1
|
Cai Y, Yu F, Wu W, Chen W. Study on the mechanism of Dexmedetomidine's effect on postoperative cognitive dysfunction in elderly people. Front Physiol 2025; 16:1508661. [PMID: 40144543 PMCID: PMC11936804 DOI: 10.3389/fphys.2025.1508661] [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: 10/09/2024] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication among elderly patients following surgical procedures, significantly impairing postoperative recovery and quality of life. The selection and dosage of intraoperative anaesthetic drugs are frequently implicated as contributing factors in the development of POCD. In recent years, dexmedetomidine (DEX), a novel α2-adrenoceptor agonist, has been increasingly utilized in surgical anaesthesia for elderly patients, showing potential as both a preventive and therapeutic agent for POCD. This paper provides a comprehensive review of current research on the mechanisms by which DEX affects POCD in the elderly. Additionally, it explores DEX's mechanisms of action in the context of neuroprotection, anti-inflammation, antioxidative stress, and the regulation of apoptosis, autophagy, and analgesia. The objective is to provide reliable theoretical support and a reference point for the clinical application of DEX in POCD among the elderly, thereby promoting its broader use in clinical practice to improve outcomes and enhance quality of life.
Collapse
Affiliation(s)
- Yuanbin Cai
- Department of Anesthesiology, Putuo District Central Hospital, Shanghai, China
| | - Fan Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Wei Wu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Wurong Chen
- Department of Anesthesiology, Putuo District Central Hospital, Shanghai, China
| |
Collapse
|
2
|
Kuo YH, How CM, Liao VHC. Sedimentary co-exposure to bis(2-ethylhexyl) phthalate and titanium dioxide nanoparticles aggravate ecotoxicity and ecological risks through disrupted bioenergetics in Caenorhabditis elegans. MARINE POLLUTION BULLETIN 2025; 212:117515. [PMID: 39752818 DOI: 10.1016/j.marpolbul.2024.117515] [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: 11/08/2024] [Revised: 12/28/2024] [Accepted: 12/28/2024] [Indexed: 03/05/2025]
Abstract
Emerging contaminants in estuarine sediments, such as bis(2-ethylhexyl) phthalate (DEHP) and titanium dioxide nanoparticles (nTiO2), pose ecotoxicological risks that may be exacerbated by co-contamination. This study investigated the impacts of DEHP, nTiO2, and their combinations at environmentally relevant concentrations (1, 10, and 100 μg/g) on the soil nematode Caenorhabditis elegans in estuarine-like sediment (14.25‰ salinity). Life history traits and bioenergetics endpoints were examined, with a sample size of ≥ 45 worms or 9 technical repeats per treatment. While individual exposures did not affect growth, the combination of DEHP (1 μg/g) and nTiO2 (100 μg/g) significantly reduced body length by 19%. Single exposure reduced total offspring by 18-41%, whereas the combination of DEHP and nTiO2 synergistically worsened reproductive toxicity (52-74% inhibition), as revealed by Loewe's additivity model and Bliss's independence. DEBtox modeling revealed a shift in physiological mode of action from "increased reproductive costs" in singular exposures to "increased growth and reproductive cost" in co-exposure. Moreover, co-exposure significantly intensified the impacts on bioenergetics-related endpoints, including ATP level (single exposure: 33-34%; co-exposure: 56%), mitochondrial damage (single exposure: 15-17%; co-exposure: 40%), and oxidative stress (single exposure: 5-7%; co-exposure: 13%). Risk quotients based on reproductive toxicity EC10 and DEBtox-derived zb suggested that environmental concentrations of DEHP and nTiO2 pose high risks in global estuarine sediments, with a 2-fold increase during co-exposure. This study demonstrates that co-contamination of DEHP and nTiO2 synergistically aggravates ecotoxicities through disrupted energy allocation, highlighting the importance of assessing mixture toxicity in environmental risk assessment of estuarine sediments.
Collapse
Affiliation(s)
- Yu-Hsuan Kuo
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Chun Ming How
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
3
|
Wu Z, Cardona EA, Cohn JA, Pierce JT. Nonapoptotic role of EGL-1 in exopher production and neuronal health in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2025; 122:e2407909122. [PMID: 39786930 PMCID: PMC11745333 DOI: 10.1073/pnas.2407909122] [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: 04/22/2024] [Accepted: 11/20/2024] [Indexed: 01/30/2025] Open
Abstract
While traditionally studied for their proapoptotic functions in activating the caspase, research suggests BH3-only proteins also have other roles such as mitochondrial dynamics regulation. Here, we find that EGL-1, the BH3-only protein in Caenorhabditis elegans, promotes the cell-autonomous production of exophers in adult neurons. Exophers are large, micron-scale vesicles that are ejected from the cell and contain cellular components such as mitochondria. EGL-1 facilitates exopher production potentially through regulation of mitochondrial dynamics. Moreover, an endogenous, low level of EGL-1 expression appears to benefit dendritic health. Our findings provide insights into the role of neuronal BH3-only protein in mitochondrial dynamics, downstream exopher production, and ultimately neuronal health.
Collapse
Affiliation(s)
- Zheng Wu
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX78712
| | - Eric A. Cardona
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX78712
| | - Jesse A. Cohn
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX78712
| | - Jonathan T. Pierce
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX78712
| |
Collapse
|
4
|
Griffin EF, Owens MG. Dopaminergic neurodegeneration in C. elegans cultivated with Porphorymonas gingivalis. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001423. [PMID: 39839711 PMCID: PMC11749262 DOI: 10.17912/micropub.biology.001423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/09/2024] [Accepted: 01/02/2025] [Indexed: 01/23/2025]
Abstract
Disruption of the human microbiome has emerged as a major contributing factor in the etiology of neurodegenerative disease. Previous work suggests a positive correlation between periodontal inflammation and Parkinson's disease. Here, we show that feeding C. elegans animals Porphorymonas gingivalis causes neurodegeneration that is not additive with neurodegeneration induced by the Parkinson's-associated protein, α-synuclein. In contrast, α-synuclein-expressing animals fed P. gingivalis show additional disruption in basal slowing, suggesting that P. gingivalis induces neurodegeneration while altering neuronal function of extant neurons. Though the mechanism is unclear, these results suggest a relationship between P. gingivalis and neurodegeneration that warrants further investigation.
