
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhai Yuqi
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Raymond Chuen Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Jacky Chi Ki Ngo
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ho Yin Edwin Chan
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
- Laboratory of Drosophila Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kwok-Fai Lau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
16
|
Yang Q, Yang C, Lv H, Zheng X, Mao S, Liu N, Mo S, Liao B, Yang M, Lu Z, Tang L, Huang X, Jian C, Shang J. Autophagy Regulation Attenuates Neuroinflammation and Cognitive Decline in an Alzheimer's Disease Mouse Model with Chronic Cerebral Hypoperfusion. Inflammation 2024:10.1007/s10753-024-02043-0. [PMID: 38951357 DOI: 10.1007/s10753-024-02043-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 07/03/2024]
Abstract
This study investigates the role of autophagy regulation in modulating neuroinflammation and cognitive function in an Alzheimer's disease (AD) mouse model with chronic cerebral hypoperfusion (CCH). Using the APP23/PS1 mice plus CCH model, we examined the impact of autophagy regulation on cognitive function, neuroinflammation, and autophagic activity. Our results demonstrate significant cognitive impairments in AD mice, exacerbated by CCH, but mitigated by treatment with the autophagy inhibitor 3-methyladenine (3-MA). Dysregulation of autophagy-related proteins, accentuated by CCH, underscores the intricate relationship between cerebral blood flow and autophagy dysfunction in AD pathology. While 3-MA restored autophagic balance, rapamycin (RAPA) treatment did not induce significant changes, suggesting alternative therapeutic approaches are necessary. Dysregulated microglial polarization and neuroinflammation in AD+CCH were linked to cognitive decline, with 3-MA attenuating neuroinflammation. Furthermore, alterations in M2 microglial polarization and the levels of inflammatory markers NLRP3 and MCP1 were observed, with 3-MA treatment exhibiting potential anti-inflammatory effects. Our findings shed light on the crosstalk between autophagy and neuroinflammation in AD+CCH and suggest targeting autophagy as a promising strategy for mitigating neuroinflammation and cognitive decline in AD+CCH.
Collapse
Affiliation(s)
- Qin Yang
- Medical School, Jinan University, Guangzhou, Guangdong, China
- Department of Neurology, Baise People's Hospital, Baise, Guangxi, China
| | - Chengmin Yang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Hui Lv
- Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xingwu Zheng
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Sanyin Mao
- Department of Neurology, The First People's Hospital of Jiande, Hangzhou, China
| | - Ning Liu
- School of Basic Medical Sciences, Beihua University, Jilin, China
| | - Shenglong Mo
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Bao Liao
- Department of Neurology, Baise People's Hospital, Baise, Guangxi, China
| | - Meiling Yang
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Zhicheng Lu
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Lina Tang
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Xiaorui Huang
- Department of Psychiatry and Psychology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chongdong Jian
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| | - Jingwei Shang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| |
Collapse
|
17
|
Singh MK, Shin Y, Ju S, Han S, Kim SS, Kang I. Comprehensive Overview of Alzheimer's Disease: Etiological Insights and Degradation Strategies. Int J Mol Sci 2024; 25:6901. [PMID: 39000011 PMCID: PMC11241648 DOI: 10.3390/ijms25136901] [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: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and affects millions of individuals globally. AD is associated with cognitive decline and memory loss that worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to prevent and manage its debilitating effects. Amyloid-β and tau proteins are primarily associated with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on degrading amyloid-β and tau or inhibiting their synthesis, particularly targeting APP processing and tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly make a difference. Genome-wide association studies (GWASs) across various cohorts identified 40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to be understood about the functions of these genes and their role in the disease process, prompting continued investigation. By delving deeper into these genetic associations, novel targets such as kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in AD and identifies how genetic variations within these pathways could serve as potential targets for drug discovery and treatment strategies. Through a comprehensive understanding of the molecular underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate the burden of this devastating disease.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
18
|
Zhao Y, Sun B, Fu X, Zuo Z, Qin H, Yao K. YAP in development and disease: Navigating the regulatory landscape from retina to brain. Biomed Pharmacother 2024; 175:116703. [PMID: 38713948 DOI: 10.1016/j.biopha.2024.116703] [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: 01/17/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/09/2024] Open
Abstract
The distinctive role of Yes-associated protein (YAP) in the nervous system has attracted widespread attention. This comprehensive review strategically uses the retina as a vantage point, embarking on an extensive exploration of YAP's multifaceted impact from the retina to the brain in development and pathology. Initially, we explore the crucial roles of YAP in embryonic and cerebral development. Our focus then shifts to retinal development, examining in detail YAP's regulatory influence on the development of retinal pigment epithelium (RPE) and retinal progenitor cells (RPCs), and its significant effects on the hierarchical structure and functionality of the retina. We also investigate the essential contributions of YAP in maintaining retinal homeostasis, highlighting its precise regulation of retinal cell proliferation and survival. In terms of retinal-related diseases, we explore the epigenetic connections and pathophysiological regulation of YAP in diabetic retinopathy (DR), glaucoma, and proliferative vitreoretinopathy (PVR). Lastly, we broaden our exploration from the retina to the brain, emphasizing the research paradigm of "retina: a window to the brain." Special focus is given to the emerging studies on YAP in brain disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD), underlining its potential therapeutic value in neurodegenerative disorders and neuroinflammation.
Collapse
Affiliation(s)
- Yaqin Zhao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Bin Sun
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xuefei Fu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhuan Zuo
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
19
|
Kubat Oktem E. Biomarkers of Alzheimer's Disease Associated with Programmed Cell Death Reveal Four Repurposed Drugs. J Mol Neurosci 2024; 74:51. [PMID: 38700745 DOI: 10.1007/s12031-024-02228-0] [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: 12/29/2023] [Accepted: 04/21/2024] [Indexed: 07/20/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the most common cause of dementia. Programmed cell death (PCD) is mainly characterized by unique morphological features and energy-dependent biochemical processes. The predominant pathway leading to cell death in AD has not been thoroughly analyzed, although there is evidence of neuron loss in AD and numerous pathways of PCD have been associated with this process. A better understanding of the systems biology underlying the relationship between AD and PCD could lead to the development of new therapeutic approaches. To this end, publicly available transcriptome data were examined using bioinformatic methods such as differential gene expression and weighted gene coexpression network analysis (WGCNA) to find PCD-related AD biomarkers. The diagnostic significance of these biomarkers was evaluated using a logistic regression-based predictive model. Using these biomarkers, a multifactorial regulatory network was developed. Last, a drug repositioning study was conducted to propose new drugs for the treatment of AD targeting PCD. The development of 3PM (predictive, preventive, and personalized) drugs for the treatment of AD would be enabled by additional research on the effects of these drugs on this disease.
Collapse
Affiliation(s)
- Elif Kubat Oktem
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Medeniyet University, North Campus, Istanbul, 34700, Turkey.
| |
Collapse
|
20
|
Kiss E, Kins S, Gorgas K, Venczel Szakács KH, Kirsch J, Kuhse J. Another Use for a Proven Drug: Experimental Evidence for the Potential of Artemisinin and Its Derivatives to Treat Alzheimer's Disease. Int J Mol Sci 2024; 25:4165. [PMID: 38673751 PMCID: PMC11049906 DOI: 10.3390/ijms25084165] [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: 02/05/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Plant-derived multitarget compounds may represent a promising therapeutic strategy for multifactorial diseases, such as Alzheimer's disease (AD). Artemisinin and its derivatives were indicated to beneficially modulate various aspects of AD pathology in different AD animal models through the regulation of a wide range of different cellular processes, such as energy homeostasis, apoptosis, proliferation and inflammatory pathways. In this review, we aimed to provide an up-to-date overview of the experimental evidence documenting the neuroprotective activities of artemi-sinins to underscore the potential of these already-approved drugs for treating AD also in humans and propose their consideration for carefully designed clinical trials. In particular, the benefits to the main pathological hallmarks and events in the pathological cascade throughout AD development in different animal models of AD are summarized. Moreover, dose- and context-dependent effects of artemisinins are noted.
Collapse
Affiliation(s)
- Eva Kiss
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
- Department of Cellular and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania;
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, 69120 Kaiserslautern, Germany;
| | - Karin Gorgas
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
| | - Kinga Hajnal Venczel Szakács
- Department of Cellular and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania;
| | - Joachim Kirsch
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
| | - Jochen Kuhse
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
| |
Collapse
|
21
|
Ma X, Shyer M, Harris K, Wang D, Hsu YC, Farrell C, Goodwin N, Anjum S, Bukhbinder AS, Dean S, Khan T, Hunter D, Schulz PE, Jiang X, Kim Y. Deep learning to predict rapid progression of Alzheimer's disease from pooled clinical trials: A retrospective study. PLOS DIGITAL HEALTH 2024; 3:e0000479. [PMID: 38598464 PMCID: PMC11006164 DOI: 10.1371/journal.pdig.0000479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024]
Abstract
The rate of progression of Alzheimer's disease (AD) differs dramatically between patients. Identifying the most is critical because when their numbers differ between treated and control groups, it distorts the outcome, making it impossible to tell whether the treatment was beneficial. Much recent effort, then, has gone into identifying RPs. We pooled de-identified placebo-arm data of three randomized controlled trials (RCTs), EXPEDITION, EXPEDITION 2, and EXPEDITION 3, provided by Eli Lilly and Company. After processing, the data included 1603 mild-to-moderate AD patients with 80 weeks of longitudinal observations on neurocognitive health, brain volumes, and amyloid-beta (Aβ) levels. RPs were defined by changes in four neurocognitive/functional health measures. We built deep learning models using recurrent neural networks with attention mechanisms to predict RPs by week 80 based on varying observation periods from baseline (e.g., 12, 28 weeks). Feature importance scores for RP prediction were computed and temporal feature trajectories were compared between RPs and non-RPs. Our evaluation and analysis focused on models trained with 28 weeks of observation. The models achieved robust internal validation area under the receiver operating characteristic (AUROCs) ranging from 0.80 (95% CI 0.79-0.82) to 0.82 (0.81-0.83), and the area under the precision-recall curve (AUPRCs) from 0.34 (0.32-0.36) to 0.46 (0.44-0.49). External validation AUROCs ranged from 0.75 (0.70-0.81) to 0.83 (0.82-0.84) and AUPRCs from 0.27 (0.25-0.29) to 0.45 (0.43-0.48). Aβ plasma levels, regional brain volumetry, and neurocognitive health emerged as important factors for the model prediction. In addition, the trajectories were stratified between predicted RPs and non-RPs based on factors such as ventricular volumes and neurocognitive domains. Our findings will greatly aid clinical trialists in designing tests for new medications, representing a key step toward identifying effective new AD therapies.
