1
|
Baysah CZ, Dohoney RA, Palanikumar L, Stillman NH, Penney AL, Sola AD, Paredes DA, Magzoub M, Kumar S. A Brain-Penetrating Foldamer Rescues Aβ Aggregation-Associated Alzheimer's Disease Phenotypes in In Vivo Models. ACS Chem Neurosci 2025; 16:1309-1322. [PMID: 40070152 DOI: 10.1021/acschemneuro.4c00753] [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: 04/03/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is the leading cause of dementia, affecting nearly 55 million people across the world. One of the central pathological factors associated with AD is the aggregation of Aβ42, a peptide product cleaved through pathological processes in AD. Under pathological conditions, Aβ42 aggregates into insoluble plaques in the brain and impairs the function of neurons, which contributes to the cognitive decline associated with AD. Therefore, the modulation of Aβ42 aggregation is considered a potential therapeutic intervention for AD. Using an Oligoquinoline-based foldamer library, we have identified a potent foldamer antagonist (SK-131) of Aβ42 aggregation. SK-131 inhibits the aggregation by inducing a α-helical structure in monomeric Aβ42. Here, we demonstrated that SK-131 rescues Aβ42 aggregation-associated phenotypes in AD cellular and multiple Caenorhabditis elegans AD models, including intracellular inhibition of Aβ42 aggregation, rescue of behavioral deficits, and attenuation of reactive oxygen species. It efficiently crosses the blood-brain barrier and demonstrates favorable pharmaceutical properties. It also potently inhibits Zn2+-mediated Aβ42 aggregation by potentially displacing Zn2+ from Aβ42. In summary, we have identified a potent brain-penetrating foldamer that efficiently rescues AD phenotypes in in vivo models. Unlike most of the therapeutic approaches that target Aβ aggregates, we have identified and validated a novel therapeutic pathway by inducing structural change in Aβ and rescuing AD phenotypes in in vivo models. This study will further aid in the quest to identify lead therapeutics to slow or stop the progression of AD.
Collapse
Affiliation(s)
- Charles Zuwu Baysah
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
| | - Ryan A Dohoney
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
| | - L Palanikumar
- Biology Program, Division of Science, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, UAE
| | - Nicholas H Stillman
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
| | - Alexandra L Penney
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
- Department of Biological Sciences, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
| | - Andres D Sola
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
| | - Daniel A Paredes
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- Ritchie School of Engineering and Computer Science, University of Denver, 2155 E Wesley Ave, Denver, Colorado 80210, United States
| | - Mazin Magzoub
- Biology Program, Division of Science, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, UAE
| | - Sunil Kumar
- Department of Chemistry and Biochemistry, University of Denver, F.W. Olin Hall, 2190 E Iliff Ave, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Ave, Suite 579, Denver, Colorado 80208, United States
- Molecular and Cellular Biophysics Program, University of Denver, Boettcher West, Room 228, 2050 E. Iliff Ave, Denver, Colorado 80210, United States
| |
Collapse
|
2
|
Mickiewicz M, Nowek Z, Czopowicz M, Moroz-Fik A, Potărniche AV, Biernacka K, Szaluś-Jordanow O, Górski P, Antonopoulos A, Markowska-Daniel I, Várady M, Kaba J. Inhibitory effect of dimethyl sulfoxide on the development of gastrointestinal nematode larvae in the larval development test. J Vet Res 2025; 69:83-90. [PMID: 40144061 PMCID: PMC11936094 DOI: 10.2478/jvetres-2025-0016] [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: 09/12/2024] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction Dimethyl sulfoxide (DMSO) is an amphipathic solvent for molecules in in vitro tests for detection of anthelmintic resistance of gastrointestinal nematodes (GIN). It has been shown to have a concentration-dependent detrimental effect on Caenorhabditis elegans, a free-living nematode. If GIN are likewise affected, using DMSO in egg-hatch test and larval development test (LDT) may confound their results. Therefore, the DMSO concentration was determined at which it exerted an inhibitory effect on GIN larval development to the third stage. Material and Methods A standard LDT was performed in 30 replications at DMSO concentrations of 0.0% (control), 0.6%, 1.3%, 2.6%, 5.2%, 10.4%, and 20.8%. The numbers of all developmental stages of Haemonchus contortus, Trichostrongylus spp. and Oesophagostomum spp. (unhatched eggs, larvae of the first, second and third stages (L1-L3) were determined, the proportion of L3 (the percentage of larval development - PD) was calculated and L3 were identified at the species or genus level. A five-parameter logistic curve was fitted to the observed PDs and modelled the DMSO-larval development relationship. Results The PD significantly decreased with increasing DMSO concentration and was significantly reduced at the 2.6% concentration. The median inhibitory concentration (IC50) was 3.79%, the concentration for 10% inhibition (IC10) was 1.75% and for 90% inhibition (IC90) was 8.20%. The percentage of L1 and L2 followed an analogical but opposite pattern to that of PD and was complementary to it at each DMSO concentration. The unhatched egg percentage was rarely >1% and showed no pattern. Conclusion At ≥2.6% concentration, DMSO significantly inhibited the L3 development of all three GIN species. It had a practically important inhibitory effect (IC10) at as low concentration as 1.75%. At lower concentrations, DMSO did not appear to inhibit larval development. The compound did not seem to exert an in vitro ovicidal effect regardless of the concentration.
Collapse
Affiliation(s)
| | - Zofia Nowek
- Division of Veterinary Epidemiology and Economics
| | | | | | - Adrian-Valentin Potărniche
- Department of Infectious Diseases and Preventive Medicine, Law and Ethics, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca400372, Romania
| | | | - Olga Szaluś-Jordanow
- Department of Small Animal Diseases with Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences–SGGW, 02-776Warsaw, Poland
| | - Paweł Górski
- Division of Parasitology and Invasiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences–SGGW, 02-786Warsaw, Poland
| | | | | | - Marián Várady
- Institute of Parasitology, Slovak Academy of Sciences, 04001Košice, Slovakia
| | | |
Collapse
|
3
|
Kwon Y, Kim J, Son YB, Lee SA, Choi SS, Cho Y. Advanced Neural Functional Imaging in C. elegans Using Lab-on-a-Chip Technology. MICROMACHINES 2024; 15:1027. [PMID: 39203678 PMCID: PMC11356251 DOI: 10.3390/mi15081027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024]
Abstract
The ability to perceive and adapt to environmental changes is crucial for the survival of all organisms. Neural functional imaging, particularly in model organisms, such as Caenorhabditis elegans, provides valuable insights into how animals sense and process external cues through their nervous systems. Because of its fully mapped neural anatomy, transparent body, and genetic tractability, C. elegans serves as an ideal model for these studies. This review focuses on advanced methods for neural functional imaging in C. elegans, highlighting calcium imaging techniques, lab-on-a-chip technologies, and their applications in the study of various sensory modalities, including chemosensation, mechanosensation, thermosensation, photosensation, and magnetosensation. We discuss the benefits of these methods in terms of precision, reproducibility, and ability to study dynamic neural processes in real time, ultimately advancing our understanding of the fundamental principles of neural activity and connectivity.
Collapse
Affiliation(s)
- Youngeun Kwon
- Department of Chemical Engineering, Myongji University, Yongin 17058, Republic of Korea; (Y.K.); (J.K.); (Y.B.S.)
| | - Jihye Kim
- Department of Chemical Engineering, Myongji University, Yongin 17058, Republic of Korea; (Y.K.); (J.K.); (Y.B.S.)
| | - Ye Bin Son
- Department of Chemical Engineering, Myongji University, Yongin 17058, Republic of Korea; (Y.K.); (J.K.); (Y.B.S.)
| | - Sol Ah Lee
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Shin Sik Choi
- Department of Bio-Pharmaceutical Sciences, Myongji University, Yongin 17058, Republic of Korea;
- The Natural Science Research Institute, Department of Food and Nutrition, Myongji University, Yongin 17058, Republic of Korea
- elegslab Inc., Seoul 06083, Republic of Korea
| | - Yongmin Cho
- Department of Chemical Engineering, Myongji University, Yongin 17058, Republic of Korea; (Y.K.); (J.K.); (Y.B.S.)
- elegslab Inc., Seoul 06083, Republic of Korea
| |
Collapse
|
4
|
Holcom A, Fuentealba M, Sivapatham R, King CD, Osman H, Foulger A, Bhaumik D, Schilling B, Furman D, Andersen JK, Lithgow GJ. Neuronal expression of human amyloid-β and Tau drives global phenotypic and multi-omic changes in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.01.542377. [PMID: 37398058 PMCID: PMC10312529 DOI: 10.1101/2023.06.01.542377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Alzheimer's disease (AD) and Alzheimer's related diseases (ADRD) are prevalent age-related neurodegenerative disorders characterized by the accumulation of amyloid-β (Aβ) plaques and Tau neurofibrillary tangles. The nematode Caenorhabditis elegan s ( C. elegans ) serves as an invaluable model organism in diseases of old age-due to its rapid aging. Here we performed an unbiased systems analysis of a C. elegans strain expressing both Aβ and Tau proteins within neurons. We set out to determine if there was a phenotypic interaction between Aβ and Tau. In addition, we were interested in determining the temporal order of the phenotypic and multi-omic (geromic) outcomes. At an early stage of adulthood, we observed reproductive impairments and mitochondrial dysfunction consistent with disruptions in mRNA transcript abundance, protein solubility, and metabolite levels. Notably, the expression of these neurotoxic proteins exhibited a synergistic effect, leading to accelerated aging. Our findings shed light on the close relationship between normal aging and ADRD. Specifically, we demonstrate alterations to metabolic functions preceding age-related neurotoxicity, offering a resource for the development of new therapeutic strategies.
Collapse
|
5
|
Wang L, Lin H, Zhu Y, Ge X, Li M, Liu J, Chen F, Zhang M, Cheng JX. Overtone photothermal microscopy for high-resolution and high-sensitivity vibrational imaging. Nat Commun 2024; 15:5374. [PMID: 38918400 PMCID: PMC11199576 DOI: 10.1038/s41467-024-49691-2] [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: 06/30/2023] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
Photothermal microscopy is a highly sensitive pump-probe method for mapping nanostructures and molecules through the detection of local thermal gradients. While visible photothermal microscopy and mid-infrared photothermal microscopy techniques have been developed, they possess inherent limitations. These techniques either lack chemical specificity or encounter significant light attenuation caused by water absorption. Here, we present an overtone photothermal (OPT) microscopy technique that offers high chemical specificity, detection sensitivity, and spatial resolution by employing a visible probe for local heat detection in the C-H overtone region. We demonstrate its capability for high-fidelity chemical imaging of polymer nanostructures, depth-resolved intracellular chemical mapping of cancer cells, and imaging of multicellular C. elegans organisms and highly scattering brain tissues. By bridging the gap between visible and mid-infrared photothermal microscopy, OPT establishes a new modality for high-resolution and high-sensitivity chemical imaging. This advancement complements large-scale shortwave infrared imaging approaches, facilitating multiscale structural and chemical investigations of materials and biological metabolism.
Collapse
Affiliation(s)
- Le Wang
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| | - Haonan Lin
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| | - Yifan Zhu
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Xiaowei Ge
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| | - Mingsheng Li
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| | - Jianing Liu
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| | - Fukai Chen
- Department of Biology, Boston University, Boston, MA, 02215, USA
| | - Meng Zhang
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| | - Ji-Xin Cheng
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA.
- Department of Chemistry, Boston University, Boston, MA, 02215, USA.