Collapse
Affiliation(s)
- Edward F. Griffin
- Natural Sciences, Converse University, Spartanburg, South Carolina, United States
| | - Madeline G. Owens
- Natural Sciences, Converse University, Spartanburg, South Carolina, United States
| |
Collapse
|
5
|
Dundee JM, Brown GC. The microglial P2Y 6 receptor as a therapeutic target for neurodegenerative diseases. Transl Neurodegener 2024; 13:47. [PMID: 39243044 PMCID: PMC11380353 DOI: 10.1186/s40035-024-00438-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024] Open
Abstract
Neurodegenerative diseases are associated with chronic neuroinflammation in the brain, which can result in microglial phagocytosis of live synapses and neurons that may contribute to cognitive deficits and neuronal loss. The microglial P2Y6 receptor (P2Y6R) is a G-protein coupled receptor, which stimulates microglial phagocytosis when activated by extracellular uridine diphosphate, released by stressed neurons. Knockout or inhibition of P2Y6R can prevent neuronal loss in mouse models of Alzheimer's disease (AD), Parkinson's disease, epilepsy, neuroinflammation and aging, and prevent cognitive deficits in models of AD, epilepsy and aging. This review summarises the known roles of P2Y6R in the physiology and pathology of the brain, and its potential as a therapeutic target to prevent neurodegeneration and other brain pathologies.
Collapse
Affiliation(s)
- Jacob M Dundee
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
6
|
Wu Z, Cardona EA, Pierce JT. Non-apoptotic role of EGL-1 in exopher production and neuronal health in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590348. [PMID: 38712027 PMCID: PMC11071422 DOI: 10.1101/2024.04.19.590348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
While traditionally studied for their pro-apoptotic functions, recent research suggests BH3-only proteins also have non-apoptotic roles. Here, we find that EGL-1, the BH3-only protein in Caenorhabditis elegans, promotes the cell-autonomous production of exophers in adult neurons. Exophers are large, micron-scale vesicles that are ejected from the cell and contain cellular components such as mitochondria. EGL-1 facilitates exopher production potentially through regulation of mitochondrial dynamics. Moreover, an endogenous, low level of EGL-1 expression appears to benefit dendritic health. Our findings provide insights into the mechanistic role of BH3-only protein in mitochondrial dynamics, downstream exopher production, and ultimately neuronal health.
Collapse
Affiliation(s)
- Zheng Wu
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX
| | - Eric A. Cardona
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX
| | - Jonathan T. Pierce
- Department of Neuroscience, Center for Learning and Memory, Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX
| |
Collapse
|
7
|
Campbell D, Zuryn S. The mechanisms and roles of mitochondrial dynamics in C. elegans. Semin Cell Dev Biol 2024; 156:266-275. [PMID: 37919144 DOI: 10.1016/j.semcdb.2023.10.006] [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: 06/18/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023]
Abstract
If mitochondria are the powerhouses of the cell, then mitochondrial dynamics are the power grid that regulates how that energy output is directed and maintained in response to unique physiological demands. Fission and fusion dynamics are highly regulated processes that fine-tune the mitochondrial networks of cells to enable appropriate responses to intrinsic and extrinsic stimuli, thereby maintaining cellular and organismal homeostasis. These dynamics shape many aspects of an organism's healthspan including development, longevity, stress resistance, immunity, and response to disease. In this review, we discuss the latest findings regarding the mechanisms and roles of mitochondrial dynamics by focussing on the nematode Caenorhabditis elegans. Whole live-animal studies in C. elegans have enabled a true organismal-level understanding of the impact that mitochondrial dynamics play in homeostasis over a lifetime.
Collapse
Affiliation(s)
- Daniel Campbell
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Steven Zuryn
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
8
|
Mack KL, Kim H, Barbieri EM, Lin J, Braganza S, Jackrel ME, DeNizio JE, Yan X, Chuang E, Tariq A, Cupo RR, Castellano LM, Caldwell KA, Caldwell GA, Shorter J. Tuning Hsp104 specificity to selectively detoxify α-synuclein. Mol Cell 2023; 83:3314-3332.e9. [PMID: 37625404 PMCID: PMC10530207 DOI: 10.1016/j.molcel.2023.07.029] [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] [Revised: 07/19/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023]
Abstract
Hsp104 is an AAA+ protein disaggregase that solubilizes and reactivates proteins trapped in aggregated states. We have engineered potentiated Hsp104 variants to mitigate toxic misfolding of α-synuclein, TDP-43, and FUS implicated in fatal neurodegenerative disorders. Though potent disaggregases, these enhanced Hsp104 variants lack substrate specificity and can have unfavorable off-target effects. Here, to lessen off-target effects, we engineer substrate-specific Hsp104 variants. By altering Hsp104 pore loops that engage substrate, we disambiguate Hsp104 variants that selectively suppress α-synuclein toxicity but not TDP-43 or FUS toxicity. Remarkably, α-synuclein-specific Hsp104 variants emerge that mitigate α-synuclein toxicity via distinct ATPase-dependent mechanisms involving α-synuclein disaggregation or detoxification of soluble α-synuclein conformers. Importantly, both types of α-synuclein-specific Hsp104 variant reduce dopaminergic neurodegeneration in a C. elegans model of Parkinson's disease more effectively than non-specific variants. We suggest that increasing the substrate specificity of enhanced disaggregases could be applied broadly to tailor therapeutics for neurodegenerative disease.
Collapse
Affiliation(s)
- Korrie L Mack
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hanna Kim
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Edward M Barbieri
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - JiaBei Lin
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sylvanne Braganza
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meredith E Jackrel
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jamie E DeNizio
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaohui Yan
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Edward Chuang
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amber Tariq
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan R Cupo
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura M Castellano
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA.
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Thies JL, Willicott K, Craig ML, Greene MR, DuGay CN, Caldwell GA, Caldwell KA. Xanthine Dehydrogenase Is a Modulator of Dopaminergic Neurodegeneration in Response to Bacterial Metabolite Exposure in C. elegans. Cells 2023; 12:1170. [PMID: 37190079 PMCID: PMC10136629 DOI: 10.3390/cells12081170] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Oxidative stress is a contributing factor to Parkinson's disease (PD). Considering the prevalence of sporadic PD, environmental exposures are postulated to increase reactive oxygen species and either incite or exacerbate neurodegeneration. We previously determined that exposure to the common soil bacterium, Streptomyces venezuelae (S. ven), enhanced oxidative stress and mitochondrial dysfunction in Caenorhabditis elegans, leading to dopaminergic (DA) neurodegeneration. Here, S. ven metabolite exposure in C. elegans was followed by RNA-Seq analysis. Half of the differentially identified genes (DEGs) were associated with the transcription factor DAF-16 (FOXO), which is a key node in regulating stress response. Our DEGs were enriched for Phase I (CYP) and Phase II (UGT) detoxification genes and non-CYP Phase I enzymes associated with oxidative metabolism, including the downregulated xanthine dehydrogenase gene, xdh-1. The XDH-1 enzyme exhibits reversible interconversion to xanthine oxidase (XO) in response to calcium. S. ven metabolite exposure enhanced XO activity in C. elegans. The chelation of calcium diminishes the conversion of XDH-1 to XO and results in neuroprotection from S. ven exposure, whereas CaCl2 supplementation enhanced neurodegeneration. These results suggest a defense mechanism that delimits the pool of XDH-1 available for interconversion to XO, and associated ROS production, in response to metabolite exposure.