Collapse
Affiliation(s)
- Xiaotian Ma
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Madison Shyer
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Kristofer Harris
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Dulin Wang
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Yu-Chun Hsu
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Christine Farrell
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Nathan Goodwin
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Sahar Anjum
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Avram S. Bukhbinder
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Division of Pediatric Neurology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Sarah Dean
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Tanveer Khan
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - David Hunter
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Paul E. Schulz
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Xiaoqian Jiang
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Yejin Kim
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
22
|
Bahnassawy L, Nicolaisen N, Untucht C, Mielich-Süss B, Reinhardt L, Ried JS, Morawe MP, Geist D, Finck A, Käfer E, Korffmann J, Townsend M, Ravikumar B, Lakics V, Cik M, Reinhardt P. Establishment of a high-content imaging assay for tau aggregation in hiPSC-derived neurons differentiated from two protocols to routinely evaluate compounds and genetic perturbations. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100137. [PMID: 38128829 DOI: 10.1016/j.slasd.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Aberrant protein aggregation is a pathological cellular hallmark of many neurodegenerative diseases, such as Alzheimer's disease (AD) and frontotemporal dementia (FTD), where the tau protein is aggregating, forming neurofibrillary tangles (NFTs), and propagating from neuron to neuron. These processes have been linked to disease progression and a decline in cognitive function. Various therapeutic approaches aim at the prevention or reduction of tau aggregates in neurons. Human induced pluripotent stem cells (hiPSCs) are a very valuable tool in neuroscience discovery, as they offer access to potentially unlimited amounts of cell types that are affected in disease, including cortical neurons, for in vitro studies. We have generated an in vitro model for tau aggregation that uses hiPSC - derived neurons expressing an aggregation prone, fluorescently tagged version of the human tau protein after lentiviral transduction. Upon addition of tau seeds in the form of recombinant sonicated paired helical filaments (sPHFs), the neurons show robust, disease-like aggregation of the tau protein. The model was developed as a plate-based high content screening assay coupled with an image analysis algorithm to evaluate the impact of small molecules or genetic perturbations on tau. We show that the assay can be used to evaluate small molecules or screen targeted compound libraries. Using siRNA-based gene knockdown, genes of interest can be evaluated, and we could show that a targeted gene library can be screened, by screening nearly 100 deubiquitinating enzymes (DUBs) in that assay. The assay uses an imaging-based readout, a relatively short timeline, quantifies the extent of tau aggregation, and also allows the assessment of cell viability. Furthermore, it can be easily adapted to different hiPSC lines or neuronal subtypes. Taken together, this complex and highly relevant approach can be routinely applied on a weekly basis in the screening funnels of several projects and generates data with a turnaround time of approximately five weeks.
Collapse
Affiliation(s)
- Lamiaa Bahnassawy
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Nathalie Nicolaisen
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Christopher Untucht
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Benjamin Mielich-Süss
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Lydia Reinhardt
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Janina S Ried
- Genomics Research Center, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Martina P Morawe
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Daniela Geist
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Anja Finck
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Elke Käfer
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Jürgen Korffmann
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Matthew Townsend
- Cambridge Research Center, AbbVie Inc., 200 Sidney Street, Cambridge, MA 02139, USA
| | - Brinda Ravikumar
- Cambridge Research Center, AbbVie Inc., 200 Sidney Street, Cambridge, MA 02139, USA
| | - Viktor Lakics
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Miroslav Cik
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany.
| | - Peter Reinhardt
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co KG, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany.
| |
Collapse
|
23
|
Liu Z, Cecarini V, Cuccioloni M, Bonfili L, Gong C, Angeletti M, Eleuteri AM. Ginsenosides Rg1 and Rg2 Activate Autophagy and Attenuate Oxidative Stress in Neuroblastoma Cells Overexpressing Aβ(1-42). Antioxidants (Basel) 2024; 13:310. [PMID: 38539843 PMCID: PMC10967604 DOI: 10.3390/antiox13030310] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 01/04/2025] Open
Abstract
Alzheimer's disease is a neurodegeneration with protein deposits, altered proteolysis, and inflammatory and oxidative processes as major hallmarks. Despite the continuous search for potential therapeutic treatments, no cure is available to date. The use of natural molecules as adjuvants in the treatment of Alzheimer's disease is a very promising strategy. In this regard, ginsenosides from ginseng root show a variety of biological effects. Here, we dissected the role of ginsenosides Rg1 and Rg2 in modulating autophagy and oxidative stress in neuroblastoma cells overexpressing Aβ(1-42). Key hallmarks of these cellular processes were detected through immunomethods and fluorometric assays. Our findings indicate that ginsenosides are able to upregulate autophagy in neuronal cells as demonstrated by increased levels of LC3II and Beclin-1 proteins and decreased amounts of p62. Simultaneously, an activation of lysosomal hydrolases was observed. Furthermore, autophagy activation promoted the clearance of Aβ(1-42). Rg1 and Rg2 also reduced oxidative stress sources and macromolecule oxidation, promoting NRF2 nuclear translocation and the expression of antioxidant enzymes. Our data further clarify the mechanisms of action of Rg1 and Rg2, indicating new insights into their role in the management of disorders like Alzheimer's disease.
Collapse
Affiliation(s)
- Ziqi Liu
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy; (Z.L.); (M.C.); (L.B.); (C.G.); (M.A.); (A.M.E.)
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy; (Z.L.); (M.C.); (L.B.); (C.G.); (M.A.); (A.M.E.)
| | - Massimiliano Cuccioloni
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy; (Z.L.); (M.C.); (L.B.); (C.G.); (M.A.); (A.M.E.)
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy; (Z.L.); (M.C.); (L.B.); (C.G.); (M.A.); (A.M.E.)
| | - Chunmei Gong
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy; (Z.L.); (M.C.); (L.B.); (C.G.); (M.A.); (A.M.E.)
| | - Mauro Angeletti
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy; (Z.L.); (M.C.); (L.B.); (C.G.); (M.A.); (A.M.E.)
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy; (Z.L.); (M.C.); (L.B.); (C.G.); (M.A.); (A.M.E.)
| |
Collapse
|
24
|
Griñán-Ferré C, Jarne-Ferrer J, Bellver-Sanchis A, Ribalta-Vilella M, Barroso E, Salvador JM, Jurado-Aguilar J, Palomer X, Vázquez-Carrera M, Pallàs M. Deletion of Gadd45a Expression in Mice Leads to Cognitive and Synaptic Impairment Associated with Alzheimer's Disease Hallmarks. Int J Mol Sci 2024; 25:2595. [PMID: 38473843 DOI: 10.3390/ijms25052595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/10/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
Gadd45 genes have been implicated in survival mechanisms, including apoptosis, autophagy, cell cycle arrest, and DNA repair, which are processes related to aging and life span. Here, we analyzed if the deletion of Gadd45a activates pathways involved in neurodegenerative disorders such as Alzheimer's Disease (AD). This study used wild-type (WT) and Gadd45a knockout (Gadd45a-/-) mice to evaluate AD progression. Behavioral tests showed that Gadd45a-/- mice presented lower working and spatial memory, pointing out an apparent cognitive impairment compared with WT animals, accompanied by an increase in Tau hyperphosphorylation and the levels of kinases involved in its phosphorylation in the hippocampus. Moreover, Gadd45a-/- animals significantly increased the brain's pro-inflammatory cytokines and modified autophagy markers. Notably, neurotrophins and the dendritic spine length of the neurons were reduced in Gadd45a-/- mice, which could contribute to the cognitive alterations observed in these animals. Overall, these findings demonstrate that the lack of the Gadd45a gene activates several pathways that exacerbate AD pathology, suggesting that promoting this protein's expression or function might be a promising therapeutic strategy to slow down AD progression.