- Department of Biology, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
6
|
Tan X, Xu R, Li AP, Li D, Wang Y, Zhao Q, Long LP, Fan YZ, Zhao CX, Liu Y, Li SH. Antioxidant and anti-Alzheimer's disease activities of 1,8-cineole and its cyclodextrin inclusion complex. Biomed Pharmacother 2024; 175:116784. [PMID: 38781865 DOI: 10.1016/j.biopha.2024.116784] [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: 02/28/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
1,8-Cineole is a bicyclic monoterpene widely distributed in the essential oils of various medicinal plants, and it exhibits significant anti-inflammatory and antioxidant activities. We aimed to investigate the therapeutic effect of 1,8-cineole on anti-Alzheimer's disease by using transgenic Caenorhabditis elegans models. Our studies demonstrated that 1,8-cineole significantly relieved Aβ1-42-induced paralysis and exhibited remarkable antioxidant and anti-Aβ1-42 aggregation activities in transgenic nematodes CL4176, CL2006 and CL2355. We developed a 1,8-cineole/cyclodextrin inclusion complex, displaying enhanced anti-paralysis, anti-Aβ aggregation and antioxidant activities compared to 1,8-cineole. In addition, we found 1,8-cineole treatment activated the SKN-1/Nrf-2 pathway and induced autophagy in nematodes. Our results demonstrated the antioxidant and anti-Alzheimer's disease activities of 1,8-cineole, which provide a potential therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Xin Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Rui Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Ai-Pei Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Yun Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Li-Ping Long
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Yu-Zhou Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Chen-Xiao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Yan Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| | - Sheng-Hong Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, and Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China; State Key Laboratory of Phytochemistry and Plant Resources in West China and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, PR China.
| |
Collapse
|
7
|
Bellver-Sanchis A, Geng Q, Navarro G, Ávila-López PA, Companys-Alemany J, Marsal-García L, Larramona-Arcas R, Miró L, Perez-Bosque A, Ortuño-Sahagún D, Banerjee DR, Choudhary BS, Soriano FX, Poulard C, Pallàs M, Du HN, Griñán-Ferré C. G9a Inhibition Promotes Neuroprotection through GMFB Regulation in Alzheimer's Disease. Aging Dis 2024; 15:311-337. [PMID: 37307824 PMCID: PMC10796087 DOI: 10.14336/ad.2023.0424-2] [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: 01/22/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023] Open
Abstract
Epigenetic alterations are a fundamental pathological hallmark of Alzheimer's disease (AD). Herein, we show the upregulation of G9a and H3K9me2 in the brains of AD patients. Interestingly, treatment with a G9a inhibitor (G9ai) in SAMP8 mice reversed the high levels of H3K9me2 and rescued cognitive decline. A transcriptional profile analysis after G9ai treatment revealed increased gene expression of glia maturation factor β (GMFB) in SAMP8 mice. Besides, a H3K9me2 ChIP-seq analysis after G9a inhibition treatment showed the enrichment of gene promoters associated with neural functions. We observed the induction of neuronal plasticity and a reduction of neuroinflammation after G9ai treatment, and more strikingly, these neuroprotective effects were reverted by the pharmacological inhibition of GMFB in mice and cell cultures; this was also validated by the RNAi approach generating the knockdown of GMFB/Y507A.10 in Caenorhabditis elegans. Importantly, we present evidence that GMFB activity is controlled by G9a-mediated lysine methylation as well as we identified that G9a directly bound GMFB and catalyzed the methylation at lysine (K) 20 and K25 in vitro. Furthermore, we found that the neurodegenerative role of G9a as a GMFB suppressor would mainly rely on methylation of the K25 position of GMFB, and thus G9a pharmacological inhibition removes this methylation promoting neuroprotective effects. Then, our findings confirm an undescribed mechanism by which G9a inhibition acts at two levels, increasing GMFB and regulating its function to promote neuroprotective effects in age-related cognitive decline.
Collapse
Affiliation(s)
- Aina Bellver-Sanchis
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Qizhi Geng
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, RNA Institute, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Department Biochemistry and Physiology, Faculty of Pharmacy. Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Pedro A. Ávila-López
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| | - Júlia Companys-Alemany
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Laura Marsal-García
- Department of Biochemistry, McGill University, Montréal, Québec, Canada.
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Québec, Canada.
| | - Raquel Larramona-Arcas
- Department of Cell Biology, Physiology, and Immunology, Celltec-UB, University of Barcelona, Barcelona, Spain, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Lluisa Miró
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Anna Perez-Bosque
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunología Molecular, Instituto de Investigación de Ciencias Biomédicas (IICB) CUCS, Universidad de Guadalajara, Jalisco 44340, México.
| | | | - Bhanwar Singh Choudhary
- Department of Pharmacy, Central University of Rajasthan, Ajmer, Rajasthan, India.
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana, Gujarat, India.
| | - Francesc X Soriano
- Department of Cell Biology, Physiology, and Immunology, Celltec-UB, University of Barcelona, Barcelona, Spain, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Coralie Poulard
- Cancer Research Cancer Lyon, Université de Lyon, F-69000 Lyon, France.
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.
- CNRS UMR5286, Centre de Recherche en Cancérlogie de Lyon, F-69000 Lyon, France.
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Hai-Ning Du
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, RNA Institute, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
8
|
Kozin SA, Kechko OI, Adzhubei AA, Makarov AA, Mitkevich VA. Switching On/Off Amyloid Plaque Formation in Transgenic Animal Models of Alzheimer's Disease. Int J Mol Sci 2023; 25:72. [PMID: 38203242 PMCID: PMC10778642 DOI: 10.3390/ijms25010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
A hallmark of Alzheimer's disease (AD) are the proteinaceous aggregates formed by the amyloid-beta peptide (Aβ) that is deposited inside the brain as amyloid plaques. The accumulation of aggregated Aβ may initiate or enhance pathologic processes in AD. According to the amyloid hypothesis, any agent that has the capability to inhibit Aβ aggregation and/or destroy amyloid plaques represents a potential disease-modifying drug. In 2023, a humanized IgG1 monoclonal antibody (lecanemab) against the Aβ-soluble protofibrils was approved by the US FDA for AD therapy, thus providing compelling support to the amyloid hypothesis. To acquire a deeper insight on the in vivo Aβ aggregation, various animal models, including aged herbivores and carnivores, non-human primates, transgenic rodents, fish and worms were widely exploited. This review is based on the recent data obtained using transgenic animal AD models and presents experimental verification of the critical role in Aβ aggregation seeding of the interactions between zinc ions, Aβ with the isomerized Asp7 (isoD7-Aβ) and the α4β2 nicotinic acetylcholine receptor.
Collapse
Affiliation(s)
- Sergey A. Kozin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.I.K.); (A.A.A.); (A.A.M.)
| | | | | | | | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.I.K.); (A.A.A.); (A.A.M.)
| |
Collapse
|
9
|
Kakraba S, Ayyadevara S, Mainali N, Balasubramaniam M, Bowroju S, Penthala NR, Atluri R, Barger SW, Griffin ST, Crooks PA, Shmookler Reis RJ. Thiadiazolidinone (TDZD) Analogs Inhibit Aggregation-Mediated Pathology in Diverse Neurodegeneration Models, and Extend C. elegans Life- and Healthspan. Pharmaceuticals (Basel) 2023; 16:1498. [PMID: 37895969 PMCID: PMC10610358 DOI: 10.3390/ph16101498] [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: 08/25/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
Chronic, low-grade inflammation has been implicated in aging and age-dependent conditions, including Alzheimer's disease, cardiomyopathy, and cancer. One of the age-associated processes underlying chronic inflammation is protein aggregation, which is implicated in neuroinflammation and a broad spectrum of neurodegenerative diseases such as Alzheimer's, Huntington's, and Parkinson's diseases. We screened a panel of bioactive thiadiazolidinones (TDZDs) from our in-house library for rescue of protein aggregation in human-cell and C. elegans models of neurodegeneration. Among the tested TDZD analogs, PNR886 and PNR962 were most effective, significantly reducing both the number and intensity of Alzheimer-like tau and amyloid aggregates in human cell-culture models of pathogenic aggregation. A C. elegans strain expressing human Aβ1-42 in muscle, leading to AD-like amyloidopathy, developed fewer and smaller aggregates after PNR886 or PNR962 treatment. Moreover, age-progressive paralysis was reduced 90% by PNR886 and 75% by PNR962, and "healthspan" (the median duration of spontaneous motility) was extended 29% and 62%, respectively. These TDZD analogs also extended wild-type C. elegans lifespan by 15-30% (p < 0.001), placing them among the most effective life-extension drugs. Because the lead drug in this family, TDZD-8, inhibits GSK3β, we used molecular-dynamic tools to assess whether these analogs may also target GSK3β. In silico modeling predicted that PNR886 or PNR962 would bind to the same allosteric pocket of inactive GSK3β as TDZD-8, employing the same pharmacophore but attaching with greater avidity. PNR886 and PNR962 are thus compelling candidate drugs for treatment of tau- and amyloid-associated neurodegenerative diseases such as AD, potentially also reducing all-cause mortality.
Collapse
Affiliation(s)
- Samuel Kakraba
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Srinivas Ayyadevara
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Nirjal Mainali
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Meenakshisundaram Balasubramaniam
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Suresh Bowroju
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.B.); (N.R.P.); (P.A.C.)
| | - Narsimha Reddy Penthala
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.B.); (N.R.P.); (P.A.C.)
| | - Ramani Atluri
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Steven W. Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Sue T. Griffin
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Peter A. Crooks
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.B.); (N.R.P.); (P.A.C.)
| | - Robert J. Shmookler Reis
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| |
Collapse
|
10
|
Long T, Chen X, Qin DL, Zhu YF, Zhou YJ, He YN, Fu HJ, Tang Y, Yu L, Huang FH, Wang L, Yu CL, Law BYK, Wu JM, Wu AG, Zhou XG. Ameliorative effect of Luffa cylindrica fruits on Caenorhabditis elegans and cellular models of Alzheimer's disease-related pathology via autophagy induction. Phytother Res 2023; 37:4639-4654. [PMID: 37394882 DOI: 10.1002/ptr.7931] [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: 02/27/2023] [Revised: 06/03/2023] [Accepted: 06/16/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a prevalent neurodegenerative disorder without an effective cure. Natural products, while showing promise as potential therapeutics for AD, remain underexplored. AIMS This study was conducted with the goal of identifying potential anti-AD candidates from natural sources using Caenorhabditis elegans (C. elegans) AD-like models and exploring their mechanisms of action. MATERIALS & METHODS Our laboratory's in-house herbal extract library was utilized to screen for potential anti-AD candidates using the C. elegans AD-like model CL4176. The neuroprotective effects of the candidates were evaluated in multiple C. elegans AD-like models, specifically targeting Aβ- and Tau-induced pathology. In vitro validation was conducted using PC-12 cells. To investigate the role of autophagy in mediating the anti-AD effects of the candidates, RNAi bacteria and autophagy inhibitors were employed. RESULTS The ethanol extract of air-dried fruits of Luffa cylindrica (LCE), a medicine-food homology species, was found to inhibit Aβ- and Tau-induced pathology (paralysis, ROS production, neurotoxicity, and Aβ and pTau deposition) in C. elegans AD-like models. LCE was non-toxic and enhanced C. elegans' health. It was shown that LCE activates autophagy and its anti-AD efficacy is weakened with the RNAi knockdown of autophagy-related genes. Additionally, LCE induced mTOR-mediated autophagy, reduced the expression of AD-associated proteins, and decreased cell death in PC-12 cells, which was reversed by autophagy inhibitors (bafilomycin A1 and 3-methyladenine). DISCUSSION LCE, identified from our natural product library, emerged as a valuable autophagy enhancer that effectively protects against neurodegeneration in multiple AD-like models. RNAi knockdown of autophagy-related genes and cotreatment with autophagy inhibitors weakened its anti-AD efficacy, implying a critical role of autophagy in mediating the neuroprotective effects of LCE. CONCLUSION Our findings highlight the potential of LCE as a functional food or drug for targeting AD pathology and promoting human health.