Collapse
Affiliation(s)
- Jennifer L. Thies
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Karolina Willicott
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Maici L. Craig
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Madeline R. Greene
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Cassandra N. DuGay
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Guy A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kim A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
10
|
Keizer HG, Brands R, Seinen W. An AMP Kinase-pathway dependent integrated stress response regulates ageing and longevity. Biogerontology 2023:10.1007/s10522-023-10024-3. [PMID: 36877293 DOI: 10.1007/s10522-023-10024-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/18/2023] [Indexed: 03/07/2023]
Abstract
The purpose of this article is to investigate the role of the AMP-kinase pathway (AMPK pathway) in the induction of a concomitant set of health benefits by exercise, numerous drugs, and health ingredients, all of which are adversely affected by ageing. Despite the AMPK pathway being frequently mentioned in relation to both these health effects and ageing, it appears challenging to understand how the activation of a single biochemical pathway by various treatments can produce such a diverse range of concurrent health benefits, involving so many organs. We discovered that the AMPK pathway functions as an integrated stress response system because of the presence of a feedback loop in it. This evolutionary conserved stress response system detects changes in AMP/ATP and NAD/NADH ratios, as well as the presence of potential toxins, and responds by activating a common protective transcriptional response that protects against aging and promotes longevity. The inactivation of the AMPK pathway with age most likely explains why ageing has a negative impact on the above-mentioned set of health benefits. We conclude that the presence of a feedback loop in the AMP-kinase pathway positions this pathway as an AMPK-ISR (AMP Kinase-dependent integrated stress response) system that responds to almost any type of (moderate) environmental stress by inducing various age-related health benefits and longevity.
Collapse
Affiliation(s)
- H G Keizer
- AMRIF Biotechnology, Agrobusiness Park 10, 6708 PW, Wageningen, The Netherlands.
| | - R Brands
- AMRIF Biotechnology, Agrobusiness Park 10, 6708 PW, Wageningen, The Netherlands.,Institute for Risk Assessment Sciences (IRAS), Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| | - W Seinen
- AMRIF Biotechnology, Agrobusiness Park 10, 6708 PW, Wageningen, The Netherlands.,Institute for Risk Assessment Sciences (IRAS), Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| |
Collapse
|
11
|
Srivastava V, Zelmanovich V, Shukla V, Abergel R, Cohen I, Ben-Sasson SA, Gross E. Distinct designer diamines promote mitophagy, and thereby enhance healthspan in C. elegans and protect human cells against oxidative damage. Autophagy 2023; 19:474-504. [PMID: 35579620 PMCID: PMC9851263 DOI: 10.1080/15548627.2022.2078069] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Impaired mitophagy is a primary pathogenic event underlying diverse aging-associated diseases such as Alzheimer and Parkinson diseases and sarcopenia. Therefore, augmentation of mitophagy, the process by which defective mitochondria are removed, then replaced by new ones, is an emerging strategy for preventing the evolvement of multiple morbidities in the elderly population. Based on the scaffold of spermidine (Spd), a known mitophagy-promoting agent, we designed and tested a family of structurally related compounds. A prototypic member, 1,8-diaminooctane (VL-004), exceeds Spd in its ability to induce mitophagy and protect against oxidative stress. VL-004 activity is mediated by canonical aging genes and promotes lifespan and healthspan in C. elegans. Moreover, it enhances mitophagy and protects against oxidative injury in rodent and human cells. Initial structural characterization suggests simple rules for the design of compounds with improved bioactivity, opening the way for a new generation of agents with a potential to promote healthy aging.
Collapse
Affiliation(s)
- Vijigisha Srivastava
- Department Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Veronica Zelmanovich
- Department Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Virendra Shukla
- Department Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rachel Abergel
- Department Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Irit Cohen
- Department Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shmuel A. Ben-Sasson
- Department Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Einav Gross
- Department Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel,CONTACT Einav Gross Department Biochemistry and Molecular Biology, IMRIC, Faculty of Medicine, the Hebrew University of Jerusalem, Ein Kerem. PO Box 12271, Jerusalem9112102, Israel
| |
Collapse
|
12
|
Lipinski RJ, Krauss RS. Gene-environment interactions in birth defect etiology: Challenges and opportunities. Curr Top Dev Biol 2023; 152:1-30. [PMID: 36707208 PMCID: PMC9942595 DOI: 10.1016/bs.ctdb.2022.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Birth defects are relatively common congenital outcomes that significantly impact affected individuals, their families, and communities. Effective development and deployment of prevention and therapeutic strategies for these conditions requires sufficient understanding of etiology, including underlying genetic and environmental causes. Tremendous progress has been made in defining the genetic basis of familial and syndromic forms of birth defects. However, the majority of birth defect cases are considered nonsyndromic and thought to result from multifactorial gene-environment interactions. While substantial advances have been made in elucidating the genetic landscape of these etiologically complex conditions, significant biological and technical constraints have stymied progress toward a refined knowledge of environmental risk factors. Defining specific gene-environment interactions in birth defect etiology is even more challenging. However, progress has been made, including demonstration of critical proofs of concept and development of new conceptual and technical approaches for resolving complex gene-environment interactions. In this review, we discuss current views of multifactorial birth defect etiology, comparing them with other diseases that also involve gene-environment interactions, including primary immunodeficiency and cancer. We describe how various model systems have illuminated mechanisms of multifactorial etiology and these models' individual strengths and weaknesses. Finally, suggestions for areas of future emphasis are proposed.