Collapse
Affiliation(s)
- Christian Griñán-Ferré
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Júlia Jarne-Ferrer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
| | - Aina Bellver-Sanchis
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
| | - Marta Ribalta-Vilella
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-National Institute of Health Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Jesús M Salvador
- Department of Immunology and Oncology, National Center for Biotechnology/CSIC, 28049 Madrid, Spain
| | - Javier Jurado-Aguilar
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-National Institute of Health Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-National Institute of Health Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-National Institute of Health Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-National Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
25
|
Naeem A, Prakash R, Kumari N, Ali Khan M, Quaiyoom Khan A, Uddin S, Verma S, Ab Robertson A, Boltze J, Shadab Raza S. MCC950 reduces autophagy and improves cognitive function by inhibiting NLRP3-dependent neuroinflammation in a rat model of Alzheimer's disease. Brain Behav Immun 2024; 116:70-84. [PMID: 38040385 DOI: 10.1016/j.bbi.2023.11.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/11/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023] Open
Abstract
Alzheimer's disease (AD) is the seventh most common cause of mortality and one of the major causes of disability and vulnerability in the elderly. AD is characterized by gradual cognitive deterioration, the buildup of misfolded amyloid beta (Aβ) peptide, and the generation of neurofibrillary tangles. Despite enormous scientific progress, there is no effective cure for AD. Thus, exploring new treatment options to stop AD or at least slow down its progress is important. In this study, we investigated the potential therapeutic effects of MCC950 on NLRP3-mediated inflammasome-driven inflammation and autophagy in AD. Rats treated with streptozotocin (STZ) exhibited simultaneous activation of the NLRP3 inflammasome and autophagy, as confirmed by Western blot, immunofluorescence, and co-immunoprecipitation analyses. MCC950, a specific NLRP3 inhibitor, was intraperitoneally administered (50 mg/kg body weight) to rats with AD-like symptoms induced by intracerebroventricular STZ injections (3 mg/kg body weight). MCC950 effectively suppressed STZ-induced cognitive impairment and anxiety by inhibiting NLRP3-dependent neuroinflammation. Moreover, our findings indicate that MCC950 exerts neuroprotective effects by attenuating autophagy in neuronal cells. The inhibiting effects of MCC950 on inflammasome activation and autophagy were reproduced in vitro, provding further mechansistic insights into MCC950 therapeutic action. Our findings suggest that MCC950 impedes the progression of AD and may also improve cognitive function through the mitigation of autophagy and NLRP3 inflammasome inhibition.
Collapse
Affiliation(s)
- Abdul Naeem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India
| | - Ravi Prakash
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India
| | - Neha Kumari
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India
| | | | - Abdul Quaiyoom Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Sandeep Verma
- Department of Chemistry, Indian Institute of Technology, Kanpur, UP 208016, India
| | - Avril Ab Robertson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India; Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India.
| |
Collapse
|
26
|
Srivastav S, van der Graaf K, Jonnalagadda PC, Thawani M, McNew JA, Stern M. Motor neuron activity enhances the proteomic stress caused by autophagy defects in the target muscle. PLoS One 2024; 19:e0291477. [PMID: 38166124 PMCID: PMC10760831 DOI: 10.1371/journal.pone.0291477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/12/2023] [Indexed: 01/04/2024] Open
Abstract
Several lines of evidence demonstrate that increased neuronal excitability can enhance proteomic stress. For example, epilepsy can enhance the proteomic stress caused by the expression of certain aggregation-prone proteins implicated in neurodegeneration. However, unanswered questions remain concerning the mechanisms by which increased neuronal excitability accomplishes this enhancement. Here we test whether increasing neuronal excitability at a particular identified glutamatergic synapse, the Drosophila larval neuromuscular junction, can enhance the proteomic stress caused by mutations in the ER fusion/GTPase gene atlastin (atl). It was previously shown that larval muscle from the atl2 null mutant is defective in autophagy and accumulates protein aggregates containing ubiquitin (poly-UB aggregates). To determine if increased neuronal excitability might enhance the increased proteomic stress caused by atl2, we activated the TrpA1-encoded excitability channel within neurons. We found that TrpA1 activation had no effect on poly-UB aggregate accumulation in wildtype muscle, but significantly increased poly-UB aggregate number in atl2 muscle. Previous work has shown that atl loss from either neuron or muscle increases muscle poly-UB aggregate number. We found that neuronal TrpA1 activation enhanced poly-UB aggregate number when atl was removed from muscle, but not from neuron. Neuronal TrpA1 activation enhanced other phenotypes conferred by muscle atl loss, such as decreased pupal size and decreased viability. Taken together, these results indicate that the proteomic stress caused by muscle atl loss is enhanced by increasing neuronal excitability.
Collapse
Affiliation(s)
- Saurabh Srivastav
- Department of BioSciences, Rice University, Houston, TX, United States of America
| | - Kevin van der Graaf
- Department of BioSciences, Rice University, Houston, TX, United States of America
| | | | - Maanvi Thawani
- Department of BioSciences, Rice University, Houston, TX, United States of America
| | - James A. McNew
- Department of BioSciences, Rice University, Houston, TX, United States of America
| | - Michael Stern
- Department of BioSciences, Rice University, Houston, TX, United States of America
| |
Collapse
|
27
|
Srivastav S, van der Graaf K, Singh P, Utama AB, Meyer MD, McNew JA, Stern M. Atl (atlastin) regulates mTor signaling and autophagy in Drosophila muscle through alteration of the lysosomal network. Autophagy 2024; 20:131-150. [PMID: 37649246 PMCID: PMC10761077 DOI: 10.1080/15548627.2023.2249794] [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: 03/06/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023] Open
Abstract
ABBREVIATIONS atl atlastin; ALR autophagic lysosome reformation; ER endoplasmic reticulum; GFP green fluorescent protein; HSP hereditary spastic paraplegia; Lamp1 lysosomal associated membrane protein 1 PolyUB polyubiquitin; RFP red fluorescent protein; spin spinster; mTor mechanistic Target of rapamycin; VCP valosin containing protein.
Collapse
Affiliation(s)
| | | | - Pratibha Singh
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | | | - Matthew D. Meyer
- Shared Equipment Authority, Rice University, Houston, Texas, USA
| | - James A. McNew
- Department of BioSciences, Rice University, Houston, Texas, USA
| | - Michael Stern
- Department of BioSciences, Rice University, Houston, Texas, USA
| |
Collapse
|
28
|
Nayak V, Patra S, Rout S, Jena AB, Sharma R, Pattanaik KP, Singh J, Pandey SS, Singh RP, Majhi S, Singh KR, Kerry RG. Regulation of neuroinflammation in Alzheimer's disease via nanoparticle-loaded phytocompounds with anti-inflammatory and autophagy-inducing properties. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155150. [PMID: 37944239 DOI: 10.1016/j.phymed.2023.155150] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/23/2023] [Accepted: 10/14/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by neuroinflammation linked to amyloid β (Aβ) aggregation and phosphorylated tau (τ) protein in neurofibrillary tangles (NFTs). Key elements in Aβ production and NFT assembly, like γ-secretase and p38 mitogen-activated protein kinase (p38MAPK), contribute to neuroinflammation. In addition, impaired proteosomal and autophagic pathways increase Aβ and τ aggregation, leading to neuronal damage. Conventional neuroinflammation drugs have limitations due to unidirectional therapeutic approaches and challenges in crossing the Blood-Brain Barrier (BBB). Clinical trials for non-steroidal anti-inflammatory drugs (NSAIDs) and other therapeutics remain uncertain. Novel strategies addressing the complex pathogenesis and BBB translocation are needed to effectively tackle AD-related neuroinflammation. PURPOSE The current scenario demands for a much-sophisticated theranostic measures which could be achieved via customized engineering and designing of novel nanotherapeutics. As, these therapeutics functions as a double edge sword, having the efficiency of unambiguous targeting, multiple drug delivery and ability to cross BBB proficiently. METHODS Inclusion criteria involve selecting recent, English-language studies from the past decade (2013-2023) that explore the regulation of neuroinflammation in neuroinflammation, Alzheimer's disease, amyloid β, tau protein, nanoparticles, autophagy, and phytocompounds. Various study types, including clinical trials, experiments, and reviews, were considered. Exclusion criteria comprised non-relevant publication types, studies unrelated to Alzheimer's disease or phytocompounds, those with methodological flaws, duplicates, and studies with inaccessible data. RESULTS In this study, polymeric nanoparticles loaded with specific phytocompounds and coated with an antibody targeting the transferrin receptor (anti-TfR) present on BBB. Thereafter, the engineered nanoparticles with the ability to efficiently traverse the BBB and interact with target molecules within the brain, could induce autophagy, a cellular process crucial for neuronal health, and exhibit potent anti-inflammatory effects. Henceforth, the proposed combination of desired phytocompounds, polymeric nanoparticles, and anti-TfR coating presents a promising approach for targeted drug delivery to the brain, with potential implications in neuroinflammatory conditions such as Alzheimer's disease.
Collapse
Affiliation(s)
- Vinayak Nayak
- ICAR- National Institute on Foot and Mouth Disease-International Centre for Foot and Mouth Disease, Arugul, Bhubaneswar, Odisha (752050), India
| | - Sushmita Patra
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra (410210), India
| | - Shrushti Rout
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar, Odisha (751004), India
| | - Atala Bihari Jena
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (02115), United States of America
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh (221005), India
| | - Kali Prasad Pattanaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar 751024, India
| | - Jay Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh (221005), India
| | - Shyam S Pandey
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu (8080196), Japan
| | - Ravindra Pratap Singh
- Department of Biotechnology, Faculty of Science, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh 484887, India
| | - Sanatan Majhi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (02115), United States of America
| | - Kshitij Rb Singh
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu (8080196), Japan.
| | - Rout George Kerry
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar, Odisha (751004), India.
| |
Collapse
|
29
|
Zainuddin MS, Bhuvanendran S, Radhakrishnan AK, Azman AS. Alzheimer's Disease-Related Proteins Targeted by Secondary Metabolite Compounds from Streptomyces: A Scoping Review. J Alzheimers Dis Rep 2023; 7:1335-1350. [PMID: 38143777 PMCID: PMC10741902 DOI: 10.3233/adr-230065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/16/2023] [Indexed: 12/26/2023] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease that is characterized as rapid and progressive cognitive decline affecting 26 million people worldwide. Although immunotherapies are ideal, its clinical safety and effectiveness are controversial, hence, treatments are still reliant on symptomatic medications. Concurrently, the Streptomyces genus has attracted attention given its pharmaceutically beneficial secondary metabolites to treat neurodegenerative diseases. Objective To present secondary metabolites from Streptomyces sp. with regulatory effects on proteins and identified prospective target proteins for AD treatment. Methods Research articles published between 2010 and 2021 were collected from five databases and 83 relevant research articles were identified. Post-screening, only 12 research articles on AD-related proteins were selected for further review. Bioinformatics analyses were performed through the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) network, PANTHER Go-Slim classification system (PANTHER17.0), and Kyoto Encyclopedia of Genes and Genomes (KEGG) Mapper. Results A total of 20 target proteins were identified from the 12 shortlisted articles. Amyloid-β, BACE1, Nrf-2, Beclin-1, and ATG5 were identified as the potential target proteins, given their role in initiating AD, mitigating neuroinflammation, and autophagy. Besides, 10 compounds from Streptomyces sp., including rapamycin, alborixin, enterocin, bonnevillamides D and E, caniferolide A, anhydroexfoliamycin, rhizolutin, streptocyclinone A and B, were identified to exhibit considerable regulatory effects on these target proteins. Conclusions The review highlights several prospective target proteins that can be regulated through treatments with Streptomyces sp. compounds to prevent AD's early stages and progression. Further identification of Streptomyces sp. compounds with potential anti-AD properties is recommended.