Collapse
Grants
- 81903829 National Natural Science Foundation of China
- 81801398 National Natural Science Foundation of China
- 2022YFS0620 The Science and Technology Planning Project of Sichuan Province, China
- 2020YJ0494 The Science and Technology Planning Project of Sichuan Province, China
- 2021YJ0180 The Science and Technology Planning Project of Sichuan Province, China
- 2020LZXNYDJ37 The Joint Project of Luzhou Municipal People's Government and Southwest Medical University, China
- 20YKDYYJC0067 The Joint Project of Luzhou Municipal People's Government and Southwest Medical University, China
- 2019ZQN174 The project of Southwest Medical University, China
- 2021ZKZD018 The project of Southwest Medical University, China
- 2021ZKMS046 The project of Southwest Medical University, China
- 2020ZRZD015 The project of Southwest Medical University, China
- 2021ZKZD015 The project of Southwest Medical University, China
- MUST-SKL-2021-005 The Macao Science and Technology Development Fund of Macao SAR
- SKL-QRCM (MUST)-2020-2022 The Macao Science and Technology Development Fund of Macao SAR
- 81903829 the National Natural Science Foundation of China
- 81801398 the National Natural Science Foundation of China
Collapse
Affiliation(s)
- Tao Long
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Xue Chen
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yun-Fei Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Yu-Jia Zhou
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yan-Ni He
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Hai-Jun Fu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Fei-Hong Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Long Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| |
Collapse
|
11
|
Dohoney RA, Joseph JA, Baysah C, Thomas AG, Siwakoti A, Ball TD, Kumar S. "Common-Precursor" Protein Mimetic Approach to Rescue Aβ Aggregation-Mediated Alzheimer's Phenotypes. ACS Chem Biol 2023. [PMID: 37367833 DOI: 10.1021/acschembio.3c00120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Abberent protein-protein interactions (aPPIs) are associated with an array of pathological conditions, which make them important therapeutic targets. The aPPIs are mediated via specific chemical interactions that spread over a large and hydrophobic surface. Therefore, ligands that can complement the surface topography and chemical fingerprints could manipulate aPPIs. Oligopyridylamides (OPs) are synthetic protein mimetics that have been shown to manipulate aPPIs. However, the previous OP library used to disrupt these aPPIs was moderate in number (∼30 OPs) with very limited chemical diversity. The onus is on the laborious and time-consuming synthetic pathways with multiple chromatography steps. We have developed a novel chromatography-free technique to synthesize a highly diverse chemical library of OPs using a "common-precursor" approach. We significantly expanded the chemical diversity of OPs using a chromatography-free high-yielding method. To validate our novel approach, we have synthesized an OP with identical chemical diversity to a pre-existing OP-based potent inhibitor of Aβ aggregation, a process central to Alzheimer's disease (AD). The newly synthesized OP ligand (RD242) was very potent in inhibiting Aβ aggregation and rescuing AD phenotypes in an in vivo model. Moreover, RD242 was very effective in rescuing AD phenotypes in a post-disease onset AD model. We envision that our "common-precursor" synthetic approach will have tremendous potential as it is expandable for other oligoamide scaffolds to enhance affinity for disease-relevant targets.
Collapse
Affiliation(s)
- Ryan A Dohoney
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Johnson A Joseph
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Charles Baysah
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Alexandra G Thomas
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
- The Department of Biological Sciences, University of Denver, Denver, Colorado 80210, United States
| | - Apshara Siwakoti
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
- The Department of Biological Sciences, University of Denver, Denver, Colorado 80210, United States
| | - Tyler D Ball
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Sunil Kumar
- The Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| |
Collapse
|
12
|
Barker E, Morgan A, Barclay JW. A Caenorhabditis elegans model of autosomal dominant adult-onset neuronal ceroid lipofuscinosis identifies ethosuximide as a potential therapeutic. Hum Mol Genet 2023; 32:1772-1785. [PMID: 36282524 PMCID: PMC10196665 DOI: 10.1093/hmg/ddac263] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 09/22/2023] Open
Abstract
Autosomal dominant adult-onset neuronal ceroid lipofuscinosis (ANCL) is a rare neurodegenerative disorder characterized by progressive dementia and premature death. Four ANCL-causing mutations have been identified, all mapping to the DNAJC5 gene that encodes cysteine string protein α (CSPα). Here, using Caenorhabditis elegans, we describe an animal model of ANCL in which disease-causing mutations are introduced into their endogenous chromosomal locus, thereby mirroring the human genetic disorder. This was achieved through CRISPR/Cas9-mediated gene editing of dnj-14, the C. elegans ortholog of DNAJC5. The resultant homozygous ANCL mutant worms exhibited reduced lifespans and severely impaired chemotaxis, similar to isogenic dnj-14 null mutants. Importantly, these phenotypes were also seen in balanced heterozygotes carrying one wild-type and one ANCL mutant dnj-14 allele, mimicking the heterozygosity of ANCL patients. We observed a more severe chemotaxis phenotype in heterozygous ANCL mutant worms compared with haploinsufficient worms lacking one copy of CSP, consistent with a dominant-negative mechanism of action. Additionally, we provide evidence of CSP haploinsufficiency in longevity, as heterozygous null mutants exhibited significantly shorter lifespan than wild-type controls. The chemotaxis phenotype of dnj-14 null mutants was fully rescued by transgenic human CSPα, confirming the translational relevance of the worm model. Finally, a focused compound screen revealed that the anti-epileptic drug ethosuximide could restore chemotaxis in dnj-14 ANCL mutants to wild-type levels. This suggests that ethosuximide may have therapeutic potential for ANCL and demonstrates the utility of this C. elegans model for future larger-scale drug screening.
Collapse
Affiliation(s)
- Eleanor Barker
- Department of Molecular Physiology & Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 3BX, UK
| | - Alan Morgan
- Department of Molecular Physiology & Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 3BX, UK
| | - Jeff W Barclay
- Department of Molecular Physiology & Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 3BX, UK
| |
Collapse
|
13
|
Janizek JD, Spiro A, Celik S, Blue BW, Russell JC, Lee TI, Kaeberlin M, Lee SI. PAUSE: principled feature attribution for unsupervised gene expression analysis. Genome Biol 2023; 24:81. [PMID: 37076856 PMCID: PMC10114348 DOI: 10.1186/s13059-023-02901-4] [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: 04/30/2022] [Accepted: 03/17/2023] [Indexed: 04/21/2023] Open
Abstract
As interest in using unsupervised deep learning models to analyze gene expression data has grown, an increasing number of methods have been developed to make these models more interpretable. These methods can be separated into two groups: post hoc analyses of black box models through feature attribution methods and approaches to build inherently interpretable models through biologically-constrained architectures. We argue that these approaches are not mutually exclusive, but can in fact be usefully combined. We propose PAUSE ( https://github.com/suinleelab/PAUSE ), an unsupervised pathway attribution method that identifies major sources of transcriptomic variation when combined with biologically-constrained neural network models.
Collapse
Affiliation(s)
- Joseph D Janizek
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, USA
- Medical Scientist Training Program, University of Washington, Seattle, USA
| | - Anna Spiro
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, USA
| | | | - Ben W Blue
- Department of Pathology, University of Washington, Seattle, USA
| | - John C Russell
- Department of Pathology, University of Washington, Seattle, USA
| | - Ting-I Lee
- Department of Pathology, University of Washington, Seattle, USA
| | - Matt Kaeberlin
- Department of Pathology, University of Washington, Seattle, USA
- Department of Genome Sciences, University of Washington, Seattle, USA
| | - Su-In Lee
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, USA.
| |
Collapse
|
14
|
Caenorhabditis elegans as a Model System to Study Human Neurodegenerative Disorders. Biomolecules 2023; 13:biom13030478. [PMID: 36979413 PMCID: PMC10046667 DOI: 10.3390/biom13030478] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
In recent years, advances in science and technology have improved our quality of life, enabling us to tackle diseases and increase human life expectancy. However, longevity is accompanied by an accretion in the frequency of age-related neurodegenerative diseases, creating a growing burden, with pervasive social impact for human societies. The cost of managing such chronic disorders and the lack of effective treatments highlight the need to decipher their molecular and genetic underpinnings, in order to discover new therapeutic targets. In this effort, the nematode Caenorhabditis elegans serves as a powerful tool to recapitulate several disease-related phenotypes and provides a highly malleable genetic model that allows the implementation of multidisciplinary approaches, in addition to large-scale genetic and pharmacological screens. Its anatomical transparency allows the use of co-expressed fluorescent proteins to track the progress of neurodegeneration. Moreover, the functional conservation of neuronal processes, along with the high homology between nematode and human genomes, render C. elegans extremely suitable for the study of human neurodegenerative disorders. This review describes nematode models used to study neurodegeneration and underscores their contribution in the effort to dissect the molecular basis of human diseases and identify novel gene targets with therapeutic potential.
Collapse
|
15
|
Li Y, Li P, Zhang W, Zheng X, Gu Q. New Wine in Old Bottle: Caenorhabditis Elegans in Food Science. FOOD REVIEWS INTERNATIONAL 2023. [DOI: 10.1080/87559129.2023.2172429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Affiliation(s)
- Yonglu Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, People’s Republic of China
| | - Ping Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, People’s Republic of China
| | - Weixi Zhang
- Department of Food Science and Nutrition; Zhejiang Key Laboratory for Agro-food Processing; Fuli Institute of Food Science; National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition; Zhejiang Key Laboratory for Agro-food Processing; Fuli Institute of Food Science; National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou, People’s Republic of China
| | - Qing Gu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, People’s Republic of China
| |
Collapse
|
16
|
Kleawyothatis W, Jattujan P, Chumphoochai K, Chalorak P, Sobhon P, Meemon K. Holothuria scabra extracts confer neuroprotective effect in C. elegans model of Alzheimer's disease by attenuating amyloid-β aggregation and toxicity. J Tradit Complement Med 2023; 13:93-104. [PMID: 36685078 PMCID: PMC9845652 DOI: 10.1016/j.jtcme.2022.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 10/08/2022] [Accepted: 10/31/2022] [Indexed: 11/15/2022] Open
Abstract
Background and aim Alzheimer's disease (AD) is the most common aged-related neurodegenerative disorder that is associated with the toxic amyloid-β (Aβ) aggregation in the brain. While the efficacies of available drugs against AD are still limited, natural products have been shown to possess neuroprotective potential for prevention and therapy of AD. This study aimed to investigate the neuroprotective effects of H. scabra extracts against Aβ aggregation and proteotoxicity in C. elegans model of Alzheimer's diseases. Experimental procedure Whole bodies (WB) and body wall (BW) of H. scabra were extracted and fractionated into ethyl acetate (WBEA, BWEA), butanol (WBBU, BWBU), and ethanol (BWET). Then C. elegans AD models were treated with these fractions and investigated for Aβ aggregation and polymerization, biochemical and behavioral changes, and level of oxidative stress, as well as lifespan extension. Results and conclusion C. elegans AD model treated with H. scabra extracts, especially triterpene glycoside-rich ethyl acetate and butanol fractions, exhibited significant reduction of Aβ deposition. These H. scabra extracts also attenuated the paralysis behavior and improved the neurological defects in chemotaxis caused by Aβ aggregation. Immunoblot analysis revealed decreased level of Aβ oligomeric forms and the increased level of Aβ monomers after treatments with H. scabra extracts. In addition, H. scabra extracts reduced reactive oxygen species and increased the mean lifespan of the treated AD worms. In conclusion, this study demonstrated strong evidence of anti-Alzheimer effects by H. scabra extracts, implying that these extracts can potentially be applied as natural preventive and therapeutic agents for AD. Taxonomy classification by EVISE Alzheimer's disease, Neurodegenerative disorder, Traditional medicine, Experimental model systems, Molecular biology.