Collapse
Affiliation(s)
- Robert J. Lipinski
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, United States,Corresponding authors: ;
| | - Robert S. Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States,Corresponding authors: ;
| |
Collapse
|
13
|
Boos JR, Jandrain HN, Hagiuda E, Taguchi AT, Hasegawa K, Fedun BL, Taylor SJ, Elad SM, Faber SE, Kumasaka T, Iwasaki T, Geldenhuys WJ. Structure and biological evaluation of Caenorhabditis elegans CISD-1/mitoNEET, a KLP-17 tail domain homologue, supports attenuation of paraquat-induced oxidative stress through a p38 MAPK-mediated antioxidant defense response. ADVANCES IN REDOX RESEARCH : AN OFFICIAL JOURNAL OF THE SOCIETY FOR REDOX BIOLOGY AND MEDICINE AND THE SOCIETY FOR FREE RADICAL RESEARCH-EUROPE 2022; 6:100048. [PMID: 36533211 PMCID: PMC9757825 DOI: 10.1016/j.arres.2022.100048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
CISD-1/mitoNEET is an evolutionarily conserved outer mitochondrial membrane [2Fe-2S] protein that regulates mitochondrial function and morphology. The [2Fe-2S] clusters are redox reactive and shown to mediate oxidative stress in vitro and in vivo. However, there is limited research studying CISD-1/mitoNEET mediation of oxidative stress in response to environmental stressors. In this study, we have determined the X-ray crystal structure of Caenorhabditis elegans CISD-1/mitoNEET homologue and evaluated the mechanisms of oxidative stress resistance to the pro-oxidant paraquat in age-synchronized populations by generating C. elegans gain and loss of function CISD-1 models. The structure of the C. elegans CISD-1/mitoNEET soluble domain refined at 1.70-Å resolution uniquely shows a reversible disulfide linkage at the homo-dimeric interface and also represents the N-terminal tail domain for dimerization of the cognate kinesin motor protein KLP-17 involved in chromosome segregation dynamics and germline development of the nematode. Moreover, overexpression of CISD-1/mitoNEET in C. elegans has revealed beneficial effects on oxidative stress resistance against paraquat-induced reactive oxygen species generation, corroborated by increased activation of the p38 mitogen-activated protein kinase (MAPK) signaling cascade.
Collapse
Affiliation(s)
- Jacob R. Boos
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Hanna N. Jandrain
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Emi Hagiuda
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Sendagi, Tokyo 113-8602, Japan
| | - Alexander T. Taguchi
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Sendagi, Tokyo 113-8602, Japan
| | - Kazuya Hasegawa
- Japan Synchrotron Radiation Research Institute (JASRI), SPring-8, Sayo, Hyogo 679-5198, Japan
| | - Bailey L. Fedun
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Sarah J. Taylor
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Sofhia M. Elad
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Sarah E. Faber
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Takashi Kumasaka
- Japan Synchrotron Radiation Research Institute (JASRI), SPring-8, Sayo, Hyogo 679-5198, Japan
| | - Toshio Iwasaki
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Sendagi, Tokyo 113-8602, Japan
| | - Werner J. Geldenhuys
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
14
|
Zhang X, Ye Y, Sun J, Xu Y, Huang Y, Wang JS, Tang L, Ji J, Chen BY, Sun X. Polygonatum sibiricum polysaccharide extract relieves FB1-induced neurotoxicity by reducing oxidative stress and mitochondrial damage in Caenorhabditis elegans. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Begelman DV, Woods G, Bhaumik D, Angeli S, Foulger AC, Lucanic M, Lan J, Andersen JK, Lithgow GJ. An aco-2::gfp knock-in enables the monitoring of mitochondrial morphology throughout C. elegans lifespan. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000599. [PMID: 35903774 PMCID: PMC9315405 DOI: 10.17912/micropub.biology.000599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/28/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022]
Abstract
We used CRISPR/Cas9 gene editing in C. elegans in order to fluorescently tag endogenous aconitase-2 (ACO-2). ACO-2 is a mitochondrially localized protein, and the aco-2::gfp strain enabled the examination of native mitochondrial morphology in live animals. Here we validate that the aco-2::gfp strain displays the prototypic changes in mitochondrial morphology known to occur during aging and upon paraquat (PQ) induced mitochondrial stress. We also provide evidence that the ACO-2::GFP reporter can serve as a superior means for tracking mitochondrial morphology than conventional MitoTracker dyes-especially in aged-worms.
Collapse
Affiliation(s)
| | | | | | - Suzanne Angeli
- The Buck Institute for Research on Aging
,
University of Maine, Molecular & Biomedical Sciences
| | | | - Mark Lucanic
- The Buck Institute for Research on Aging
,
GeroStateAlpha
| | - Jianfeng Lan
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, the Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Julie K. Andersen
- The Buck Institute for Research on Aging
,
Correspondence to: Julie K. Andersen (
)
| | - Gordon J. Lithgow
- The Buck Institute for Research on Aging
,
Correspondence to: Gordon J. Lithgow (
)
| |
Collapse
|
16
|
De Miranda BR, Goldman SM, Miller GW, Greenamyre JT, Dorsey ER. Preventing Parkinson's Disease: An Environmental Agenda. JOURNAL OF PARKINSONS DISEASE 2021; 12:45-68. [PMID: 34719434 PMCID: PMC8842749 DOI: 10.3233/jpd-212922] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fueled by aging populations and continued environmental contamination, the global burden of Parkinson's disease (PD) is increasing. The disease, or more appropriately diseases, have multiple environmental and genetic influences but no approved disease modifying therapy. Additionally, efforts to prevent this debilitating disease have been limited. As numerous environmental contaminants (e.g., pesticides, metals, industrial chemicals) are implicated in PD, disease prevention is possible. To reduce the burden of PD, we have compiled preclinical and clinical research priorities that highlight both disease prediction and primary prevention. Though not exhaustive, the "PD prevention agenda" builds upon many years of research by our colleagues and proposes next steps through the lens of modifiable risk factors. The agenda identifies ten specific areas of further inquiry and considers the funding and policy changes that will be necessary to help prevent the world's fastest growing brain disease.