Collapse
Affiliation(s)
| | | | - Ammu K. Radhakrishnan
- Jeffery Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | | |
Collapse
|
30
|
Datta D, Perone I, Morozov YM, Arellano J, Duque A, Rakic P, van Dyck CH, Arnsten AFT. Localization of PDE4D, HCN1 channels, and mGluR3 in rhesus macaque entorhinal cortex may confer vulnerability in Alzheimer's disease. Cereb Cortex 2023; 33:11501-11516. [PMID: 37874022 PMCID: PMC10724870 DOI: 10.1093/cercor/bhad382] [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: 05/01/2023] [Revised: 08/28/2023] [Accepted: 09/27/2023] [Indexed: 10/25/2023] Open
Abstract
Alzheimer's disease cortical tau pathology initiates in the layer II cell clusters of entorhinal cortex, but it is not known why these specific neurons are so vulnerable. Aging macaques exhibit the same qualitative pattern of tau pathology as humans, including initial pathology in layer II entorhinal cortex clusters, and thus can inform etiological factors driving selective vulnerability. Macaque data have already shown that susceptible neurons in dorsolateral prefrontal cortex express a "signature of flexibility" near glutamate synapses on spines, where cAMP-PKA magnification of calcium signaling opens nearby potassium and hyperpolarization-activated cyclic nucleotide-gated channels to dynamically alter synapse strength. This process is regulated by PDE4A/D, mGluR3, and calbindin, to prevent toxic calcium actions; regulatory actions that are lost with age/inflammation, leading to tau phosphorylation. The current study examined whether a similar "signature of flexibility" expresses in layer II entorhinal cortex, investigating the localization of PDE4D, mGluR3, and HCN1 channels. Results showed a similar pattern to dorsolateral prefrontal cortex, with PDE4D and mGluR3 positioned to regulate internal calcium release near glutamate synapses, and HCN1 channels concentrated on spines. As layer II entorhinal cortex stellate cells do not express calbindin, even when young, they may be particularly vulnerable to magnified calcium actions and ensuing tau pathology.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Isabella Perone
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yury M Morozov
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jon Arellano
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Alvaro Duque
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Pasko Rakic
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | - Amy F T Arnsten
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
31
|
Tumurbaatar B, Fracassi A, Scaduto P, Guptarak J, Woltjer R, Jupiter D, Taglialatela G. Preserved autophagy in cognitively intact non-demented individuals with Alzheimer's neuropathology. Alzheimers Dement 2023; 19:5355-5370. [PMID: 37191183 PMCID: PMC10651802 DOI: 10.1002/alz.13074] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Growing evidence supports that dysfunctional autophagy, the major cell mechanism responsible for removing protein aggregates and a route of clearance for Tau in healthy neurons, is a major finding in demented Alzheimer's disease (AD) patients. However, the association of autophagy with maintenance of cognitive integrity in resilient individuals who have AD neuropathology but remain non-demented (NDAN) has not been evaluated. METHODS Using post mortem brain samples from age-matched healthy control, AD, and NDAN subjects, we evaluated autophagy in relation to Tau pathology using Western blot, immunofluorescence and RNA-seq. RESULTS Compared to AD patients, NDAN subjects had preserved autophagy and reduced tauopathy. Furthermore, expression of autophagy genes and AD-related proteins were significantly associated in NDAN compared to AD and control subjects. DISCUSSION Our results suggest preserved autophagy is a protective mechanism that maintains cognitive integrity in NDAN individuals. This novel observation supports the potential of autophagy-inducing strategies in AD therapeutics. HIGHLIGHTS NDAN subjects have preserved autophagic protein levels comparable with control subjects. Compared to control subjects, NDAN subjects have significantly reduced Tau oligomers and PHF Tau phosphorylation at synapses that negatively correlate with autophagy markers. Transcription of autophagy genes strongly associates with AD-related proteins in NDAN donors.
Collapse
Affiliation(s)
- Batbayar Tumurbaatar
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Pietro Scaduto
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Jutatip Guptarak
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Randall Woltjer
- Department of Pathology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, USA
| | - Daniel Jupiter
- Department of Biostatistics & Data Science, Department of Orthopaedic Surgery and Rehabilitation, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), 301 University Blvd, Galveston, Texas, 77555 USA
| |
Collapse
|
32
|
Baek H, Sanjay, Park M, Lee HJ. Cyanidin-3-O-glucoside protects the brain and improves cognitive function in APPswe/PS1ΔE9 transgenic mice model. J Neuroinflammation 2023; 20:268. [PMID: 37978414 PMCID: PMC10655395 DOI: 10.1186/s12974-023-02950-3] [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: 07/04/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
Cyanidin-3-O-glucoside (C3G) is a natural anthocyanin with antioxidant, anti-inflammatory, and antitumor properties. However, as the effects of C3G on the amyloidogenic pathway, autophagy, tau phosphorylation, neuronal cell death, and synaptic plasticity in Alzheimer's disease models have not been reported, we attempted to investigate the same in the brains of APPswe/PS1ΔE9 mice were analyzed. After oral administration of C3G (30 mg/kg/day) for 16 weeks, the cortical and hippocampal regions in the brains of APPswe/PS1ΔE9 mice were analyzed. C3G treatment reduced the levels of soluble and insoluble Aβ (Aβ40 and Aβ42) peptides and reduced the protein expression of the amyloid precursor protein, presenilin-1, and β-secretase in the cortical and hippocampal regions. And C3G treatment upregulated the expression of autophagy-related markers, LC3B-II, LAMP-1, TFEB, and PPAR-α and downregulated that of SQSTM1/p62, improving the autophagy of Aβ plaques and neurofibrillary tangles. In addition, C3G increased the protein expression of phosphorylated-AMPK/AMPK and Sirtuin 1 and decreased that of mitogen-activated protein kinases, such as phosphorylated-Akt/Akt and phosphorylated-ERK/ERK, thus demonstrating its neuroprotective effects. Furthermore, C3G regulated the PI3K/Akt/GSK3β signaling by upregulating phosphorylated-Akt/Akt and phosphorylated-GSK3β/GSK3β expression. C3G administration mitigated tau phosphorylation and improved synaptic function and plasticity by upregulating the expression of synapse-associated proteins synaptophysin and postsynaptic density protein-95. Although the potential of C3G in the APPswe/PS1ΔE9 mouse models has not yet been reported, oral administration of the C3G is shown to protect the brain and improve cognitive behavior.
Collapse
Affiliation(s)
- Hana Baek
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Sanjay
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Miey Park
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea.
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea.
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea.
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea.
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.
| |
Collapse
|
33
|
Barmaki H, Nourazarian A, Khaki-Khatibi F. Proteostasis and neurodegeneration: a closer look at autophagy in Alzheimer's disease. Front Aging Neurosci 2023; 15:1281338. [PMID: 38020769 PMCID: PMC10652403 DOI: 10.3389/fnagi.2023.1281338] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of misfolded amyloid-beta and tau proteins. Autophagy acts as a proteostasis process to remove protein clumps, although it progressively weakens with aging and AD, thus facilitating the accumulation of toxic proteins and causing neurodegeneration. This review examines the impact of impaired autophagy on the progression of AD disease pathology. Under normal circumstances, autophagy removes abnormal proteins and damaged organelles, but any dysfunction in this process can lead to the exacerbation of amyloid and tau pathology, particularly in AD. There is increasing attention to therapeutic tactics to revitalize autophagy, including reduced caloric intake, autophagy-stimulating drugs, and genetic therapy. However, the translation of these strategies into clinical practice faces several hurdles. In summary, this review integrates the understanding of the intricate role of autophagy dysfunction in Alzheimer's disease progression and reinforces the promising prospects of autophagy as a beneficial target for treatments to modify the course of Alzheimer's disease.