Collapse
Affiliation(s)
- Warannida Kleawyothatis
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Prapaporn Jattujan
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kawita Chumphoochai
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Pawanrat Chalorak
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
- Department of Radiological Technology and Medical Physics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Krai Meemon
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| |
Collapse
|
17
|
Kozin SA. Role of Interaction between Zinc and Amyloid Beta in Pathogenesis of Alzheimer’s Disease. BIOCHEMISTRY (MOSCOW) 2023; 88:S75-S87. [PMID: 37069115 DOI: 10.1134/s0006297923140055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Progression of Alzheimer's disease is accompanied by the appearance of extracellular deposits in the brain tissues of patients with characteristic supramolecular morphology (amyloid plaques) the main components of which are β-amyloid isoforms (Aβ) and biometal ions (zinc, copper, iron). For nearly 40 years and up to the present time, the vast majority of experimental data indicate critical role of formation and accumulation of amyloid plaques (cerebral amyloidogenesis) in pathogenesis of Alzheimer's disease, however, nature of the molecular agents that initiate cerebral amyloidogenesis, as well as causes of aggregation of the native Aβ molecules in vivo remained unknown for a long time. This review discusses the current level of fundamental knowledge about the molecular mechanisms of interactions of zinc ions with a number of Aβ isoforms present in amyloid plaques of the patients with Alzheimer's disease, and also shows how this knowledge made it possible to identify driving forces of the cerebral amyloidogenesis in Alzheimer's disease and made it possible to determine fundamentally new biomarkers and drug targets as part of development of innovative strategy for diagnosis and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Sergey A Kozin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
18
|
Zommiti M, Connil N, Tahrioui A, Groboillot A, Barbey C, Konto-Ghiorghi Y, Lesouhaitier O, Chevalier S, Feuilloley MGJ. Organs-on-Chips Platforms Are Everywhere: A Zoom on Biomedical Investigation. Bioengineering (Basel) 2022; 9:646. [PMID: 36354557 PMCID: PMC9687856 DOI: 10.3390/bioengineering9110646] [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: 08/24/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 08/28/2023] Open
Abstract
Over the decades, conventional in vitro culture systems and animal models have been used to study physiology, nutrient or drug metabolisms including mechanical and physiopathological aspects. However, there is an urgent need for Integrated Testing Strategies (ITS) and more sophisticated platforms and devices to approach the real complexity of human physiology and provide reliable extrapolations for clinical investigations and personalized medicine. Organ-on-a-chip (OOC), also known as a microphysiological system, is a state-of-the-art microfluidic cell culture technology that sums up cells or tissue-to-tissue interfaces, fluid flows, mechanical cues, and organ-level physiology, and it has been developed to fill the gap between in vitro experimental models and human pathophysiology. The wide range of OOC platforms involves the miniaturization of cell culture systems and enables a variety of novel experimental techniques. These range from modeling the independent effects of biophysical forces on cells to screening novel drugs in multi-organ microphysiological systems, all within microscale devices. As in living biosystems, the development of vascular structure is the salient feature common to almost all organ-on-a-chip platforms. Herein, we provide a snapshot of this fast-evolving sophisticated technology. We will review cutting-edge developments and advances in the OOC realm, discussing current applications in the biomedical field with a detailed description of how this technology has enabled the reconstruction of complex multi-scale and multifunctional matrices and platforms (at the cellular and tissular levels) leading to an acute understanding of the physiopathological features of human ailments and infections in vitro.
Collapse
Affiliation(s)
- Mohamed Zommiti
- Research Unit Bacterial Communication and Anti-infectious Strategies (CBSA, UR4312), University of Rouen Normandie, 27000 Evreux, France
| | | | | | | | | | | | | | | | - Marc G. J. Feuilloley
- Research Unit Bacterial Communication and Anti-infectious Strategies (CBSA, UR4312), University of Rouen Normandie, 27000 Evreux, France
| |
Collapse
|
19
|
Qin Y, Chen F, Tang Z, Ren H, Wang Q, Shen N, Lin W, Xiao Y, Yuan M, Chen H, Bu T, Li Q, Huang L. Ligusticum chuanxiong Hort as a medicinal and edible plant foods: Antioxidant, anti-aging and neuroprotective properties in Caenorhabditis elegans. Front Pharmacol 2022; 13:1049890. [PMID: 36386171 PMCID: PMC9643709 DOI: 10.3389/fphar.2022.1049890] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/18/2022] [Indexed: 02/05/2023] Open
Abstract
Ligusticum chuanxiong Hort. (CX) is a medicinal and edible plant including a variety of active substances, which may be an available resource for the treatment of related diseases. To expand the medicinal uses of CX, this study aims to explore the antioxidant, anti-aging and neuroprotective effects of the Ligusticum chuanxiong leaves (CXL) and rhizome (CXR) extracts. We first characterize CX phytochemical spectrum by LC-MS as well as antioxidant capacity. Acute toxicity, anti-oxidative stress capacity, lifespan and healthspan was evaluated in C elegans N2. Neuroprotective effect was evaluated in vitro and in vivo (C elegans CL4176 and CL2355). In this study, we detected 74 and 78 compounds from CXR and CXL, respectively, including phthalides, alkaloids, organic acids, terpenes, polyphenols and others. Furthermore, we found that CXs not only protect against oxidative stress, but also prolong the lifespan, alleviate lipofuscin, malondialdehyde (MDA) and reactive oxygen species (ROS) accumulation, and improve movement level, antioxidant enzyme activity in C elegans N2. However, only CXR reduced the β-amyloid peptide (Aβ)-induced paralysis phenotype in CL4176s and alleviated chemosensory behavior dysfunction in CL2355s. In addition, CXR treatment reduced the production of Aβ and ROS, enhanced SOD activity in CL4176s. The possible mechanism of anti-aging of CXL and CXR is to promote the expression of related antioxidant pathway genes, increase the activity of antioxidant enzymes, and reduce the accumulation of ROS, which is dependent on DAF-16 and HSF-1 (only in CXR). CXR was able to activate antioxidase-related (sod-3 and sod-5) and heat shock protein genes (hsp-16.1 and hsp-70) expression, consequently ameliorating proteotoxicity related to Aβ aggregation. In summary, these findings demonstrate the antioxidant, anti-aging and neuroprotective (only in CXR) activities of the CX, which provide an important pharmacological basis for developing functional foods and drugs to relieve the symptoms of aging and AD. However, the material basis of neuroprotective activity and antiaging effects need to be elucidated, and the relationship between these activities should also be clarified in future studies.
Collapse
Affiliation(s)
- Yihan Qin
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Fangfang Chen
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Zizhong Tang
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China,*Correspondence: Zizhong Tang,
| | - Hongjiao Ren
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Qing Wang
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Nayu Shen
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Wenjie Lin
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Yirong Xiao
- Sichuan Agricultural University Hospital, Sichuan Agricultural University, Ya’an, China
| | - Ming Yuan
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Hui Chen
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Tongliang Bu
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Qingfeng Li
- College of Life Sciences, Sichuan Agricultural University, Ya’an, China
| | - Lin Huang
- Triticeae Research Institute, Sichuan Agricultural University, Wenjiang, China
| |
Collapse
|
20
|
Raquel Ferreira Paulo I, Basílio de Oliveira Caland R, Orlando Muñoz Cadavid C, Martins Melo G, Soares De Castro Bezerra L, Pons E, Peña L, de Paula Oliveira R. β-carotene genetically-enriched lyophilized orange juice increases antioxidant capacity and reduces β-amyloid proteotoxicity and fat accumulation in Caenorhabditis elegans. FOOD CHEMISTRY. MOLECULAR SCIENCES 2022; 5:100141. [PMID: 36304081 PMCID: PMC9593878 DOI: 10.1016/j.fochms.2022.100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/01/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
Abstract
β-carotene content of genetically modified orange was 33-fold higher. β-carotene-enriched LOJ provided greater antioxidant capacity and stress resistance. β-carotene-enriched LOJ reduced β-amyloid proteotoxicity. β-carotene-enriched LOJ showed higher hypolipidemic activity in glucose rich diet.
Citrus sinensis orange juice is an excellent dietary source of β-carotene, a well-known antioxidant. However, β-carotene concentrations are relatively low in most cultivars. We developed a new orange through metabolic engineering strategy (GS) with 33.72-fold increase in β-carotene content compared to its conventional counterpart (CV). Using Caenorhabditis elegans, we found that animals treated with GS showed a greater reduction in intracellular reactive oxygen species (ROS) which is associated with a greater resistance to oxidative stress and induction of the expression of antioxidant genes. Moreover, animals treated with GS orange showed a more effective protection against β-amyloid proteotoxicity and greater hypolipidemic effect under high glucose diet compared to animals treated with CV. These data demonstrate that the increased amount of β-carotene in orange actually provides a greater beneficial effect in C. elegans and a valuable proof of principle to support further studies in mammals and humans.
Collapse
Affiliation(s)
| | - Ricardo Basílio de Oliveira Caland
- Instituto Federal de Educação, Ciência e Tecnologia do Piauí-IFPI, Brazil,Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | | | - Giovanna Martins Melo
- Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | | | - Elsa Pons
- Instituto de Biología Molecular y Celular de Plantas, Consejo Superior de Investigaciones Científicas, Universidad Politécnica de Valencia, Spain
| | - Leandro Peña
- Fundo de Defesa da Citricultura (Fundecitrus), Araraquara, SP, Brazil,Instituto de Biología Molecular y Celular de Plantas, Consejo Superior de Investigaciones Científicas, Universidad Politécnica de Valencia, Spain
| | - Riva de Paula Oliveira
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil,Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil,Corresponding author at: Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Natal, RN, Brazil.
| |
Collapse
|
21
|
Identification of a Hydroxygallic Acid Derivative, Zingibroside R1 and a Sterol Lipid as Potential Active Ingredients of Cuscuta chinensis Extract That Has Neuroprotective and Antioxidant Effects in Aged Caenorhabditis elegans. Nutrients 2022; 14:nu14194199. [PMID: 36235851 PMCID: PMC9570774 DOI: 10.3390/nu14194199] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 11/25/2022] Open
Abstract
We examined the effects of the extracts from two traditional Chinese medicine plants, Cuscuta chinensis and Eucommia ulmoides, on the healthspan of the model organism Caenorhabditis elegans. C. chinensis increased the short-term memory and the mechanosensory response of aged C. elegans. Furthermore, both extracts improved the resistance towards oxidative stress, and decreased the intracellular level of reactive oxygen species. Chemical analyses of the extracts revealed the presence of several bioactive compounds such as chlorogenic acid, cinnamic acid, and quercetin. A fraction from the C. chinensis extract enriched in zingibroside R1 improved the lifespan, the survival after heat stress, and the locomotion in a manner similar to the full C. chinensis extract. Thus, zingibroside R1 could be (partly) responsible for the observed health benefits of C. chinensis. Furthermore, a hydroxygallic acid derivative and the sterol lipid 4-alpha-formyl-stigmasta-7,24(241)-dien-3-beta-ol are abundantly present in the C. chinensis extract and its most bioactive fraction, but hardly in E. ulmoides, making them good candidates to explain the overall healthspan benefits of C. chinensis compared to the specific positive effects on stress resistance by E. ulmoides. Our findings highlight the overall anti-aging effects of C. chinensis in C. elegans and provide first hints about the components responsible for these effects.
Collapse
|
22
|
Ma L, Li X, Liu C, Yan W, Ma J, Petersen RB, Peng A, Huang K. Modelling Parkinson's Disease in C. elegans: Strengths and Limitations. Curr Pharm Des 2022; 28:3033-3048. [PMID: 36111767 DOI: 10.2174/1381612828666220915103502] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/08/2022] [Indexed: 01/28/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease that affects the motor system and progressively worsens with age. Current treatment options for PD mainly target symptoms, due to our limited understanding of the etiology and pathophysiology of PD. A variety of preclinical models have been developed to study different aspects of the disease. The models have been used to elucidate the pathogenesis and for testing new treatments. These models include cell models, non-mammalian models, rodent models, and non-human primate models. Over the past few decades, Caenorhabditis elegans (C. elegans) has been widely adopted as a model system due to its small size, transparent body, short generation time and life cycle, fully sequenced genome, the tractability of genetic manipulation and suitability for large scale screening for disease modifiers. Here, we review studies using C. elegans as a model for PD and highlight the strengths and limitations of the C. elegans model. Various C. elegans PD models, including neurotoxin-induced models and genetic models, are described in detail. Moreover, met.