Collapse
Affiliation(s)
- Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama atBirmingham, Birmingham, AL, USA
| | - Samuel M Goldman
- Division of Occupational and Environmental Medicine, San Francisco VeteransAffairs Health Care System, School of Medicine, University ofCalifornia-San Francisco, San Francisco, CA, USA
| | - Gary W Miller
- Department of Environmnetal Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, Universityof Pittsburgh, Pittsburgh, PA, USA
| | - E Ray Dorsey
- Center for Health+Technology and Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
17
|
Wei CC, Yang NC, Huang CW. Zearalenone Induces Dopaminergic Neurodegeneration via DRP-1-Involved Mitochondrial Fragmentation and Apoptosis in a Caenorhabditis elegans Parkinson's Disease Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:12030-12038. [PMID: 34586801 DOI: 10.1021/acs.jafc.1c05836] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The contamination of mycotoxin zearalenone (ZEN) in foods has been reported worldwide, resulting in potential risks to food safety. However, the toxic mechanism of ZEN on neurodegenerative diseases has not been fully elucidated. Therefore, this study conducted in vivo ZEN neurotoxicity assessment on Parkinson's disease (PD)-related dopaminergic neurodegeneration and mitochondrial dysfunction using Caenorhabditis elegans. The results demonstrated that dopaminergic neuron damage was induced by ZEN exposure (1.25, 10, and 50 μM), and dopaminergic neuron-related behaviors were adversely affected subsequently. Additionally, the mitochondrial fragmentation was significantly increased by ZEN exposure. Moreover, upregulated expression of mitochondrial fission and cell apoptosis-related genes (drp-1, egl-1, ced-4, and ced-3) revealed the crucial role of DRP-1 on ZEN-induced neurotoxicity, which was further confirmed by drp-1 mutant and RNAi assays. In conclusion, our study indicates ZEN-induced dopaminergic neurodegeneration via DRP-1-involved mitochondrial fragmentation and apoptosis, which might cause harmful effects on PD-related symptoms.
Collapse
Affiliation(s)
- Chia-Cheng Wei
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan
| | - Nien-Chieh Yang
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan
| | - Chi-Wei Huang
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd., Taipei 100, Taiwan
| |
Collapse
|
18
|
Chandler R, Cogo S, Lewis P, Kevei E. Modelling the functional genomics of Parkinson's disease in Caenorhabditis elegans: LRRK2 and beyond. Biosci Rep 2021; 41:BSR20203672. [PMID: 34397087 PMCID: PMC8415217 DOI: 10.1042/bsr20203672] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
For decades, Parkinson's disease (PD) cases have been genetically categorised into familial, when caused by mutations in single genes with a clear inheritance pattern in affected families, or idiopathic, in the absence of an evident monogenic determinant. Recently, genome-wide association studies (GWAS) have revealed how common genetic variability can explain up to 36% of PD heritability and that PD manifestation is often determined by multiple variants at different genetic loci. Thus, one of the current challenges in PD research stands in modelling the complex genetic architecture of this condition and translating this into functional studies. Caenorhabditis elegans provide a profound advantage as a reductionist, economical model for PD research, with a short lifecycle, straightforward genome engineering and high conservation of PD relevant neural, cellular and molecular pathways. Functional models of PD genes utilising C. elegans show many phenotypes recapitulating pathologies observed in PD. When contrasted with mammalian in vivo and in vitro models, these are frequently validated, suggesting relevance of C. elegans in the development of novel PD functional models. This review will discuss how the nematode C. elegans PD models have contributed to the uncovering of molecular and cellular mechanisms of disease, with a focus on the genes most commonly found as causative in familial PD and risk factors in idiopathic PD. Specifically, we will examine the current knowledge on a central player in both familial and idiopathic PD, Leucine-rich repeat kinase 2 (LRRK2) and how it connects to multiple PD associated GWAS candidates and Mendelian disease-causing genes.
Collapse
Affiliation(s)
| | - Susanna Cogo
- School of Biological Sciences, University of Reading, Reading, RG6 6AH, U.K
- Department of Biology, University of Padova, Padova, Via Ugo Bassi 58/B, 35121, Italy
| | - Patrick A. Lewis
- Royal Veterinary College, University of London, London, NW1 0TU, U.K
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, U.K
| | - Eva Kevei
- School of Biological Sciences, University of Reading, Reading, RG6 6AH, U.K
| |
Collapse
|
19
|
Aberrant Mitochondrial Dynamics: An Emerging Pathogenic Driver of Abdominal Aortic Aneurysm. Cardiovasc Ther 2021; 2021:6615400. [PMID: 34221126 PMCID: PMC8221877 DOI: 10.1155/2021/6615400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 05/13/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is defined as a progressive segmental dilation of the abdominal aorta and is associated with high mortality. The characterized features of AAA indicate several underlying mechanisms of AAA formation and progression, including reactive oxygen species production, inflammation, and atherosclerosis. Mitochondrial functions are critical for determining cell fate, and mitochondrial dynamics, especially selective mitochondrial autophagy, which is termed as mitophagy, has emerged as an important player in the pathogenesis of several cardiovascular diseases. The PARKIN/PARIS/PGC1α pathway is associated with AAA formation and has been proposed to play a role in mitochondrial dynamics mediated by the PINK/PARKIN pathway in the pathogenesis underlying AAA. This review is aimed at deepening our understanding of AAA formation and progression, which is vital for the development of potential medical therapies for AAA.
Collapse
|
20
|
Steele JR, Strange N, Rodgers KJ, Padula MP. A Novel Method for Creating a Synthetic L-DOPA Proteome and In Vitro Evidence of Incorporation. Proteomes 2021; 9:24. [PMID: 34073856 PMCID: PMC8162537 DOI: 10.3390/proteomes9020024] [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: 03/19/2021] [Revised: 05/02/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022] Open
Abstract
Proteinopathies are protein misfolding diseases that have an underlying factor that affects the conformation of proteoforms. A factor hypothesised to play a role in these diseases is the incorporation of non-protein amino acids into proteins, with a key example being the therapeutic drug levodopa. The presence of levodopa as a protein constituent has been explored in several studies, but it has not been examined in a global proteomic manner. This paper provides a proof-of-concept method for enzymatically creating levodopa-containing proteins using the enzyme tyrosinase and provides spectral evidence of in vitro incorporation in addition to the induction of the unfolded protein response due to levodopa.