Collapse
Affiliation(s)
- Haleh Barmaki
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Fatemeh Khaki-Khatibi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Medina-Vera D, Zambrana-Infantes EN, López-Gambero AJ, Verheul-Campos J, Santín LJ, Baixeras E, Suarez J, Pavon FJ, Rosell-Valle C, de Fonseca FR. Transcending the amyloid-beta dominance paradigm in Alzheimer's disease: An exploration of behavioural, metabolic, and gut microbiota phenotypes in 5xFAD mice. Neurobiol Dis 2023; 187:106295. [PMID: 37717663 DOI: 10.1016/j.nbd.2023.106295] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023] Open
Abstract
The amyloid cascade hypothesis is widely accepted as an explanation for the neuropathological changes in Alzheimer's disease (AD). However, the role of amyloid-beta (Aβ) as the sole cause of these changes is being questioned. Using the 5xFAD mouse model of AD, we investigated various factors contributing to neuropathology, including genetic load (heterozygous (HTZ) versus homozygous (HZ) condition), behavioural phenotype, neuropathology markers, metabolic physiology, and gut microbiota composition at early (5 months of age) and late (12 months of age) stages of disease onset, and considering both sexes. At 5 months of age, both HTZ and HZ mice exhibited hippocampal alterations associated with Aβ accumulation, leading to increased neuroinflammation and disrupted PI3K-Akt pathway. However, only HZ mice showed cognitive impairment in the Y-maze and Morris water maze tests, worsening with age. Dysregulation of both insulin and insulin secretion-regulating GIP peptide were observed at 5 months of age, disappearing later. Circulating levels of metabolic-regulating hormones, such as Ghrelin and resisting helped to differentiates HTZ mice from HZ mice. Differences between HTZ and HZ mice were also observed in gut microbiota composition, disrupted intestinal barrier proteins, and increased proinflammatory products in the intestine. These findings suggest that cognitive impairment in 5xFAD mice may not solely result from Aβ aggregation. Other factors, including altered PI3K-Akt signalling, disrupted insulin-linked metabolic pathways, and changes in gut microbiota, contribute to disease progression. Targeting Aβ deposition alone may not suffice. Understanding AD pathogenesis and its multiple contributing factors is vital for effective therapies.
Collapse
Affiliation(s)
- Dina Medina-Vera
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; Facultad de Ciencias, Campus de Teatinos s/n, Universidad de Málaga, 29010 Málaga, Spain; Facultad de Medicina, Campus de Teatinos s/n, Universidad de Málaga, 29010 Málaga, Spain; Unidad de Gestión Clínica del Corazón-CIBERCV (Enfermedades Cardiovasculares), Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Emma N Zambrana-Infantes
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain
| | - Antonio J López-Gambero
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000 Bordeaux, France
| | - Julia Verheul-Campos
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Luis J Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain
| | - Elena Baixeras
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Juan Suarez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Francisco J Pavon
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; Unidad de Gestión Clínica del Corazón-CIBERCV (Enfermedades Cardiovasculares), Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Cristina Rosell-Valle
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain.
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain.
| |
Collapse
|
35
|
Abd Elmaaboud MA, Estfanous RS, Atef A, Kabel AM, Alnemari KA, Naguib TM, Alsufyani SE, Darwish HW, Arab HH. Dapagliflozin/Hesperidin Combination Mitigates Lipopolysaccharide-Induced Alzheimer's Disease in Rats. Pharmaceuticals (Basel) 2023; 16:1370. [PMID: 37895841 PMCID: PMC10609711 DOI: 10.3390/ph16101370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/19/2023] [Accepted: 09/24/2023] [Indexed: 10/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative disorders worldwide. Its pathologic features include massive neuroinflammation with abnormal deposition of β-amyloid peptide in the cerebral tissues leading to degeneration of the brain neurons. Adverse effects associated with the traditional drugs used for the treatment of this pathological condition have directed the research efforts towards searching for alternative effective agents with minimal adverse effects. The aim of this study was to elucidate the potential ameliorative effects of dapagliflozin and/or hesperidin on Alzheimer's disease (AD) induced by lipopolysaccharide (LPS) injection in rats. In a rodent model of AD, the effect of dapagliflozin with or without hesperidin on the biochemical parameters and the behavioral tests as well as the histopathological parameters was determined. Each of dapagliflozin and hesperidin restored the behavioral tests to the reference values, augmented the antioxidant defense mechanisms, ameliorated the neuronal inflammatory responses, combatted the changes in Toll-like receptor-4 (TLR-4)/High-mobility group box 1 (HMGB1) protein signaling and receptors of advanced glycation end products (RAGE) levels, and restored the balance between the apoptotic signals and autophagy in the hippocampal tissues. Additionally, both agents exhibited an outstanding ability to combat LPS-induced perturbations in the histopathological and electron microscopic image of the brain tissues. These favorable effects were significantly encountered in the group treated with dapagliflozin/hesperidin combination when compared versus animals treated with either dapagliflozin or hesperidin. In conclusion, inhibition of the hippocampal HMGB1/TLR4/RAGE signaling, the pro-inflammatory axis, and apoptosis alongside augmentation of the antioxidant defenses and autophagy can be regarded as beneficial effects by which dapagliflozin/hesperidin combination may combat LPS-triggered AD.
Collapse
Affiliation(s)
| | - Remon S. Estfanous
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Aliaa Atef
- Department of Pathology, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Ahmed M. Kabel
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | | | - Tamer M. Naguib
- Anesthesia and ICU Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Shuruq E. Alsufyani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (S.E.A.); or (H.H.A.)
| | - Hany W. Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Hany H. Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (S.E.A.); or (H.H.A.)
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
36
|
Funayama N, Yagyuu T, Imada M, Ueyama Y, Nakagawa Y, Kirita T. Impact of beta-tricalcium phosphate on preventing tooth extraction-triggered bisphosphonate-related osteonecrosis of the jaw in rats. Sci Rep 2023; 13:16032. [PMID: 37749392 PMCID: PMC10520003 DOI: 10.1038/s41598-023-43315-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023] Open
Abstract
Antiresorptive or antiangiogenic drugs can cause medication-related osteonecrosis of the jaw that is refractory. Bisphosphonate-related osteonecrosis of the jaw (BRONJ) may be caused by procedures such as tooth extraction damage the alveolar bone, release bisphosphonates (BPs) and impede healing. This study investigated strategies for BRONJ prevention and molecular mechanisms of its onset. We assessed the effectiveness of filling extraction sockets with beta-tricalcium phosphate (β-TCP). Rats were administered zoledronic acid (ZA) 1.2 mg/kg once per week for 2 weeks, and a molar was extracted. They were randomly assigned to the β-TCP group (bone defects filled with 0.01 g of β-TCP) or control group. Tissue content measurements indicated 2.2 ng of ZA per socket in the β-TCP group and 4.9 ng in the control group, confirming BP distribution and BP adsorption by β-TCP in vivo. At 4 weeks after extraction, the β-TCP group had normal mucosal coverage without inflammation. Moreover, at 8 weeks after extraction, enhanced bone healing, socket coverage, and new bone formation were observed in the β-TCP group. Connective tissue in the extraction sockets suggested that local increases in BP concentrations may suppress the local autophagy mechanisms involved in BRONJ. Filling extraction sockets with β-TCP may prevent BRONJ.
Collapse
Affiliation(s)
- Naoki Funayama
- Department of Oral and Maxillofacial Surgery, Nara Medical University, 840 Shijo-cho, Kashihara-shi, Nara, 634-8521, Japan
| | - Takahiro Yagyuu
- Department of Oral and Maxillofacial Surgery, Nara Medical University, 840 Shijo-cho, Kashihara-shi, Nara, 634-8521, Japan.
| | - Mitsuhiko Imada
- Department of Oral and Maxillofacial Surgery, Nara Medical University, 840 Shijo-cho, Kashihara-shi, Nara, 634-8521, Japan
| | - Yoshihiro Ueyama
- Department of Oral and Maxillofacial Surgery, Nara Medical University, 840 Shijo-cho, Kashihara-shi, Nara, 634-8521, Japan
| | - Yosuke Nakagawa
- Department of Oral and Maxillofacial Surgery, Nara Medical University, 840 Shijo-cho, Kashihara-shi, Nara, 634-8521, Japan
| | - Tadaaki Kirita
- Department of Oral and Maxillofacial Surgery, Nara Medical University, 840 Shijo-cho, Kashihara-shi, Nara, 634-8521, Japan
| |
Collapse
|
37
|
Sanghai N, Tranmer GK. Biochemical and Molecular Pathways in Neurodegenerative Diseases: An Integrated View. Cells 2023; 12:2318. [PMID: 37759540 PMCID: PMC10527779 DOI: 10.3390/cells12182318] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/05/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Neurodegenerative diseases (NDDs) like Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) are defined by a myriad of complex aetiologies. Understanding the common biochemical molecular pathologies among NDDs gives an opportunity to decipher the overlapping and numerous cross-talk mechanisms of neurodegeneration. Numerous interrelated pathways lead to the progression of neurodegeneration. We present evidence from the past pieces of literature for the most usual global convergent hallmarks like ageing, oxidative stress, excitotoxicity-induced calcium butterfly effect, defective proteostasis including chaperones, autophagy, mitophagy, and proteosome networks, and neuroinflammation. Herein, we applied a holistic approach to identify and represent the shared mechanism across NDDs. Further, we believe that this approach could be helpful in identifying key modulators across NDDs, with a particular focus on AD, PD, and ALS. Moreover, these concepts could be applied to the development and diagnosis of novel strategies for diverse NDDs.