Collapse
Affiliation(s)
- Liang Ma
- Department of Pharmacy, Wuhan Mental Health Center, Wuhan, China.,Department of Pharmacy, Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Xi Li
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanyao Yan
- Department of Pharmacy, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinlu Ma
- Human Resources Department, Wuhan Mental Health Center, Wuhan, China.,Human Resources Department, Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI, USA
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan, Tongren Hospital of Wuhan University, Wuhan, China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Wu KC, Chu PC, Cheng YJ, Li CI, Tian J, Wu HY, Wu SH, Lai YC, Kao HH, Hsu AL, Lin HW, Lin CH. Development of a traditional Chinese medicine-based agent for the treatment of cancer cachexia. J Cachexia Sarcopenia Muscle 2022; 13:2073-2087. [PMID: 35718751 PMCID: PMC9397559 DOI: 10.1002/jcsm.13028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 04/19/2022] [Accepted: 05/13/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Despite recent advances in understanding the pathophysiology of cancer cachexia, prevention/treatment of this debilitating disease remains an unmet medical need. METHODS We developed an integrated, multi-tiered strategy involving both in vitro and in vivo muscle atrophy platforms to identify traditional Chinese medicine (TCM)-based anti-cachectic agents. In the initial screening, we used inflammatory cytokine-induced atrophy of C2C12 myotubes as a phenotypic screening platform to assess the protective effects of TCMs. The selected TCMs were then evaluated for their abilities to protect Caenorhabditis elegans from age-related reduction of mobility and contractility, followed by the C-26 colon adenocarcinoma mouse model of cachexia to confirm the anti-muscle atrophy effects (body/skeletal muscle weights, fibre size distribution, grip strengths, and serum IL-6). Transcriptome analysis, quantitative real-time polymerase chain reaction, and immunoblotting were performed to gain understanding of the potential mechanism(s) by which effective TCM protected against C26 tumour-induced muscle atrophy. RESULTS Of 29 widely used TCMs, Dioscorea radix (DR) and Mu Dan Pi (MDP) showed a complete protection (all P values, 0.0002) vis-à-vis C26 conditioned medium control in the myotube atrophy platform. MDP exhibited a unique ability to ameliorate age-associated decreases in worm mobility, accompanied by improved total body contractions, relative to control (P < 0.0001 and <0.01, respectively), which, however, was not noted with DR. This differential in vivo protective effect between MDP and DR was also confirmed in the C-26 mouse model. MDP at 1000 mg/kg (MDP-H) was effective in protecting body weight loss (P < 0.05) in C-26 tumour-bearing mice without changing food or water intake, accompanied by the restoration of the fibre size distribution of hindleg skeletal muscles (P < 0.0001) and the forelimb grip strength (P < 0.05). MDP-treated C-26-tumour-bearing mice were alert, showed normal posture and better body conditions, and exhibited lower serum IL-6 levels (P = 0.06) relative to vehicle control. This decreased serum IL-6 was associated with the in vitro suppressive effect of MDP (25 and 50 μg/mL) on IL-6 secretion into culture medium by C26 cells. RNA-seq analysis, followed by quantitative real-time polymerase chain reaction and/or immunoblotting, shows that MDP's anti-cachectic effect was attributable to its ability to reverse the C-26 tumour-induced re-programming of muscle homoeostasis-associated gene expression, including that of two cachexia drivers (MuRF1 and Atrogin-1), in skeletal muscles. CONCLUSIONS All these findings suggest the translational potential of MDP to foster new strategies for the prevention and/or treatment of cachexia. The protective effect of MDP on other types of muscle atrophy such as sarcopenia might warrant investigations.
Collapse
Affiliation(s)
- Kun-Chang Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan.,Research Center for Healthy Aging, China Medical University, Taichung, Taiwan
| | - Po-Chen Chu
- Department of Cosmeceutics and Graduate Institute of Cosmeceutics, China Medical University, Taichung, Taiwan
| | - Yu-Jung Cheng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan.,Department of Rehabilitation, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Ing Li
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Jingkui Tian
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China.,College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Hsing-Yu Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Szu-Hsien Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan.,Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Yi-Chun Lai
- Research Center for Healthy Aging, China Medical University, Taichung, Taiwan
| | - Hsiang-Han Kao
- Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ao-Lin Hsu
- Research Center for Healthy Aging, China Medical University, Taichung, Taiwan.,PhD Program for Aging, China Medical University, Taichung, Taiwan.,Department of Internal Medicine, Division of Geriatrics & Palliative Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hsiang-Wen Lin
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan.,Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan.,Department of Pharmacy System, Outcomes and Policy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Chih-Hsueh Lin
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Geriatric Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
24
|
Weinkove D, Zavagno G. Applying C. elegans to the Industrial Drug Discovery Process to Slow Aging. FRONTIERS IN AGING 2022; 2:740582. [PMID: 35821999 PMCID: PMC9261450 DOI: 10.3389/fragi.2021.740582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/29/2021] [Indexed: 01/29/2023]
Abstract
The increase in our molecular understanding of the biology of aging, coupled with a recent surge in investment, has led to the formation of several companies developing pharmaceuticals to slow aging. Research using the tiny nematode worm Caenorhabditis elegans was the first to show that mutations in single genes can extend lifespan, and subsequent research has shown that this model organism is uniquely suited to testing interventions to slow aging. Yet, with a few notable exceptions, C. elegans is not in the standard toolkit of longevity companies. Here we discuss the paths to overcome the barriers to using C. elegans in industrial drug discovery. We address the predictive power of C. elegans for human aging, how C. elegans research can be applied to specific challenges in the typical drug discovery pipeline, and how standardised and quantitative assays will help C. elegans fulfil its potential in the biotech and pharmaceutical industry. We argue that correct application of this model and its knowledge base will significantly accelerate progress to slow human aging.
Collapse
Affiliation(s)
- David Weinkove
- Department of Biosciences, Durham University, Durham, United Kingdom.,Magnitude Biosciences Ltd., NETpark Plexus, Sedgefield, United Kingdom
| | - Giulia Zavagno
- Department of Biosciences, Durham University, Durham, United Kingdom.,Magnitude Biosciences Ltd., NETpark Plexus, Sedgefield, United Kingdom
| |
Collapse
|
25
|
Turcu AL, Companys-Alemany J, Phillips MB, Patel DS, Griñán-Ferré C, Loza MI, Brea JM, Pérez B, Soto D, Sureda FX, Kurnikova MG, Johnson JW, Pallàs M, Vázquez S. Design, synthesis, and in vitro and in vivo characterization of new memantine analogs for Alzheimer's disease. Eur J Med Chem 2022; 236:114354. [PMID: 35453065 PMCID: PMC9106868 DOI: 10.1016/j.ejmech.2022.114354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/28/2022]
Abstract
Currently, of the few accessible symptomatic therapies for Alzheimer's disease (AD), memantine is the only N-methyl-d-aspartate receptor (NMDAR) blocker approved by the FDA. This work further explores a series of memantine analogs featuring a benzohomoadamantane scaffold. Most of the newly synthesized compounds block NMDARs in the micromolar range, but with lower potency than previously reported hit IIc, results that were supported by molecular dynamics simulations. Subsequently, electrophysiological studies with the more potent compounds allowed classification of IIc, a low micromolar, uncompetitive, voltage-dependent, NMDAR blocker, as a memantine-like compound. The excellent in vitro DMPK properties of IIc made it a promising candidate for in vivo studies in Caenorhabditis elegans (C. elegans) and in the 5XFAD mouse model of AD. Administration of IIc or memantine improved locomotion and rescues chemotaxis behavior in C. elegans. Furthermore, both compounds enhanced working memory in 5XFAD mice and modified NMDAR and CREB signaling, which may prevent synaptic dysfunction and modulate neurodegenerative progression.
Collapse
Affiliation(s)
- Andreea L Turcu
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l'Alimentació i Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain; Neurophysiology Laboratory, Department of Biomedicine, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036, Barcelona, Spain
| | - Júlia Companys-Alemany
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neurosciences (NeuroUB), Universitat de Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
| | - Matthew B Phillips
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Dhilon S Patel
- Chemistry Department, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA, 15213, USA
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neurosciences (NeuroUB), Universitat de Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
| | - M Isabel Loza
- Innopharma Screening Platform, Biofarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidad de Santiago de Compostela, Edificio CIMUS, Av. Barcelona, S/N, E, 15706, Santiago de Compostela, Spain
| | - José M Brea
- Innopharma Screening Platform, Biofarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidad de Santiago de Compostela, Edificio CIMUS, Av. Barcelona, S/N, E, 15706, Santiago de Compostela, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutics and Toxicology, Autonomous University of Barcelona, E-08193, Bellaterra, Spain
| | - David Soto
- Neurophysiology Laboratory, Department of Biomedicine, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036, Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Francesc X Sureda
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201, Reus, Tarragona, Spain
| | - Maria G Kurnikova
- Chemistry Department, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA, 15213, USA
| | - Jon W Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neurosciences (NeuroUB), Universitat de Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l'Alimentació i Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain.
| |
Collapse
|
26
|
Claesson K, Chew YL, Ecroyd H. Exploiting flow cytometry for the unbiased quantification of protein inclusions in Caenorhabditis elegans. J Neurochem 2022; 161:281-292. [PMID: 35170035 PMCID: PMC9541147 DOI: 10.1111/jnc.15591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 01/28/2022] [Accepted: 02/12/2022] [Indexed: 11/29/2022]
Abstract
The aggregation of proteins into inclusions or plaques is a prominent hallmark of a diverse range of pathologies including neurodegenerative diseases. The quantification of such inclusions in Caenorhabditis elegans models of aggregation is usually achieved by fluorescence microscopy or other techniques involving biochemical fractionation of worm lysates. Here, we describe a simple and rapid flow cytometry-based approach that allows fluorescently tagged inclusions to be enumerated in whole worm lysate in a quantitative and unbiased fashion. We demonstrate that this technique is applicable to multiple C. elegans models of aggregation and importantly, can be used to monitor the dynamics of inclusion formation in response to heat shock and during ageing. This includes the characterisation of physicochemical properties of inclusions, such as their apparent size, which may reveal how aggregate formation is distinct in different tissues or at different stages of pathology or ageing. This new method can be used as a powerful technique for the medium- to high-throughput quantification of inclusions in future studies of genetic or chemical modulators of aggregation in C. elegans.
Collapse
Affiliation(s)
- Kristian Claesson
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health & Medical Research InstituteWollongongNew South WalesAustralia
| | - Yee Lian Chew
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health & Medical Research InstituteWollongongNew South WalesAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Heath Ecroyd
- Molecular Horizons and School of Chemistry and Molecular BioscienceUniversity of WollongongWollongongNSWAustralia
- Illawarra Health & Medical Research InstituteWollongongNew South WalesAustralia
| |
Collapse
|
27
|
Vozdek R, Pramstaller PP, Hicks AA. Functional Screening of Parkinson's Disease Susceptibility Genes to Identify Novel Modulators of α-Synuclein Neurotoxicity in Caenorhabditis elegans. Front Aging Neurosci 2022; 14:806000. [PMID: 35572147 PMCID: PMC9093606 DOI: 10.3389/fnagi.2022.806000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
Idiopathic Parkinson's disease (PD) is characterized by progressive loss of dopaminergic (DA) neurons during aging. The pathological hallmark of PD is the Lewy body detected in postmortem brain tissue, which is mainly composed of aggregated α-Synuclein (αSyn). However, it is estimated that 90% of PD cases have unknown pathogenetic triggers. Here, we generated a new transgenic Caenorhabditis elegans PD model eraIs1 expressing green fluorescent protein- (GFP-) based reporter of human αSyn in DA neurons, and exhibited a nice readout of the developed αSyn inclusions in DA neurons, leading to their degeneration during aging. Using these animals in a preliminary reverse genetic screening of >100-PD genome-wide association study- (GWAS-) based susceptibility genes, we identified 28 orthologs of C. elegans and their inactivation altered the phenotype of eraIs1; 10 knockdowns exhibited reduced penetrance of αSyn:Venus inclusions formed in the axons of cephalic (CEP) DA neurons, 18 knockdowns exhibited increased penetrance of disrupted CEP dendrite integrity among which nine knockdowns also exhibited disrupted neuronal morphology independent of the expressed αSyn reporter. Loss-of-function alleles of the five identified genes, such as sac-2, rig-6 or lfe-2, unc-43, and nsf-1, modulated the corresponding eraIs1 phenotype, respectively, and supported the RNA interference (RNAi) data. The Western blot analysis showed that the levels of insoluble αSyn:Venus were not correlated with the observed phenotypes in these mutants. However, RNAi of 12 identified modulators reduced the formation of pro-aggregating polyglutamine Q40:YFP foci in muscle cells, suggesting the possible role of these genes in cellular proteotoxicity. Therefore, modulators identified by their associated biological pathways, such as calcium signaling or vesicular trafficking, represent new potential therapeutic targets for neurodegenerative proteopathies and other diseases associated with aging.