Collapse
Affiliation(s)
- Joel Ricky Steele
- Proteomics Core Facility and School of Life Sciences, The University of Technology Sydney, Ultimo, NSW 2007, Australia;
- Neurotoxin Research Group, School of Life Sciences, The University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Natalie Strange
- School of Life Sciences, The University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Kenneth J. Rodgers
- Neurotoxin Research Group, School of Life Sciences, The University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Matthew P. Padula
- Proteomics Core Facility and School of Life Sciences, The University of Technology Sydney, Ultimo, NSW 2007, Australia;
| |
Collapse
|
21
|
Oh KH, Sheoran S, Richmond JE, Kim H. Alcohol induces mitochondrial fragmentation and stress responses to maintain normal muscle function in Caenorhabditis elegans. FASEB J 2020; 34:8204-8216. [PMID: 32294300 DOI: 10.1096/fj.201903166r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/30/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
Chronic excessive ethanol consumption has distinct toxic and adverse effects on a variety of tissues. In skeletal muscle, ethanol causes alcoholic myopathy, which is characterized by myofiber atrophy and the loss of muscle strength. Alcoholic myopathy is more prevalent than all inherited muscle diseases combined. Current evidence indicates that ethanol directly impairs muscle organization and function. However, the underlying mechanism by which ethanol causes toxicity in muscle is poorly understood. Here, we show that the nematode Caenorhabditis elegans exhibits the key features of alcoholic myopathy when exposed to ethanol. As in mammals, ethanol exposure impairs muscle strength and induces the expression of protective genes, including oxidative stress response genes. In addition, ethanol exposure causes the fragmentation of mitochondrial networks aligned with myofibril lattices. This ethanol-induced mitochondrial fragmentation is dependent on the mitochondrial fission factor DRP-1 (dynamin-related protein 1) and its receptor proteins on the outer mitochondrial membrane. Our data indicate that this fragmentation contributes to the activation of the mitochondrial unfolded protein response (UPR). We also found that robust, perpetual mitochondrial UPR activation effectively reduces muscle weakness caused by ethanol exposure. Our results strongly suggest that the modulation of mitochondrial stress responses may provide a method to ameliorate alcohol toxicity and damage to muscle.
Collapse
Affiliation(s)
- Kelly H Oh
- Department of Cell Biology & Anatomy, Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.,School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Seema Sheoran
- Department of Biological Science, University of Illinois at Chicago, Chicago, IL, USA
| | - Janet E Richmond
- Department of Biological Science, University of Illinois at Chicago, Chicago, IL, USA
| | - Hongkyun Kim
- Department of Cell Biology & Anatomy, Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.,School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
22
|
Tao J, Hao Y, Li X, Yin H, Nie X, Zhang J, Xu B, Chen Q, Li B. Systematic Identification of Housekeeping Genes Possibly Used as References in Caenorhabditis elegans by Large-Scale Data Integration. Cells 2020; 9:E786. [PMID: 32213971 PMCID: PMC7140892 DOI: 10.3390/cells9030786] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022] Open
Abstract
For accurate gene expression quantification, normalization of gene expression data against reliable reference genes is required. It is known that the expression levels of commonly used reference genes vary considerably under different experimental conditions, and therefore, their use for data normalization is limited. In this study, an unbiased identification of reference genes in Caenorhabditis elegans was performed based on 145 microarray datasets (2296 gene array samples) covering different developmental stages, different tissues, drug treatments, lifestyle, and various stresses. As a result, thirteen housekeeping genes (rps-23, rps-26, rps-27, rps-16, rps-2, rps-4, rps-17, rpl-24.1, rpl-27, rpl-33, rpl-36, rpl-35, and rpl-15) with enhanced stability were comprehensively identified by using six popular normalization algorithms and RankAggreg method. Functional enrichment analysis revealed that these genes were significantly overrepresented in GO terms or KEGG pathways related to ribosomes. Validation analysis using recently published datasets revealed that the expressions of newly identified candidate reference genes were more stable than the commonly used reference genes. Based on the results, we recommended using rpl-33 and rps-26 as the optimal reference genes for microarray and rps-2 and rps-4 for RNA-sequencing data validation. More importantly, the most stable rps-23 should be a promising reference gene for both data types. This study, for the first time, successfully displays a large-scale microarray data driven genome-wide identification of stable reference genes for normalizing gene expression data and provides a potential guideline on the selection of universal internal reference genes in C. elegans, for quantitative gene expression analysis.
Collapse
Affiliation(s)
- Jingxin Tao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Youjin Hao
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Xudong Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Huachun Yin
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Xiner Nie
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Jie Zhang
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Boying Xu
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| | - Qiao Chen
- Scientific Research Office, Chongqing Normal University, Chongqing 401331, China;
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China; (J.T.); (Y.H.); (X.L.); (H.Y.); (X.N.); (J.Z.); (B.X.)
| |
Collapse
|
23
|
Feng ST, Wang ZZ, Yuan YH, Wang XL, Sun HM, Chen NH, Zhang Y. Dynamin-related protein 1: A protein critical for mitochondrial fission, mitophagy, and neuronal death in Parkinson’s disease. Pharmacol Res 2020; 151:104553. [DOI: 10.1016/j.phrs.2019.104553] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/14/2019] [Accepted: 11/16/2019] [Indexed: 01/14/2023]
|
24
|
Madreiter‐Sokolowski CT, Ramadani‐Muja J, Ziomek G, Burgstaller S, Bischof H, Koshenov Z, Gottschalk B, Malli R, Graier WF. Tracking intra- and inter-organelle signaling of mitochondria. FEBS J 2019; 286:4378-4401. [PMID: 31661602 PMCID: PMC6899612 DOI: 10.1111/febs.15103] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/19/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022]
Abstract
Mitochondria are as highly specialized organelles and masters of the cellular energy metabolism in a constant and dynamic interplay with their cellular environment, providing adenosine triphosphate, buffering Ca2+ and fundamentally contributing to various signaling pathways. Hence, such broad field of action within eukaryotic cells requires a high level of structural and functional adaptation. Therefore, mitochondria are constantly moving and undergoing fusion and fission processes, changing their shape and their interaction with other organelles. Moreover, mitochondrial activity gets fine-tuned by intra- and interorganelle H+ , K+ , Na+ , and Ca2+ signaling. In this review, we provide an up-to-date overview on mitochondrial strategies to adapt and respond to, as well as affect, their cellular environment. We also present cutting-edge technologies used to track and investigate subcellular signaling, essential to the understanding of various physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Corina T. Madreiter‐Sokolowski
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
- Department of Health Sciences and TechnologyETH ZurichSchwerzenbachSwitzerland
| | - Jeta Ramadani‐Muja
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Gabriela Ziomek
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Sandra Burgstaller
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Helmut Bischof
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Zhanat Koshenov
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Benjamin Gottschalk
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
- BioTechMedGrazAustria
| | - Wolfgang F. Graier
- Gottfried Schatz Research Center, Molecular Biology and BiochemistryMedical University of GrazAustria
- BioTechMedGrazAustria
| |
Collapse
|
25
|
Hartman JH, Gonzalez-Hunt C, Hall SM, Ryde IT, Caldwell KA, Caldwell GA, Meyer JN. Genetic Defects in Mitochondrial Dynamics in Caenorhabditis elegans Impact Ultraviolet C Radiation- and 6-hydroxydopamine-Induced Neurodegeneration. Int J Mol Sci 2019; 20:ijms20133202. [PMID: 31261893 PMCID: PMC6651461 DOI: 10.3390/ijms20133202] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/30/2022] Open
Abstract
Background: Parkinson’s disease (PD) is one of the most common neurodegenerative disorders involving devastating loss of dopaminergic neurons in the substantia nigra. Early steps in PD pathogenesis include mitochondrial dysfunction, and mutations in mitochondrial genes have been linked to familial forms of the disease. However, low penetrance of mutations indicates a likely important role for environmental factors in PD risk through gene by environment interactions. Herein, we study how genetic deficiencies in mitochondrial dynamics processes including fission, fusion, and mitophagy interact with environmental exposures to impact neurodegeneration. Methods: We utilized the powerful model organism Caenorhabditis elegans to study ultraviolet C radiation (UVC)- and 6-hydroxydopamine-induced degeneration of fluorescently-tagged dopaminergic neurons in the background of fusion deficiency (MFN1/2 homolog, fzo-1), fission deficiency (DMN1L homolog, drp-1), and mitochondria-specific autophagy (mitophagy) deficiency (PINK1 and PRKN homologs, pink-1 and pdr-1). Results: Overall, we found that deficiency in either mitochondrial fusion or fission sensitizes nematodes to UVC exposure (used to model common environmental pollutants) but protects from 6-hydroxydopamine-induced neurodegeneration. By contrast, mitophagy deficiency makes animals more sensitive to these stressors with an interesting exception—pink-1 deficiency conferred remarkable protection from 6-hydroxydopamine. We found that this protection could not be explained by compensatory antioxidant gene expression in pink-1 mutants or by differences in mitochondrial morphology. Conclusions: Together, our results support a strong role for gene by environment interactions in driving dopaminergic neurodegeneration and suggest that genetic deficiency in mitochondrial processes can have complex effects on neurodegeneration.