Collapse
Affiliation(s)
- Nitesh Sanghai
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada;
| | - Geoffrey K. Tranmer
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada;
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
38
|
Bellettini-Santos T, Batista-Silva H, Marcolongo-Pereira C, Quintela-Castro FCDA, Barcelos RM, Chiepe KCMB, Rossoni JV, Passamani-Ambrosio R, da Silva BS, Chiarelli-Neto O, Garcez ML. Move Your Body toward Healthy Aging: Potential Neuroprotective Mechanisms of Irisin in Alzheimer's Disease. Int J Mol Sci 2023; 24:12440. [PMID: 37569815 PMCID: PMC10420140 DOI: 10.3390/ijms241512440] [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: 06/27/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in older adults, having a significant global burden and increasing prevalence. Current treatments for AD only provide symptomatic relief and do not cure the disease. Physical activity has been extensively studied as a potential preventive measure against cognitive decline and AD. Recent research has identified a hormone called irisin, which is produced during exercise, that has shown promising effects on cognitive function. Irisin acts on the brain by promoting neuroprotection by enhancing the growth and survival of neurons. It also plays a role in metabolism, energy regulation, and glucose homeostasis. Furthermore, irisin has been found to modulate autophagy, which is a cellular process involved in the clearance of protein aggregates, which are a hallmark of AD. Additionally, irisin has been shown to protect against cell death, apoptosis, oxidative stress, and neuroinflammation, all of which are implicated in AD pathogenesis. However, further research is needed to fully understand the mechanisms and therapeutic potential of irisin in AD. Despite the current gaps in knowledge, irisin holds promise as a potential therapeutic target for slowing cognitive decline and improving quality of life in AD patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Michelle Lima Garcez
- Graduate Program of Research and Extension (CEPEG), University Center of Espirito Santo, Espírito Santo 29703-858, Brazil; (T.B.-S.); (H.B.-S.); (C.M.-P.); (F.C.d.A.Q.-C.); (R.M.B.); (K.C.M.B.C.); (J.V.R.J.); (R.P.-A.); (B.S.d.S.); (O.C.-N.)
| |
Collapse
|
39
|
Zhang H, Bezprozvanny I. "Dirty Dancing" of Calcium and Autophagy in Alzheimer's Disease. Life (Basel) 2023; 13:life13051187. [PMID: 37240832 DOI: 10.3390/life13051187] [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: 03/24/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. There is a growing body of evidence that dysregulation in neuronal calcium (Ca2+) signaling plays a major role in the initiation of AD pathogenesis. In particular, it is well established that Ryanodine receptor (RyanR) expression levels are increased in AD neurons and Ca2+ release via RyanRs is augmented in AD neurons. Autophagy is important for removing unnecessary or dysfunctional components and long-lived protein aggregates, and autophagy impairment in AD neurons has been extensively reported. In this review we discuss recent results that suggest a causal link between intracellular Ca2+ signaling and lysosomal/autophagic dysregulation. These new results offer novel mechanistic insight into AD pathogenesis and may potentially lead to identification of novel therapeutic targets for treating AD and possibly other neurodegenerative disorders.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnical University, St. Petersburg 195251, Russia
| |
Collapse
|
40
|
Brase L, You SF, D'Oliveira Albanus R, Del-Aguila JL, Dai Y, Novotny BC, Soriano-Tarraga C, Dykstra T, Fernandez MV, Budde JP, Bergmann K, Morris JC, Bateman RJ, Perrin RJ, McDade E, Xiong C, Goate AM, Farlow M, Sutherland GT, Kipnis J, Karch CM, Benitez BA, Harari O. Single-nucleus RNA-sequencing of autosomal dominant Alzheimer disease and risk variant carriers. Nat Commun 2023; 14:2314. [PMID: 37085492 PMCID: PMC10121712 DOI: 10.1038/s41467-023-37437-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/15/2023] [Indexed: 04/23/2023] Open
Abstract
Genetic studies of Alzheimer disease (AD) have prioritized variants in genes related to the amyloid cascade, lipid metabolism, and neuroimmune modulation. However, the cell-specific effect of variants in these genes is not fully understood. Here, we perform single-nucleus RNA-sequencing (snRNA-seq) on nearly 300,000 nuclei from the parietal cortex of AD autosomal dominant (APP and PSEN1) and risk-modifying variant (APOE, TREM2 and MS4A) carriers. Within individual cell types, we capture genes commonly dysregulated across variant groups. However, specific transcriptional states are more prevalent within variant carriers. TREM2 oligodendrocytes show a dysregulated autophagy-lysosomal pathway, MS4A microglia have dysregulated complement cascade genes, and APOEε4 inhibitory neurons display signs of ferroptosis. All cell types have enriched states in autosomal dominant carriers. We leverage differential expression and single-nucleus ATAC-seq to map GWAS signals to effector cell types including the NCK2 signal to neurons in addition to the initially proposed microglia. Overall, our results provide insights into the transcriptional diversity resulting from AD genetic architecture and cellular heterogeneity. The data can be explored on the online browser ( http://web.hararilab.org/SNARE/ ).
Collapse
Affiliation(s)
- Logan Brase
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Shih-Feng You
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Ricardo D'Oliveira Albanus
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | | | - Yaoyi Dai
- Baylor College of Medicine, Houston, TX, USA
| | - Brenna C Novotny
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Carolina Soriano-Tarraga
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Taitea Dykstra
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Maria Victoria Fernandez
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John P Budde
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Kristy Bergmann
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John C Morris
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Randall J Bateman
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Richard J Perrin
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Eric McDade
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martin Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Greg T Sutherland
- School of Medical Sciences and Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Bruno A Benitez
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
- NeuroGenomics and Informatics, Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
41
|
Smit C, De Wet S, Barron T, Loos B. Rooibos tea-in the cross fire of ROS, mitochondrial dysfunction and loss of proteostasis-positioned for healthy aging. Biogerontology 2023; 24:149-162. [PMID: 36781516 DOI: 10.1007/s10522-022-10012-z] [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: 09/20/2022] [Accepted: 12/16/2022] [Indexed: 02/15/2023]
Abstract
Impaired mitochondrial function and loss of cellular proteostasis control are key hallmarks of aging and are implicated in the development of neurodegenerative diseases. A common denominator is the cell's inability to handle reactive oxygen species (ROS), leading to major downstream oxidative damage that exacerbates neuronal dysfunction. Although we have progressed in understanding the molecular defects associated with neuronal aging, many unanswered questions remain. How much ROS is required to serve cellular function before it becomes detrimental and how does the cell's oxidative status impact mitochondrial function and protein degradation through autophagy? How does ROS regulate autophagy? Aspalathus linearis, also commonly known as rooibos, is an endemic South African plant that is gaining globally acclaim for its antioxidant properties and its role as functional medicinal beverage. In this article we dissect the role of rooibos in the context of the cell's ROS handling capacity, mitochondrial function and autophagy activity. By addressing the dynamic relationship between these critical interconnected systems, and by evaluating the functional properties of rooibos, we unravel its position for preserving cell viability and promoting healthy aging.
Collapse
Affiliation(s)
- Catherine Smit
- Department of Physiological Sciences, Stellenbosch University, Merriman Avenue, Mike de Vries Building, Stellenbosch, 7600, South Africa
| | - Sholto De Wet
- Department of Physiological Sciences, Stellenbosch University, Merriman Avenue, Mike de Vries Building, Stellenbosch, 7600, South Africa
| | - Tamryn Barron
- Department of Physiological Sciences, Stellenbosch University, Merriman Avenue, Mike de Vries Building, Stellenbosch, 7600, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Merriman Avenue, Mike de Vries Building, Stellenbosch, 7600, South Africa.
| |
Collapse
|
42
|
D'Incal CP, Van Rossem KE, De Man K, Konings A, Van Dijck A, Rizzuti L, Vitriolo A, Testa G, Gozes I, Vanden Berghe W, Kooy RF. Chromatin remodeler Activity-Dependent Neuroprotective Protein (ADNP) contributes to syndromic autism. Clin Epigenetics 2023; 15:45. [PMID: 36945042 PMCID: PMC10031977 DOI: 10.1186/s13148-023-01450-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/16/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Individuals affected with autism often suffer additional co-morbidities such as intellectual disability. The genes contributing to autism cluster on a relatively limited number of cellular pathways, including chromatin remodeling. However, limited information is available on how mutations in single genes can result in such pleiotropic clinical features in affected individuals. In this review, we summarize available information on one of the most frequently mutated genes in syndromic autism the Activity-Dependent Neuroprotective Protein (ADNP). RESULTS Heterozygous and predicted loss-of-function ADNP mutations in individuals inevitably result in the clinical presentation with the Helsmoortel-Van der Aa syndrome, a frequent form of syndromic autism. ADNP, a zinc finger DNA-binding protein has a role in chromatin remodeling: The protein is associated with the pericentromeric protein HP1, the SWI/SNF core complex protein BRG1, and other members of this chromatin remodeling complex and, in murine stem cells, with the chromodomain helicase CHD4 in a ChAHP complex. ADNP has recently been shown to possess R-loop processing activity. In addition, many additional functions, for instance, in association with cytoskeletal proteins have been linked to ADNP. CONCLUSIONS We here present an integrated evaluation of all current aspects of gene function and evaluate how abnormalities in chromatin remodeling might relate to the pleiotropic clinical presentation in individual"s" with Helsmoortel-Van der Aa syndrome.
Collapse
Affiliation(s)
- Claudio Peter D'Incal
- Department of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, 2650, Edegem, Belgium
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Epigenetic Signaling Lab (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Kirsten Esther Van Rossem
- Department of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, 2650, Edegem, Belgium
| | - Kevin De Man
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Epigenetic Signaling Lab (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Anthony Konings
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Epigenetic Signaling Lab (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Anke Van Dijck
- Department of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, 2650, Edegem, Belgium
| | - Ludovico Rizzuti
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
- Human Technopole, V. Le Rita Levi-Montalcini, 1, 20157, Milan, Italy
| | - Alessandro Vitriolo
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
- Human Technopole, V. Le Rita Levi-Montalcini, 1, 20157, Milan, Italy
| | - Giuseppe Testa
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
- Human Technopole, V. Le Rita Levi-Montalcini, 1, 20157, Milan, Italy
| | - Illana Gozes
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Sackler School of Medicine, 727, 69978, Tel Aviv, Israel
| | - Wim Vanden Berghe
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Epigenetic Signaling Lab (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, 2650, Edegem, Belgium.