Collapse
Affiliation(s)
- Roman Vozdek
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | | | | |
Collapse
|
28
|
Orientin Prolongs the Longevity of Caenorhabditis elegans and Postpones the Development of Neurodegenerative Diseases via Nutrition Sensing and Cellular Protective Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8878923. [PMID: 35237385 PMCID: PMC8885179 DOI: 10.1155/2022/8878923] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022]
Abstract
Age is the major risk factor for most of the deadliest diseases. Developing small molecule drugs with antiaging effects could improve the health of aged people and retard the onset and progress of aging-associated disorders. Bioactive secondary metabolites from medicinal plants are the main source for development of medication. Orientin is a water-soluble flavonoid monomer compound widely found in many medicinal plants. Orientin inhibits fat production, antioxidation, and anti-inflammatory activities. In this study, we explored whether orientin could affect the aging of C. elegans. We found that orientin improved heat, oxidative, and pathogenic stress resistances through activating stress responses, including HSF-1-mediated heat shock response, SKN-1-mediated xenobiotic and oxidation response, mitochondria unfolded responses, endoplasmic unfolded protein response, and increased autophagy activity. Orientin also could activate key regulators of the nutrient sensing pathway, including AMPK and insulin downstream transcription factor FOXO/DAF-16 to further improve the cellular health status. The above effects of orientin reduced the accumulation of toxic proteins (α-synuclein, β-amyloid, and poly-Q) and delayed the onset of neurodegenerative disorders in AD, PD, and HD models of C. elegans and finally increased the longevity and health span of C. elegans. Our results suggest that orientin has promising antiaging effects and could be a potential natural source for developing novel therapeutic drugs for aging and its related diseases.
Collapse
|
29
|
Modeling Alzheimer's Disease in Caenorhabditis elegans. Biomedicines 2022; 10:biomedicines10020288. [PMID: 35203497 PMCID: PMC8869312 DOI: 10.3390/biomedicines10020288] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is the most frequent cause of dementia. After decades of research, we know the importance of the accumulation of protein aggregates such as β-amyloid peptide and phosphorylated tau. We also know that mutations in certain proteins generate early-onset Alzheimer’s disease (EOAD), and many other genes modulate the disease in its sporadic form. However, the precise molecular mechanisms underlying AD pathology are still unclear. Because of ethical limitations, we need to use animal models to investigate these processes. The nematode Caenorhabditis elegans has received considerable attention in the last 25 years, since the first AD models overexpressing Aβ peptide were described. We review here the main results obtained using this model to study AD. We include works studying the basic molecular mechanisms of the disease, as well as those searching for new therapeutic targets. Although this model also has important limitations, the ability of this nematode to generate knock-out or overexpression models of any gene, single or combined, and to carry out toxicity, recovery or survival studies in short timeframes with many individuals and at low cost is difficult to overcome. We can predict that its use as a model for various diseases will certainly continue to increase.
Collapse
|
30
|
Tamagno WA, Santini W, Dos Santos A, Alves C, Bilibio D, Sutorillo NT, Zamberlan DC, Kaizer RR, Barcellos LJG. Pitaya fruit extract ameliorates the healthspan on copper-induced toxicity of Caenorhabditis elegans. J Food Biochem 2022; 46:e14050. [PMID: 34981523 DOI: 10.1111/jfbc.14050] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/10/2021] [Accepted: 11/26/2021] [Indexed: 12/19/2022]
Abstract
Copper (Cu) is an essential metal and it is important for metabolism. However, in high concentrations, it becomes toxic. Metal-induced toxicity is the cause of many neurodegenerative diseases. So it is necessary to search mechanisms to find ways of healthy aging. Natural compounds and diets based on fruits are increasingly common and could lead to a healthy life. Pitaya (Hylocereus undatus) is a tropical and Latin American, fruit that is gaining more popularity due to its antioxidant properties. Here, we evaluate the preventive and curative effect of different doses of microencapsulated pulp H. undatus extract on copper-induced toxicity. For this we use the nematode Caenorhabditis elegans, to investigate the effects of pitaya extract on behavior, lipid peroxidation, antioxidant chaperon, and cholinergic nervous system (ColNS). Results showed behavioral changes, decreased cell death biomarkers, and lipid peroxidation caused by copper, and these toxic effects were prevented and reverted by Pitaya's extract. After all, the extract can be used in diet as a supplement and studied to treat or prevent specific diseases, some of them linked to contamination and senility-related conditions. PRACTICAL APPLICATIONS: This research has been aimed to provide the uses of Hylocereus undatus microencapsulated pulp extract for the prevention and treatment of copper-induced toxicity. We have been shown that Pitaya is a good source of antioxidant compounds that can ameliorate the antioxidant system as well as the cholinergic nervous system avoiding behavior changes before and after the metal toxicity of copper. Therefore, the potential applications and common use of this extract can serve as food supplementation to prevent metal oxidative damage as well as to repair clinical cases of copper poisoning.
Collapse
Affiliation(s)
- Wagner Antonio Tamagno
- Biochemistry and Molecular Biology Laboratory, Federal Institute of Education, Science, and Technology of Rio Grande do Sul, Sertão, Rio Grande do Sul, Brazil
| | - Wallace Santini
- Biochemistry and Molecular Biology Laboratory, Federal Institute of Education, Science, and Technology of Rio Grande do Sul, Sertão, Rio Grande do Sul, Brazil
| | - Amanda Dos Santos
- Biochemistry and Molecular Biology Laboratory, Federal Institute of Education, Science, and Technology of Rio Grande do Sul, Sertão, Rio Grande do Sul, Brazil
| | - Carla Alves
- Biochemistry and Molecular Biology Laboratory, Federal Institute of Education, Science, and Technology of Rio Grande do Sul, Sertão, Rio Grande do Sul, Brazil.,Graduate Program in Bioexperimentation, Universidade de Passo Fundo, São José, Passo Fundo, Brazil
| | - Denise Bilibio
- Biochemistry and Molecular Biology Laboratory, Federal Institute of Education, Science, and Technology of Rio Grande do Sul, Sertão, Rio Grande do Sul, Brazil
| | - Nathália Tafarel Sutorillo
- Biochemistry and Molecular Biology Laboratory, Federal Institute of Education, Science, and Technology of Rio Grande do Sul, Sertão, Rio Grande do Sul, Brazil
| | - Daniele Coradini Zamberlan
- Biochemistry and Molecular Biology Department, Center of Natural and Exacts Science, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Rosilene Rodrigues Kaizer
- Biochemistry and Molecular Biology Laboratory, Federal Institute of Education, Science, and Technology of Rio Grande do Sul, Sertão, Rio Grande do Sul, Brazil.,Graduate Program in Environmental Science and Technology, Federal University of Fronteira Sul (UFFS) - Erechim Campus, Erechim, Rio Grande do Sul, Brazil
| | - Leonardo José Gil Barcellos
- Graduate Program in Bioexperimentation, Universidade de Passo Fundo, São José, Passo Fundo, Brazil.,Graduate Program in Pharmacology, Universidade Federal de Santa Maria, Cidade Universitária, Camobi, Santa Maria, Brazil
| |
Collapse
|
31
|
Mitkevich VA, Barykin EP, Eremina S, Pani B, Katkova-Zhukotskaya O, Polshakov VI, Adzhubei AA, Kozin SA, Mironov AS, Makarov AA, Nudler E. Zn-dependent β-amyloid Aggregation and its Reversal by the Tetrapeptide HAEE. Aging Dis 2022; 14:309-318. [PMID: 37008059 PMCID: PMC10017155 DOI: 10.14336/ad.2022.0827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/27/2022] [Indexed: 11/18/2022] Open
Abstract
The pathogenesis of Alzheimer's disease (AD) is associated with the formation of cerebral amyloid plaques, the main components of which are the modified Aβ molecules as well as the metal ions. Aβ isomerized at Asp7 residue (isoD7-Aβ) is the most abundant isoform in amyloid plaques. We hypothesized that the pathogenic effect of isoD7-Aβ is due to the formation of zinc-dependent oligomers, and that this interaction can be disrupted by the rationally designed tetrapeptide (HAEE). Here, we utilized surface plasmon resonance, nuclear magnetic resonance, and molecular dynamics simulation to demonstrate Zn2+-dependent oligomerization of isoD7-Aβ and the formation of a stable isoD7-Aβ:Zn2+:HAEE complex incapable of forming oligomers. To demonstrate the physiological importance of zinc-dependent isoD7-Aβ oligomerization and the ability of HAEE to interfere with this process at the organismal level, we employed transgenic nematodes overexpressing human Aβ. We show that the presence of isoD7-Aβ in the medium triggers extensive amyloidosis that occurs in a Zn2+-dependent manner, enhances paralysis, and shortens the animals' lifespan. Exogenous HAEE completely reverses these pathological effects of isoD7-Aβ. We conclude that the synergistic action of isoD7-Aβ and Zn2+ promotes Aβ aggregation and that the selected small molecules capable of interrupting this process, such as HAEE, can potentially serve as anti-amyloid therapeutics.
Collapse
Affiliation(s)
- Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Evgeny P Barykin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Svetlana Eremina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Bibhusita Pani
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, USA.
| | | | - Vladimir I Polshakov
- Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University, Moscow, Russia.
| | - Alexei A Adzhubei
- Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| | - Sergey A Kozin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Alexander S Mironov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, USA.
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, USA.
- Correspondence should be addressed to: Dr. Evgeny Nudler, Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA. .
| |
Collapse
|
32
|
Zhang Y, Zhao C, Zhang H, Lu Q, Zhou J, Liu R, Wang S, Pu Y, Yin L. Trans-generational effects of copper on nerve damage in Caenorhabditis elegans. CHEMOSPHERE 2021; 284:131324. [PMID: 34225113 DOI: 10.1016/j.chemosphere.2021.131324] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 05/15/2023]
Abstract
The potential toxicity of copper has received great attention for a long time, however, trans-generational effects of copper have not been extensively investigated. Caenorhabditis elegans (C. elegans) was used to evaluate the trans-generational toxicities of copper several physiological endpoints: growth, head thrashes and body bends and degree of neuronal damage. Copper significantly inhibited growth, body bends, head thrashes and caused degeneration of dopaminergic neurons in a concentration-dependent manner in parental worms. Further we found oxidative damage was to underlying the onset of neuron degeneration. In our study copper promoted ROS accumulation, and led to an increased expression of the oxidative stress response-related genes sod-3 and a decreased expression of metal detoxification genes mtl-1 and mtl-2. Moreover, copper increased the fluorescence intensity of the transgenic strain that encodes the antioxidant enzyme SOD-3. Gradually decline in copper-induced impairments were observed in the filial generations without exposure. No growth impairment was shown in F3, the trend of head thrashes recovery gradually appeared in F2 and no growth impairment was shown in F3, the body bends impairment caused by the parental copper exposure was recovery until F4 and no growth impairment was shown in F5. Besides, dopamine neurons revealed damage related to neurobehavioral endpoints, with hereditary effects in the progeny together. In addition, sequencing results suggested that copper exposure could cause epigenetic changes. QRT-PCR results showed that differentially expressed genes can also be passed on to offspring.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| | - Chao Zhao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| | - Hu Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| | - Qiang Lu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| | - Jingjing Zhou
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| | - Shizhi Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
33
|
Tamagno WA, Vanin AP, Sutorillo NT, Bilibio D, Dada RA, Colla LM, Zamberlan DC, Kaizer RR, Barcellos LJG. Fruit extract of red pitaya (Hylocereus undatus) prevents and reverses stress-induced impairments in the cholinergic and antioxidant systems of Caenorhabditis elegans. J Food Biochem 2021; 46:e13981. [PMID: 34698395 DOI: 10.1111/jfbc.13981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/27/2022]
Abstract
The addition of fruit to the diet is very important, and we can use nutraceutical and functional foods for this supplement. A little-known fruit is a red pitaya (Hylocereus undatus) that has been widely reported to have a high antioxidant potential. In this study, we analyzed the in vitro and in vivo antioxidant capacity of microencapsulated pitaya extract on the behavior, antioxidant, and nervous system of the nematode Caenorhabditis elegans. The worms were treated with fruit extract before and after juglone-induced stress, to determine the protective or curative effects of pitaya. We have been evaluated cholinergic, antioxidant, and behavioral biomarkers. We have evidenced that the pulp of pitaya contains antioxidant compounds and can serve as a potential nutraceutical product. In addition, the fruit extract was effective in preventing and/or reverse the stress-induced damages, even at high levels of chemical stress at all evaluated parameters. PRACTICAL APPLICATIONS: The potential applications and uses aimed by this research are related to the supplementation of foods given the antioxidant effect. Our data suggested that the effect of the pitaya fruit microencapsulated pulp extract was effective to prevent and repair the damage caused by oxidative stress. Besides the use of this microencapsulated extract can be an auxiliary in the treatment of diseases related to oxidative damage as well as promoting senescent aging. Another important use is the application of this extract as a dietary supplement to fortify the antioxidant system.