Collapse
Affiliation(s)
- Jessica H Hartman
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | | | - Samantha M Hall
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Ian T Ryde
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Joel N Meyer
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
26
|
Byrne JJ, Soh MS, Chandhok G, Vijayaraghavan T, Teoh JS, Crawford S, Cobham AE, Yapa NMB, Mirth CK, Neumann B. Disruption of mitochondrial dynamics affects behaviour and lifespan in Caenorhabditis elegans. Cell Mol Life Sci 2019; 76:1967-1985. [PMID: 30840087 PMCID: PMC6478650 DOI: 10.1007/s00018-019-03024-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 01/11/2019] [Accepted: 01/22/2019] [Indexed: 01/29/2023]
Abstract
Mitochondria are essential components of eukaryotic cells, carrying out critical physiological processes that include energy production and calcium buffering. Consequently, mitochondrial dysfunction is associated with a range of human diseases. Fundamental to their function is the ability to transition through fission and fusion states, which is regulated by several GTPases. Here, we have developed new methods for the non-subjective quantification of mitochondrial morphology in muscle and neuronal cells of Caenorhabditis elegans. Using these techniques, we uncover surprising tissue-specific differences in mitochondrial morphology when fusion or fission proteins are absent. From ultrastructural analysis, we reveal a novel role for the fusion protein FZO-1/mitofusin 2 in regulating the structure of the inner mitochondrial membrane. Moreover, we have determined the influence of the individual mitochondrial fission (DRP-1/DRP1) and fusion (FZO-1/mitofusin 1,2; EAT-3/OPA1) proteins on animal behaviour and lifespan. We show that loss of these mitochondrial fusion or fission regulators induced age-dependent and progressive deficits in animal movement, as well as in muscle and neuronal function. Our results reveal that disruption of fusion induces more profound defects than lack of fission on animal behaviour and tissue function, and imply that while fusion is required throughout life, fission is more important later in life likely to combat ageing-associated stressors. Furthermore, our data demonstrate that mitochondrial function is not strictly dependent on morphology, with no correlation found between morphological changes and behavioural defects. Surprisingly, we find that disruption of either mitochondrial fission or fusion significantly reduces median lifespan, but maximal lifespan is unchanged, demonstrating that mitochondrial dynamics play an important role in limiting variance in longevity across isogenic populations. Overall, our study provides important new insights into the central role of mitochondrial dynamics in maintaining organismal health.
Collapse
Affiliation(s)
- Joseph J Byrne
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Ming S Soh
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Gursimran Chandhok
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Tarika Vijayaraghavan
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Jean-Sébastien Teoh
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Simon Crawford
- Monash Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Melbourne, VIC, 3800, Australia
| | - Ansa E Cobham
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - Nethmi M B Yapa
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Christen K Mirth
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - Brent Neumann
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
27
|
Griffin EF, Scopel SE, Stephen CA, Holzhauer AC, Vaji MA, Tuckey RA, Berkowitz LA, Caldwell KA, Caldwell GA. ApoE-associated modulation of neuroprotection from Aβ-mediated neurodegeneration in transgenic Caenorhabditis elegans. Dis Model Mech 2019; 12:dmm.037218. [PMID: 30683808 PMCID: PMC6398492 DOI: 10.1242/dmm.037218] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/17/2019] [Indexed: 12/13/2022] Open
Abstract
Allele-specific distinctions in the human apolipoprotein E (APOE) locus represent the best-characterized genetic predictor of Alzheimer's disease (AD) risk. Expression of isoform APOEε2 is associated with reduced risk, while APOEε3 is neutral and APOEε4 carriers exhibit increased susceptibility. Using Caenorhabditis elegans, we generated a novel suite of humanized transgenic nematodes to facilitate neuronal modeling of amyloid-beta peptide (Aβ) co-expression in the context of distinct human APOE alleles. We found that co-expression of human APOEε2 with Aβ attenuated Aβ-induced neurodegeneration, whereas expression of the APOEε4 allele had no effect on neurodegeneration, indicating a loss of neuroprotective capacity. Notably, the APOEε3 allele displayed an intermediate phenotype; it was not neuroprotective in young adults but attenuated neurodegeneration in older animals. There was no functional impact from the three APOE isoforms in the absence of Aβ co-expression. Pharmacological treatment that examined neuroprotective effects of APOE alleles on calcium homeostasis showed allele-specific responses to changes in ER-associated calcium dynamics in the Aβ background. Additionally, Aβ suppressed survival, an effect that was rescued by APOEε2 and APOEε3, but not APOEε4. Expression of the APOE alleles in neurons, independent of Aβ, exerted no impact on survival. Taken together, these results illustrate that C. elegans provides a powerful in vivo platform with which to explore how AD-associated neuronal pathways are modulated by distinct APOE gene products in the context of Aβ-associated neurotoxicity. The significance of both ApoE and Aβ to AD highlights the utility of this new pre-clinical model as a means to dissect their functional inter-relationship.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Edward F Griffin
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487-0344, USA
| | - Samuel E Scopel
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487-0344, USA
| | - Cayman A Stephen
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487-0344, USA
| | - Adam C Holzhauer
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487-0344, USA
| | - Madeline A Vaji
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487-0344, USA
| | - Ryan A Tuckey
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487-0344, USA
| | - Laura A Berkowitz
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487-0344, USA
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487-0344, USA.,Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center for Research on the Basic Biology of Aging, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487-0344, USA .,Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center for Research on the Basic Biology of Aging, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| |
Collapse
|
28
|