| |
Collapse
|
43
|
Zhang H, Knight C, Chen SRW, Bezprozvanny I. A Gating Mutation in Ryanodine Receptor Type 2 Rescues Phenotypes of Alzheimer's Disease Mouse Models by Upregulating Neuronal Autophagy. J Neurosci 2023; 43:1441-1454. [PMID: 36627208 PMCID: PMC9987572 DOI: 10.1523/jneurosci.1820-22.2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/26/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
It is well established that ryanodine receptors (RyanRs) are overactive in Alzheimer's disease (AD), and it has been suggested that inhibition of RyanR is potentially beneficial for AD treatment. In the present study, we explored a potential connection between basal RyanR activity and autophagy in neurons. Autophagy plays an important role in clearing damaged organelles and long-lived protein aggregates, and autophagy dysregulation occurs in both AD patients and AD animal models. Autophagy is known to be regulated by intracellular calcium (Ca2+) signals, and our results indicated that basal RyanR2 activity in hippocampal neurons inhibited autophagy through activation of calcineurin and the resulting inhibition of the AMPK (AMP-activated protein kinase)-ULK1 (unc-51-like autophagy-activating kinase 1) pathway. Thus, we hypothesized that increased basal RyanR2 activity in AD may lead to the inhibition of neuronal autophagy and accumulation of β-amyloid. To test this hypothesis, we took advantage of the RyanR2-E4872Q knock-in mouse model (EQ) in which basal RyanR2 activity is reduced because of shortened channel open time. We discovered that crossing EQ mice with the APPKI and APPPS1 mouse models of AD (both males and females) rescued amyloid accumulation and LTP impairment in these mice. Our results revealed that reduced basal activity of RyanR2-EQ channels disinhibited the autophagic pathway and led to increased amyloid clearance in these models. These data indicated a potential pathogenic outcome of RyanR2 overactivation in AD and also provided additional targets for therapeutic intervention in AD. Basal activity of ryanodine receptors controls neuronal autophagy and contributes to development of the AD phenotype.SIGNIFICANCE STATEMENT It is well established that neuronal autophagy is impaired in Alzheimer's disease (AD). Our results suggest that supranormal calcium (Ca2+) release from endoplasmic reticulum contributes to the inhibition of autophagy in AD and that reduction in basal activity of type 2 ryanodine receptors disinhibits the neuronal autophagic pathway and leads to increased amyloid clearance in AD models. Our findings directly link neuronal Ca2+ dysregulation with autophagy dysfunction in AD and point to additional targets for therapeutic intervention.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Caitlynn Knight
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas 75390
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas 75390
- Laboratory of Molecular Neurodegeneration, St. Petersburg State Polytechnical Universty, St. Petersburg 195251, Russian Federation
| |
Collapse
|
44
|
Tang Z, Cao J, Yao J, Fan X, Zhao J, Zhao M, Duan Q, Han B, Duan S. KDM1A-mediated upregulation of METTL3 ameliorates Alzheimer's disease via enhancing autophagic clearance of p-Tau through m6A-dependent regulation of STUB1. Free Radic Biol Med 2023; 195:343-358. [PMID: 36587923 DOI: 10.1016/j.freeradbiomed.2022.12.099] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a severe neurodegenerative disorder that progressively destroys cognitive skills. Exploring the mechanism underlying autophagic clearance of phosphorylated tau (p-Tau) contributes to developing novel therapeutic strategies for AD. METHODS SH-SY5Y and HT22 cells were treated with Aβ1-42 to establish an in vitro model of AD. Cell viability was examined using CCK-8. TUNEL staining was applied to evaluate cell apoptosis. LC3 puncta was examined by IF staining. m6A modification level was evaluated through MeRIP. RNA pull-down and RIP assays were used for analyzing the interaction between IGF2BP1 and STUB1 transcripts. The binding of KDM1A to the promoter of METTL3 was confirmed by ChIP assays. APP/PS1 transgenic mice were used as an in vivo model of AD. Cognitive skills of mice were evaluated with the Morris water maze. Hippocampal damage and Aβ deposition were detected through H&E and IHC staining. RESULTS Dysregulated levels of autophagy, p-Tau and m6A was observed in an in vitro model of AD. Overexpression of METTL3 or STUB1 enhanced autophagy but reduced p-Tau level in Aβ1-42-treated cells. METTL3 stabilized STUB1 mRNA through the m6A-IGF2BP1-dependent mechanism and naturally promoted STUB1 expression, thereby enhancing autophagic p-Tau clearance in Aβ1-42-treated cells. Overexpression of KDM1A enhanced autophagy, m6A modification and autophagic p-Tau clearance in Aβ1-42-treated cells. KDM1A-mediated upregulation of METTL3 promoted autophagic p-Tau clearance and ameliorated Alzheimer's disease both in vitro and in vivo. CONCLUSION KDM1A-mediated upregulation of METTL3 enhances autophagic clearance of p-Tau through m6A-dependent regulation of STUB1, thus ameliorating Alzheimer's disease. Our study provides novel mechanistic insights into AD pathogenesis and potential drug targets for AD.
Collapse
Affiliation(s)
- Zhanbin Tang
- The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang, PR China
| | - Jingwei Cao
- The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang, PR China
| | - Jialin Yao
- Harbin First Hospital, Harbin, 150081, Heilongjiang, PR China
| | - Xuehui Fan
- The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang, PR China
| | - Jingkun Zhao
- The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang, PR China
| | - Mianqiao Zhao
- Harbin Second Hospital, Harbin, 150081, Heilongjiang, PR China
| | - Qiong Duan
- The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang, PR China
| | - Baichao Han
- The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang, PR China
| | - Shurong Duan
- The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang, PR China.
| |
Collapse
|
45
|
Emran TB, Chopra H, Dhama K. Autophagy and Alzheimer's disease: How far science has to be progressed? - correspondence. Ann Med Surg (Lond) 2023; 85:323-325. [PMID: 36845802 PMCID: PMC9949842 DOI: 10.1097/ms9.0000000000000156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 12/25/2022] [Indexed: 02/28/2023] Open
Affiliation(s)
- Talha B. Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh India
| |
Collapse
|
46
|
Andronie-Cioara FL, Ardelean AI, Nistor-Cseppento CD, Jurcau A, Jurcau MC, Pascalau N, Marcu F. Molecular Mechanisms of Neuroinflammation in Aging and Alzheimer's Disease Progression. Int J Mol Sci 2023; 24:ijms24031869. [PMID: 36768235 PMCID: PMC9915182 DOI: 10.3390/ijms24031869] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/01/2023] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Aging is the most prominent risk factor for late-onset Alzheimer's disease. Aging associates with a chronic inflammatory state both in the periphery and in the central nervous system, the evidence thereof and the mechanisms leading to chronic neuroinflammation being discussed. Nonetheless, neuroinflammation is significantly enhanced by the accumulation of amyloid beta and accelerates the progression of Alzheimer's disease through various pathways discussed in the present review. Decades of clinical trials targeting the 2 abnormal proteins in Alzheimer's disease, amyloid beta and tau, led to many failures. As such, targeting neuroinflammation via different strategies could prove a valuable therapeutic strategy, although much research is still needed to identify the appropriate time window. Active research focusing on identifying early biomarkers could help translating these novel strategies from bench to bedside.
Collapse
Affiliation(s)
- Felicia Liana Andronie-Cioara
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Adriana Ioana Ardelean
- Department of Preclinical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Carmen Delia Nistor-Cseppento
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
- Correspondence: (C.D.N.-C.); (N.P.)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | | | - Nicoleta Pascalau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
- Correspondence: (C.D.N.-C.); (N.P.)
| | - Florin Marcu
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
47
|
Kamaruzzaman MA, Romli MH, Abas R, Vidyadaran S, Hidayat Baharuldin MT, Nasaruddin ML, Thirupathirao V, Sura S, Warsito K, Mohd Nor NH, Azwaruddin MA, Alshawsh MA, Mohd Moklas MA. Regulatory role of the endocannabinoid system on glial cells toward cognitive function in Alzheimer's disease: A systematic review and meta-analysis of animal studies. Front Pharmacol 2023; 14:1053680. [PMID: 36959856 PMCID: PMC10028478 DOI: 10.3389/fphar.2023.1053680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/08/2023] [Indexed: 03/06/2023] Open
Abstract
Objective: Over the last decade, researchers have sought to develop novel medications against dementia. One potential agent under investigation is cannabinoids. This review systematically appraised and meta-analyzed published pre-clinical research on the mechanism of endocannabinoid system modulation in glial cells and their effects on cognitive function in animal models of Alzheimer's disease (AD). Methods: A systematic review complying with PRISMA guidelines was conducted. Six databases were searched: EBSCOHost, Scopus, PubMed, CINAHL, Cochrane, and Web of Science, using the keywords AD, cannabinoid, glial cells, and cognition. The methodological quality of each selected pre-clinical study was evaluated using the SYRCLE risk of bias tool. A random-effects model was applied to analyze the data and calculate the effect size, while I2 and p-values were used to assess heterogeneity. Results: The analysis included 26 original articles describing (1050 rodents) with AD-like symptoms. Rodents treated with cannabinoid agonists showed significant reductions in escape latency (standard mean difference [SMD] = -1.26; 95% confidence interval [CI]: -1.77 to -0.76, p < 0.00001) and ability to discriminate novel objects (SMD = 1.40; 95% CI: 1.04 to 1.76, p < 0.00001) compared to the control group. Furthermore, a significant decrease in Aβ plaques (SMD = -0.91; 95% CI: -1.55 to -0.27, p = 0.006) was observed in the endocannabinoid-treated group compared to the control group. Trends were observed toward neuroprotection, as represented by decreased levels of glial cell markers including glial fibrillary acid protein (SMD = -1.47; 95% CI: -2.56 to -0.38, p = 0.008) and Iba1 (SMD = -1.67; 95% CI: -2.56 to -0.79, p = 0.0002). Studies on the wild-type mice demonstrated significantly decreased levels of pro-inflammatory markers TNF-α, IL-1, and IL-6 (SMD = -2.28; 95% CI: -3.15 to -1.41, p = 0.00001). Despite the non-significant decrease in pro-inflammatory marker levels in transgenic mice (SMD = -0.47; 95% CI: -1.03 to 0.08, p = 0.09), the result favored the endocannabinoid-treated group over the control group. Conclusion: The revised data suggested that endocannabinoid stimulation promotes cognitive function via modulation of glial cells by decreasing pro-inflammatory markers in AD-like rodent models. Thus, cannabinoid agents may be required to modulate the downstream chain of effect to enhance cognitive stability against concurrent neuroinflammation in AD. Population-based studies and well-designed clinical trials are required to characterize the acceptability and real-world effectiveness of cannabinoid agents. Systematic Review Registration: [https://inplasy.com/inplasy-2022-8-0094/], identifier [Inplasy Protocol 3770].