Collapse
Affiliation(s)
- Wagner Antonio Tamagno
- Biochemistry and Molecular Biology Laboratory of the Federal Institute of Education, Science and Technology of Rio Grande do Sul - Sertão Campus, City of Sertão, Brazil
| | - Ana Paula Vanin
- Biochemistry and Molecular Biology Laboratory of the Federal Institute of Education, Science and Technology of Rio Grande do Sul - Sertão Campus, City of Sertão, Brazil.,Graduate Program in Environmental Science and Technology, Federal University of Fronteira Sul (UFFS) - Erechim Campus, City of Erechim, Brazil
| | - Nathália Tafarel Sutorillo
- Biochemistry and Molecular Biology Laboratory of the Federal Institute of Education, Science and Technology of Rio Grande do Sul - Sertão Campus, City of Sertão, Brazil
| | - Denise Bilibio
- Biochemistry and Molecular Biology Laboratory of the Federal Institute of Education, Science and Technology of Rio Grande do Sul - Sertão Campus, City of Sertão, Brazil
| | - Renata Affeldt Dada
- Biochemistry and Molecular Biology Laboratory of the Federal Institute of Education, Science and Technology of Rio Grande do Sul - Sertão Campus, City of Sertão, Brazil
| | - Luciane Maria Colla
- Graduate Program in Food Science and Technology, Universidade de Passo Fundo, Passo Fundo, Brazil
| | - Daniele Coradini Zamberlan
- Biochemistry and Molecular Biology Department, Center of Natural and Exacts Science, Federal University of Santa Maria, Santa Maria, Brazil
| | - Rosilene Rodrigues Kaizer
- Biochemistry and Molecular Biology Laboratory of the Federal Institute of Education, Science and Technology of Rio Grande do Sul - Sertão Campus, City of Sertão, Brazil.,Graduate Program in Environmental Science and Technology, Federal University of Fronteira Sul (UFFS) - Erechim Campus, City of Erechim, Brazil
| | - Leonardo José Gil Barcellos
- Graduate Program in Bioexperimentation, University of Passo Fundo, Passo Fundo, Brazil.,Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| |
Collapse
|
34
|
Mitochondria-affecting small molecules ameliorate proteostasis defects associated with neurodegenerative diseases. Sci Rep 2021; 11:17733. [PMID: 34489512 PMCID: PMC8421394 DOI: 10.1038/s41598-021-97148-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023] Open
Abstract
Macroautophagic recycling of dysfunctional mitochondria, known as mitophagy, is essential for mitochondrial homeostasis and cell viability. Accumulation of defective mitochondria and impaired mitophagy have been widely implicated in many neurodegenerative diseases, and loss-of-function mutations of PINK1 and Parkin, two key regulators of mitophagy, are amongst the most common causes of heritable parkinsonism. This has led to the hypothesis that pharmacological stimulation of mitophagy may be a feasible approach to combat neurodegeneration. Toward this end, we screened ~ 45,000 small molecules using a high-throughput, whole-organism, phenotypic screen that monitored accumulation of PINK-1 protein, a key event in mitophagic activation, in a Caenorhabditis elegans strain carrying a Ppink-1::PINK-1::GFP reporter. We obtained eight hits that increased mitochondrial fragmentation and autophagosome formation. Several of the compounds also reduced ATP production, oxygen consumption, mitochondrial mass, and/or mitochondrial membrane potential. Importantly, we found that treatment with two compounds, which we named PS83 and PS106 (more commonly known as sertraline) reduced neurodegenerative disease phenotypes, including delaying paralysis in a C. elegans β-amyloid aggregation model in a PINK-1-dependent manner. This report presents a promising step toward the identification of compounds that will stimulate mitochondrial turnover.
Collapse
|
35
|
Carregosa D, Mota S, Ferreira S, Alves-Dias B, Loncarevic-Vasiljkovic N, Crespo CL, Menezes R, Teodoro R, dos Santos CN. Overview of Beneficial Effects of (Poly)phenol Metabolites in the Context of Neurodegenerative Diseases on Model Organisms. Nutrients 2021; 13:2940. [PMID: 34578818 PMCID: PMC8464690 DOI: 10.3390/nu13092940] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022] Open
Abstract
The rise of neurodegenerative diseases in an aging population is an increasing problem of health, social and economic consequences. Epidemiological and intervention studies have demonstrated that diets rich in (poly)phenols can have potent health benefits on cognitive decline and neurodegenerative diseases. Meanwhile, the role of gut microbiota is ever more evident in modulating the catabolism of (poly)phenols to dozens of low molecular weight (poly)phenol metabolites that have been identified in plasma and urine. These metabolites can reach circulation in higher concentrations than parent (poly)phenols and persist for longer periods of time. However, studies addressing their potential brain effects are still lacking. In this review, we will discuss different model organisms that have been used to study how low molecular weight (poly)phenol metabolites affect neuronal related mechanisms gathering critical insight on their potential to tackle the major hallmarks of neurodegeneration.
Collapse
Affiliation(s)
- Diogo Carregosa
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
| | - Sara Mota
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- iBET, Institute of Experimental and Technological Biology, Apartado 12, 2781-901 Oeiras, Portugal
| | - Sofia Ferreira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- CBIOS, University Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Beatriz Alves-Dias
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
| | - Natasa Loncarevic-Vasiljkovic
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- Department of Neurobiology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Carolina Lage Crespo
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
| | - Regina Menezes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- iBET, Institute of Experimental and Technological Biology, Apartado 12, 2781-901 Oeiras, Portugal
- CBIOS, University Lusófona’s Research Center for Biosciences & Health Technologies, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Rita Teodoro
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
| | - Cláudia Nunes dos Santos
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisboa, Portugal; (D.C.); (S.M.); (S.F.); (B.A.-D.); (N.L.-V.); (C.L.C.); (R.M.); (R.T.)
- iBET, Institute of Experimental and Technological Biology, Apartado 12, 2781-901 Oeiras, Portugal
| |
Collapse
|
36
|
Giong HK, Subramanian M, Yu K, Lee JS. Non-Rodent Genetic Animal Models for Studying Tauopathy: Review of Drosophila, Zebrafish, and C. elegans Models. Int J Mol Sci 2021; 22:8465. [PMID: 34445171 PMCID: PMC8395099 DOI: 10.3390/ijms22168465] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Tauopathy refers to a group of progressive neurodegenerative diseases, including frontotemporal lobar degeneration and Alzheimer's disease, which correlate with the malfunction of microtubule-associated protein Tau (MAPT) due to abnormal hyperphosphorylation, leading to the formation of intracellular aggregates in the brain. Despite extensive efforts to understand tauopathy and develop an efficient therapy, our knowledge is still far from complete. To find a solution for this group of devastating diseases, several animal models that mimic diverse disease phenotypes of tauopathy have been developed. Rodents are the dominating tauopathy models because of their similarity to humans and established disease lines, as well as experimental approaches. However, powerful genetic animal models using Drosophila, zebrafish, and C. elegans have also been developed for modeling tauopathy and have contributed to understanding the pathophysiology of tauopathy. The success of these models stems from the short lifespans, versatile genetic tools, real-time in-vivo imaging, low maintenance costs, and the capability for high-throughput screening. In this review, we summarize the main findings on mechanisms of tauopathy and discuss the current tauopathy models of these non-rodent genetic animals, highlighting their key advantages and limitations in tauopathy research.
Collapse
Affiliation(s)
- Hoi-Khoanh Giong
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Manivannan Subramanian
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Kweon Yu
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Jeong-Soo Lee
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| |
Collapse
|
37
|
Ijomone OM, Gubert P, Okoh COA, Varão AM, Amara LDO, Aluko OM, Aschner M. Application of Fluorescence Microscopy and Behavioral Assays to Demonstrating Neuronal Connectomes and Neurotransmitter Systems in C. elegans. NEUROMETHODS 2021; 172:399-426. [PMID: 34754139 PMCID: PMC8575032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The nematode Caenorhabditis elegans (C. elegans) is a prevailing model which is commonly utilized in a variety of biomedical research arenas, including neuroscience. Due to its transparency and simplicity, it is becoming a choice model organism for conducting imaging and behavioral assessment crucial to understanding the intricacies of the nervous system. Here, the methods required for neuronal characterization using fluorescent proteins and behavioral tasks are described. These are simplified protocols using fluorescent microscopy and behavioral assays to examine neuronal connections and associated neurotransmitter systems involved in normal physiology and aberrant pathology of the nervous system. Our aim is to make available to readers some streamlined and replicable procedures using C. elegans models as well as highlighting some of the limitations.
Collapse
Affiliation(s)
- Omamuyovwi M. Ijomone
- The Neuro- Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
- Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Priscila Gubert
- Department of Biochemistry, Laboratório de Imunopatologia Keizo Asami, LIKA, Federal University of Pernambuco, Recife, Brazil
- Postgraduate Program in Pure and Applied Chemistry, Federal University of Western of Bahia, Bahia, Brazil
| | - Comfort O. A. Okoh
- The Neuro- Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Alexandre M. Varão
- Postgraduate Program in Pure and Applied Chemistry, Federal University of Western of Bahia, Bahia, Brazil
| | - Leandro de O. Amara
- Postgraduate Program in Pure and Applied Chemistry, Federal University of Western of Bahia, Bahia, Brazil
| | - Oritoke M. Aluko
- The Neuro- Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
- Department of Physiology, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology and Neurosciences, Albert Einstein College of Medicine, NY, USA
| |
Collapse
|
38
|
Calahorro F, Holden-Dye L, O'Connor V. Impact of drug solvents on C. elegans pharyngeal pumping. Toxicol Rep 2021; 8:1240-1247. [PMID: 34195015 PMCID: PMC8233170 DOI: 10.1016/j.toxrep.2021.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 05/20/2021] [Accepted: 06/14/2021] [Indexed: 02/05/2023] Open
Abstract
Caenorhabditis elegans provides a multi-cellular model organism for toxicology and drug discovery. These studies usually require solvents such as dimethyl sulfoxide (DMSO), ethanol or acetone as a vehicle. This raises the need to carefully consider whether the chemical vehicles used in these screens are anodyne towards C. elegans. Here, we use pharyngeal pumping as a bioassay to assess this. Pharyngeal pumping is a visually scoreable behaviour that is controlled by environmental cues activating sensory and integrative neural signalling to coordinate pharyngeal activity. As such it serves as a rich bioassay to screen for chemical modulation. We found that while pumping was insensitive to high concentrations of the widely used drug solvents ethanol and acetone, it was perturbed by concentrations of DMSO above 0.5 % v/v encompassing concentrations used as drug vehicle. This was manifested as an inhibition of pharyngeal pump rate followed by a slow recovery in the continued presence of the solvent. The inhibition was not observed in a neuroligin mutant, nlg-1, consistent with DMSO acting at the level of sensory processing that modulates pumping. We found that bus-17 mutants, which have enhanced cuticle penetration to drugs are more sensitive to DMSO. The effect of DMSO is accompanied by a progressive morphological disruption in which internal membrane-like structures of varying size accumulate. These internal structures are seen in all three genotypes investigated in this study and likely arise independent of the effects on pharyngeal pumping. Overall, these results highlight sensory signalling and strain dependent vehicle sensitivity. Although we define concentrations at which this can be mitigated, it highlights the need to consider time-dependent vehicle effects when evaluating control responses in C. elegans chemical biology.
Collapse
|
39
|
Liu L, Guo P, Wang P, Zheng S, Qu Z, Liu N. The Review of Anti-aging Mechanism of Polyphenols on Caenorhabditis elegans. Front Bioeng Biotechnol 2021; 9:635768. [PMID: 34327192 PMCID: PMC8314386 DOI: 10.3389/fbioe.2021.635768] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
Micronutrients extracted from natural plants or made by biological synthesis are widely used in anti-aging research and applications. Among more than 30 effective anti-aging substances, employing polyphenol organic compounds for modification or delaying of the aging process attracts great interest because of their distinct contribution in the prevention of degenerative diseases, such as cardiovascular disease and cancer. There is a profound potential for polyphenol extracts in the research of aging and the related diseases of the elderly. Previous studies have mainly focused on the properties of polyphenols implicated in free radical scavenging; however, the anti-oxidant effect cannot fully elaborate its biological functions, such as neuroprotection, Aβ protein production, ion channel coupling, and signal transduction pathways. Caenorhabditis elegans (C. elegans) has been considered as an ideal model organism for exploring the mechanism of anti-aging research and is broadly utilized in screening for natural bioactive substances. In this review, we have described the molecular mechanisms and pathways responsible for the slowdown of aging processes exerted by polyphenols. We also have discussed the possible mechanisms for their anti-oxidant and anti-aging properties in C. elegans from the perspective of different classifications of the specific polyphenols, such as flavonols, anthocyanins, flavan-3-ols, hydroxybenzoic acid, hydroxycinnamic acid, and stilbenes.