Nitric oxide mediated redox regulation of protein homeostasis. Cell Signal 2018; 53:348-356. [PMID: 30408515 DOI: 10.1016/j.cellsig.2018.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Nitric oxide is a versatile diffusible signaling molecule, whose biosynthesis by three NO synthases (NOS) is tightly regulated at transcriptional and posttranslational levels, availability of co-factors, and calcium binding. Above normal levels of NO have beneficial protective effects for example in the cardiovascular system, but also contribute to the pathophysiology in the context of inflammatory diseases, and to aging and neurodegeneration in the nervous system. The effect specificity relies on the functional and spatial specificity of the NOS isoenzymes, and on the duality of two major signaling mechanisms (i) activation of soluble guanylycylase (sGC)-dependent cGMP production and (ii) direct S-nitrosylation of redox sensitive cysteines of susceptible proteins. The present review summarizes the functional implications of S-nitrosylation in the context of proteostasis, and focuses on two NO target proteins, heat shock cognate of 70 kDa (Hsc70/HSPA8) and the ubiquitin 2 ligase (UBE2D), because both are modified on functionally critical cysteines and are key regulators of chaperone mediated and assisted autophagy and proteasomal protein degradation. SNO modifications of these candidates are associated with protein accumulations and adoption of a senescent phenotype of neuronal cells suggesting that S-nitrosylations of protein homeostatic machineries contribute to aging phenomena.
Collapse
|
29
|
Shan Y, Sun S, Yang F, Shang N, Liu H. Dexmedetomidine protects the developing rat brain against the neurotoxicity wrought by sevoflurane: role of autophagy and Drp1-Bax signaling. Drug Des Devel Ther 2018; 12:3617-3624. [PMID: 30464393 PMCID: PMC6214411 DOI: 10.2147/dddt.s180343] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The effect of sevoflurane on the nervous system is controversial. As an adjuvant anesthetic, dexmedetomidine has a protective role in various nerve-injury diseases. We investigated the effect of dexmedetomidine on injury to the developing brain induced by sevoflurane anesthesia, and if autophagy and mitochondrial damage are involved in the neuroprotective effects of dexmedetomidine. METHODS Pregnant rats on gestational day 20 were exposed to 3% sevoflurane for 4 hours. Saline and dexmedetomidine were injected intraperitoneally 15 minutes before exposure to sevoflurane or control gas. Bilateral hippocampi were harvested on postnatal day 1. Hippocampal morphology was observed by Nissl staining and expression of the microtubule-related protein LC3I/II, p62, Drp1, Bax, and Bcl2 were evaluated by Western blotting and immunohistochemistry. RESULTS Nissl staining showed that sevoflurane anesthesia during the third trimester caused neuronal damage to the hippocampi of rat pups. Western blotting and immunohistochemistry showed that pregnant rats exposed to sevoflurane during the third trimester led to pups having increased expression of LC3 and p62, suggesting that sevoflurane blocked autophagic flow in the hippocampus. Expression of Drp1 and Bax was increased after sevoflurane exposure, whereas Bcl2 expression was downregulated. All these effects were alleviated by pretreatment with dexmedetomidine. CONCLUSION Sevoflurane exposure during the third trimester caused neurological injury to rat pups. Autophagy and abnormalities in mitochondrial dynamics were involved in this neurotoxic process and were antagonized by dexmedetomidine.
Collapse
Affiliation(s)
- Yangyang Shan
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, 110004, China,
| | - Shiwei Sun
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, 110004, China,
| | - Fan Yang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, 110004, China,
| | - Nan Shang
- Department of Respiration, No. 202 Hospital of PLA, Shenyang, 110003, China
| | - Hongtao Liu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, 110004, China, ,Correspondence: Hongtao Liu, Department of Anesthesiology, Shengjing Hospital, China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, China, Email
| |
Collapse
|
30
|
Caldwell KA, Thies JL, Caldwell GA. No Country for Old Worms: A Systematic Review of the Application of C. elegans to Investigate a Bacterial Source of Environmental Neurotoxicity in Parkinson's Disease. Metabolites 2018; 8:metabo8040070. [PMID: 30380609 PMCID: PMC6315381 DOI: 10.3390/metabo8040070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 10/21/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022] Open
Abstract
While progress has been made in discerning genetic associations with Parkinson's disease (PD), identifying elusive environmental contributors necessitates the application of unconventional hypotheses and experimental strategies. Here, we provide an overview of studies that we conducted on a neurotoxic metabolite produced by a species of common soil bacteria, Streptomyces venezuelae (S. ven), indicating that the toxicity displayed by this bacterium causes stress in diverse cellular mechanisms, such as the ubiquitin proteasome system and mitochondrial homeostasis. This dysfunction eventually leads to age and dose-dependent neurodegeneration in the nematode Caenorhabditis elegans. Notably, dopaminergic neurons have heightened susceptibility, but all of the neuronal classes eventually degenerate following exposure. Toxicity further extends to human SH-SY5Y cells, which also degenerate following exposure. Additionally, the neurons of nematodes expressing heterologous aggregation-prone proteins display enhanced metabolite vulnerability. These mechanistic analyses collectively reveal a unique metabolomic fingerprint for this bacterially-derived neurotoxin. In considering that epidemiological distinctions in locales influence the incidence of PD, we surveyed soils from diverse regions of Alabama, and found that exposure to ~30% of isolated Streptomyces species caused worm dopaminergic neurons to die. In addition to aging, one of the few established contributors to PD appears to be a rural lifestyle, where exposure to soil on a regular basis might increase the risk of interaction with bacteria producing such toxins. Taken together, these data suggest that a novel toxicant within the Streptomyces genus might represent an environmental contributor to the progressive neurodegeneration that is associated with PD.
Collapse
Affiliation(s)
- Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487, USA.
- Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center for Research on the Basic Biology of Aging, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA.
| | - Jennifer L Thies
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487, USA.
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487, USA.
- Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center for Research on the Basic Biology of Aging, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA.
| |
Collapse
|