Collapse
Affiliation(s)
- Mohd Amir Kamaruzzaman
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Muhammad Hibatullah Romli
- Department of Nursing and Rehabilitation, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Razif Abas
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Sharmili Vidyadaran
- Department of Pathology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | | | | | | | - Sreenivasulu Sura
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
- Department of Preclinical Sciences, Faculty of Medicine and Health Sciences, University Tunku Abdul Rahman, Kampar, Malaysia
| | - Kabul Warsito
- Department of Agrotechnology, Faculty of Science and Technology, University of Pembangunan Panca Budi, Medan, Indonesia
| | - Nurul Huda Mohd Nor
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Muhammad Amsyar Azwaruddin
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
| | - Mohammed Abdullah Alshawsh
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Paediatrics, School of Clinical Sciences, Faculty of Medicine, Nursing and Health Science, Monash University, Clayton, VIC, Australia
- *Correspondence: Mohamad Aris Mohd Moklas, ; Mohammed Abdullah Alshawsh,
| | - Mohamad Aris Mohd Moklas
- Department of Human Anatomy, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Kuala Lumpur, Malaysia
- *Correspondence: Mohamad Aris Mohd Moklas, ; Mohammed Abdullah Alshawsh,
| |
Collapse
|
48
|
Deolankar SC, Najar MA, Ramesh P, Kanichery A, Kudva AK, Raghu SV, Prasad TSK. Discovery of Molecular Networks of Neuroprotection Conferred by Brahmi Extract in Aβ 42-Induced Toxicity Model of Drosophila melanogaster Using a Quantitative Proteomic Approach. Mol Neurobiol 2022; 60:303-316. [PMID: 36261695 DOI: 10.1007/s12035-022-03066-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022]
Abstract
Accumulation of Aβ42 peptides forming plaque in various regions of the brain is a hallmark of Alzheimer's disease (AD) progression. However, to date, there is no effective management strategy reported for attenuation of Aβ42-induced toxicity in the early stages of the disease. Alternate medicinal systems such as Ayurveda in the past few decades show promising results in the management of neuronal complications. Medhya Rasayana such as Brahmi is known for its neuroprotective properties via resolving memory-related issues, while the underlying molecular mechanism of the same remains unclear. In the present study, we aimed to understand the neuroprotective effects of the aqueous extract of Bacopa monnieri and Centella asiatica (both commonly known as Brahmi) against the Aβ42 expressing model of the Drosophila melanogaster. By applying a quantitative proteomics approach, the study identified > 90% of differentially expressed proteins from Aβ42 expressing D. melanogaster were either restored to their original expression pattern or showed no change in expression pattern upon receiving either Brahmi extract treatment. The Brahmi restored proteins were part of neuronal pathways associated with cell cycle re-entry, apoptosis, and mitochondrial dynamics. The neuroprotective effect of Brahmi was also validated by negative geotaxis behavioral analysis suggesting its protective role against behavioral deficits exerted by Aβ42 toxicity. We believe that these discoveries will provide a platform for developing novel therapeutics for AD management by deciphering molecular targets of neuroprotection conferred by an aqueous extract of Bacopa monnieri or Centella asiatica.
Collapse
Affiliation(s)
- Sayali Chandrashekhar Deolankar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, India
| | - Mohd Altaf Najar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, India
| | - Poornima Ramesh
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, India
| | - Anagha Kanichery
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, India
| | - Avinash K Kudva
- Department of Biochemistry, Mangalore University, Mangalore, India
| | | | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, India.
| |
Collapse
|
49
|
François M, Karpe AV, Liu JW, Beale DJ, Hor M, Hecker J, Faunt J, Maddison J, Johns S, Doecke JD, Rose S, Leifert WR. Multi-Omics, an Integrated Approach to Identify Novel Blood Biomarkers of Alzheimer's Disease. Metabolites 2022; 12:949. [PMID: 36295851 PMCID: PMC9610280 DOI: 10.3390/metabo12100949] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
The metabolomic and proteomic basis of mild cognitive impairment (MCI) and Alzheimer's disease (AD) is poorly understood, and the relationships between systemic abnormalities in metabolism and AD/MCI pathogenesis is unclear. This study compared the metabolomic and proteomic signature of plasma from cognitively normal (CN) and dementia patients diagnosed with MCI or AD, to identify specific cellular pathways and new biomarkers altered with the progression of the disease. We analysed 80 plasma samples from individuals with MCI or AD, as well as age- and gender-matched CN individuals, by utilising mass spectrometry methods and data analyses that included combined pathway analysis and model predictions. Several proteins clearly identified AD from the MCI and CN groups and included plasma actins, mannan-binding lectin serine protease 1, serum amyloid A2, fibronectin and extracellular matrix protein 1 and Keratin 9. The integrated pathway analysis showed various metabolic pathways were affected in AD, such as the arginine, alanine, aspartate, glutamate and pyruvate metabolism pathways. Therefore, our multi-omics approach identified novel plasma biomarkers for the MCI and AD groups, identified changes in metabolic processes, and may form the basis of a biomarker panel for stratifying dementia participants in future clinical trials.
Collapse
Affiliation(s)
- Maxime François
- CSIRO Health & Biosecurity, Human Health Program, Molecular Diagnostic Solutions Group, Adelaide, SA 5000, Australia
| | - Avinash V. Karpe
- CSIRO Land & Water, Metabolomics Unit, Ecosciences Precinct, Dutton Park, QLD 4001, Australia
| | - Jian-Wei Liu
- CSIRO Land & Water, Agricultural and Environmental Sciences Precinct, Acton, Canberra, ACT 2601, Australia
| | - David J. Beale
- CSIRO Land & Water, Metabolomics Unit, Ecosciences Precinct, Dutton Park, QLD 4001, Australia
| | - Maryam Hor
- CSIRO Health & Biosecurity, Human Health Program, Molecular Diagnostic Solutions Group, Adelaide, SA 5000, Australia
| | - Jane Hecker
- Department of Internal Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Jeff Faunt
- Department of General Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - John Maddison
- Aged Care Rehabilitation & Palliative Care, SA Health, Modbury Hospital, Modbury, SA 5092, Australia
| | - Sally Johns
- Aged Care Rehabilitation & Palliative Care, SA Health, Modbury Hospital, Modbury, SA 5092, Australia
| | - James D. Doecke
- Australian e-Health Research Centre, CSIRO, Level 7, Surgical Treatment and Rehabilitation Service—STARS, Herston, QLD 4029, Australia
| | - Stephen Rose
- Australian e-Health Research Centre, CSIRO, Level 7, Surgical Treatment and Rehabilitation Service—STARS, Herston, QLD 4029, Australia
| | - Wayne R. Leifert
- CSIRO Health & Biosecurity, Human Health Program, Molecular Diagnostic Solutions Group, Adelaide, SA 5000, Australia
| |
Collapse
|
50
|
Jahangiri B, Saei AK, Obi PO, Asghari N, Lorzadeh S, Hekmatirad S, Rahmati M, Velayatipour F, Asghari MH, Saleem A, Moosavi MA. Exosomes, autophagy and ER stress pathways in human diseases: Cross-regulation and therapeutic approaches. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166484. [PMID: 35811032 DOI: 10.1016/j.bbadis.2022.166484] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/01/2022] [Accepted: 07/03/2022] [Indexed: 02/08/2023]
Abstract
Exosomal release pathway and autophagy together maintain homeostasis and survival of cells under stressful conditions. Autophagy is a catabolic process through which cell entities, such as malformed biomacromolecules and damaged organelles, are degraded and recycled via the lysosomal-dependent pathway. Exosomes, a sub-type of extracellular vesicles (EVs) formed by the inward budding of multivesicular bodies (MVBs), are mostly involved in mediating communication between cells. The unfolded protein response (UPR) is an adaptive response that is activated to sustain survival in the cells faced with the endoplasmic reticulum (ER) stress through a complex network that involves protein synthesis, exosomes secretion and autophagy. Disruption of the critical crosstalk between EVs, UPR and autophagy may be implicated in various human diseases, including cancers and neurodegenerative diseases, yet the molecular mechanism(s) behind the coordination of these communication pathways remains obscure. Here, we review the available information on the mechanisms that control autophagy, ER stress and EV pathways, with the view that a better understanding of their crosstalk and balance may improve our knowledge on the pathogenesis and treatment of human diseases, where these pathways are dysregulated.
Collapse
Affiliation(s)
- Babak Jahangiri
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran
| | - Ali Kian Saei
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran
| | - Patience O Obi
- Applied Health Sciences, University of Manitoba, Winnipeg R3T 2N2, Canada; Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg R3T 2N2, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada
| | - Narjes Asghari
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Shirin Hekmatirad
- Department of Pharmacology and Toxicology, School of Medicine, Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Velayatipour
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran
| | - Mohammad Hosseni Asghari
- Department of Pharmacology and Toxicology, School of Medicine, Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Ayesha Saleem
- Applied Health Sciences, University of Manitoba, Winnipeg R3T 2N2, Canada; Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg R3T 2N2, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada.
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran.
| |
Collapse
|