Collapse
Affiliation(s)
- Limin Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China.,Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China
| | - Peisen Guo
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Peixi Wang
- Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China
| | - Shanqing Zheng
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Zhi Qu
- Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China
| | - Nan Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China.,Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China.,Institute of Environment and Health, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
40
|
Giunti S, Andersen N, Rayes D, De Rosa MJ. Drug discovery: Insights from the invertebrate Caenorhabditis elegans. Pharmacol Res Perspect 2021; 9:e00721. [PMID: 33641258 PMCID: PMC7916527 DOI: 10.1002/prp2.721] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
Therapeutic drug development is a long, expensive, and complex process that usually takes 12-15 years. In the early phases of drug discovery, in particular, there is a growing need for animal models that ensure the reduction in both cost and time. Caenorhabditis elegans has been traditionally used to address fundamental aspects of key biological processes, such as apoptosis, aging, and gene expression regulation. During the last decade, with the advent of large-scale platforms for screenings, this invertebrate has also emerged as an essential tool in the pharmaceutical research industry to identify novel drugs and drug targets. In this review, we discuss the reasons why C. elegans has been positioned as an outstanding cost-effective option for drug discovery, highlighting both the advantages and drawbacks of this model. Particular attention is paid to the suitability of this nematode in large-scale genetic and pharmacological screenings. High-throughput screenings in C. elegans have indeed contributed to the breakthrough of a wide variety of candidate compounds involved in extensive fields including neurodegeneration, pathogen infections and metabolic disorders. The versatility of this nematode, which enables its instrumentation as a model of human diseases, is another attribute also herein underscored. As illustrative examples, we discuss the utility of C. elegans models of both human neurodegenerative diseases and parasitic nematodes in the drug discovery industry. Summing up, this review aims to demonstrate the impact of C. elegans models on the drug discovery pipeline.
Collapse
Affiliation(s)
- Sebastián Giunti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - Natalia Andersen
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - Diego Rayes
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - María José De Rosa
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| |
Collapse
|
41
|
Zbinden A, Pérez-Berlanga M, De Rossi P, Polymenidou M. Phase Separation and Neurodegenerative Diseases: A Disturbance in the Force. Dev Cell 2021; 55:45-68. [PMID: 33049211 DOI: 10.1016/j.devcel.2020.09.014] [Citation(s) in RCA: 280] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/09/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022]
Abstract
Protein aggregation is the main hallmark of neurodegenerative diseases. Many proteins found in pathological inclusions are known to undergo liquid-liquid phase separation, a reversible process of molecular self-assembly. Emerging evidence supports the hypothesis that aberrant phase separation behavior may serve as a trigger of protein aggregation in neurodegeneration, and efforts to understand and control the underlying mechanisms are underway. Here, we review similarities and differences among four main proteins, α-synuclein, FUS, tau, and TDP-43, which are found aggregated in different diseases and were independently shown to phase separate. We discuss future directions in the field that will help shed light on the molecular mechanisms of aggregation and neurodegeneration.
Collapse
Affiliation(s)
- Aurélie Zbinden
- Department of Quantitative Biomedicine, University of Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Manuela Pérez-Berlanga
- Department of Quantitative Biomedicine, University of Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Pierre De Rossi
- Department of Quantitative Biomedicine, University of Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Magdalini Polymenidou
- Department of Quantitative Biomedicine, University of Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
42
|
Youssef K, Archonta D, Kubiseski TJ, Tandon A, Rezai P. Electric egg-laying: a new approach for regulating C. elegans egg-laying behaviour in a microchannel using electric field. LAB ON A CHIP 2021; 21:821-834. [PMID: 33527103 DOI: 10.1039/d0lc00964d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In this paper, the novel effect of electric field (EF) on adult C. elegans egg-laying in a microchannel is discovered and correlated with neural and muscular activities. The quantitative effects of worm aging and EF strength, direction, and exposure duration on egg-laying are studied phenotypically using egg-count, body length, head movement, and transient neuronal activity readouts. Electric egg-laying rate increases significantly when worms face the anode and the response is EF-dependent, i.e. stronger (6 V cm-1) and longer EF (40 s) exposure result in a shorter egg laying response duration. Worm aging significantly deteriorates the electric egg-laying behaviour with an 88% decrease in the egg-count from day-1 to day-4 post young-adult stage. Fluorescent imaging of intracellular calcium dynamics in the main parts of the egg-laying neural circuit demonstrates the involvement and sensitivity of the serotonergic hermaphrodite specific neurons (HSNs), vulva muscles, and ventral cord neurons to the EF. HSN mutation also results in a reduced rate of electric egg-laying allowing the use of this technique for cellular screening and mapping of the neural basis of electrosensation in C. elegans. This novel assay can be parallelized and performed in a high-throughput manner for drug and gene screening applications.
Collapse
Affiliation(s)
- Khaled Youssef
- Department of Mechanical Engineering, York University, Toronto, ON, Canada.
| | - Daphne Archonta
- Department of Mechanical Engineering, York University, Toronto, ON, Canada.
| | | | - Anurag Tandon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, Ontario, Canada and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Pouya Rezai
- Department of Mechanical Engineering, York University, Toronto, ON, Canada.
| |
Collapse
|
43
|
Health and longevity studies in C. elegans: the "healthy worm database" reveals strengths, weaknesses and gaps of test compound-based studies. Biogerontology 2021; 22:215-236. [PMID: 33683565 PMCID: PMC7973913 DOI: 10.1007/s10522-021-09913-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/20/2021] [Indexed: 12/11/2022]
Abstract
Several biogerontology databases exist that focus on genetic or gene expression data linked to health as well as survival, subsequent to compound treatments or genetic manipulations in animal models. However, none of these has yet collected experimental results of compound-related health changes. Since quality of life is often regarded as more valuable than length of life, we aim to fill this gap with the “Healthy Worm Database” (http://healthy-worm-database.eu). Literature describing health-related compound studies in the aging model Caenorhabditis elegans was screened, and data for 440 compounds collected. The database considers 189 publications describing 89 different phenotypes measured in 2995 different conditions. Besides enabling a targeted search for promising compounds for further investigations, this database also offers insights into the research field of studies on healthy aging based on a frequently used model organism. Some weaknesses of C. elegans-based aging studies, like underrepresented phenotypes, especially concerning cognitive functions, as well as the convenience-based use of young worms as the starting point for compound treatment or phenotype measurement are discussed. In conclusion, the database provides an anchor for the search for compounds affecting health, with a link to public databases, and it further highlights some potential shortcomings in current aging research.
Collapse
|
44
|
Gil-Hernández A, Silva-Palacios A. Relevance of endoplasmic reticulum and mitochondria interactions in age-associated diseases. Ageing Res Rev 2020; 64:101193. [PMID: 33069818 DOI: 10.1016/j.arr.2020.101193] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/29/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Although the elixir of youth remains in the darkness, medical and scientific advances have succeeded in increasing human longevity; however, the predisposition to disease and its high economic cost are raising. Different strategies (e.g., antioxidants) and signaling pathways (e.g., Nrf2) have been identified to help regulate disease progression, nevertheless, there are still missing links that we need to understand. Contact sites called mitochondria-associated membranes (MAM) allow bi-directional communication between organelles as part of the essential functions in the cell to maintain its homeostasis. Different groups have deeply studied the role of MAM in aging; however, it's necessary to analyze their involvement in the progression of age-related diseases. In this review, we highlight the role of contact sites in these conditions, as well as the morphological and functional changes of mitochondria and ER in aging. We emphasize the intimate relationship between both organelles as a reflection of the biological processes that take place in the cell to try to regulate the deterioration characteristic of the aging process; proposing MAM as a potential target to help limit the disease progression with age.
Collapse
|
45
|
Parallel-Channel Electrotaxis and Neuron Screening of Caenorhabditis elegans. MICROMACHINES 2020; 11:mi11080756. [PMID: 32759767 PMCID: PMC7465510 DOI: 10.3390/mi11080756] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022]
Abstract
In this paper, we report a novel microfluidic method to conduct a Caenorhabditis elegans electrotaxis movement assay and neuronal imaging on up to 16 worms in parallel. C. elegans is a model organism for neurodegenerative disease and movement disorders such as Parkinson’s disease (PD), and for screening chemicals that alleviate protein aggregation, neuronal death, and movement impairment in PD. Electrotaxis of C. elegans in microfluidic channels has led to the development of neurobehavioral screening platforms, but enhancing the throughput of the electrotactic behavioral assay has remained a challenge. Our device consisted of a hierarchy of tree-like channels for worm loading into 16 parallel electrotaxis screening channels with equivalent electric fields. Tapered channels at the ends of electrotaxis channels were used for worm immobilization and fluorescent imaging of neurons. Parallel electrotaxis of worms was first validated against established single-worm electrotaxis phenotypes. Then, mutant screening was demonstrated using the NL5901 strain, carrying human α-synuclein in the muscle cells, by showing the associated electrotaxis defects in the average speed, body bend frequency (BBF), and electrotaxis time index (ETI). Moreover, chemical screening of a PD worm model was shown by exposing the BZ555 strain, expressing green fluorescence protein (GFP) in the dopaminergic neurons (DNs), to 6-hydroxydopamine neurotoxin. The neurotoxin-treated worms exhibited a reduction in electrotaxis swimming speed, BBF, ETI, and DNs fluorescence intensity. We envision our technique to be used widely in C. elegans-based movement disorder assays to accelerate behavioral and cellular phenotypic investigations.
Collapse
|
46
|
A Dihydroflavonoid Naringin Extends the Lifespan of C. elegans and Delays the Progression of Aging-Related Diseases in PD/AD Models via DAF-16. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6069354. [PMID: 32832002 PMCID: PMC7422489 DOI: 10.1155/2020/6069354] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/20/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Naringin is a dihydroflavonoid, which is rich in several plant species used for herbal medicine. It has a wide range of biological activities, including antineoplastic, anti-inflammatory, antiphotoaging, and antioxidative activities. So it would be interesting to know if naringin has an effect on aging and aging-related diseases. We examined the effect of naringin on the aging of Caenorhabditis elegans (C. elegans). Our results showed that naringin could extend the lifespan of C. elegans. Moreover, naringin could also increase the thermal and oxidative stress tolerance, reduce the accumulation of lipofuscin, and delay the progress of aging-related diseases in C. elegans models of AD and PD. Naringin could not significantly extend the lifespan of long-lived mutants from genes in insulin/IGF-1 signaling (IIS) and nutrient-sensing pathways, such as daf-2, akt-2, akt-1, eat-2, sir-2.1, and rsks-1. Naringin treatment prolonged the lifespan of long-lived glp-1 mutants, which have decreased reproductive stem cells. Naringin could not extend the lifespan of a null mutant of the fox-head transcription factor DAF-16. Moreover, naringin could increase the mRNA expression of genes regulated by daf-16 and itself. In conclusion, we show that a natural product naringin could extend the lifespan of C. elegans and delay the progression of aging-related diseases in C. elegans models via DAF-16.
Collapse
|
47
|
Pretsch D, Rollinger JM, Schmid A, Genov M, Wöhrer T, Krenn L, Moloney M, Kasture A, Hummel T, Pretsch A. Prolongation of metallothionein induction combats Aß and α-synuclein toxicity in aged transgenic Caenorhabditis elegans. Sci Rep 2020; 10:11707. [PMID: 32678125 PMCID: PMC7366685 DOI: 10.1038/s41598-020-68561-7] [Citation(s) in RCA: 15] [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: 03/11/2019] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative disorders (ND) like Alzheimer's (AD), Parkinson's (PD), Huntington's or Prion diseases share similar pathological features. They are all age dependent and are often associated with disruptions in analogous metabolic processes such as protein aggregation and oxidative stress, both of which involve metal ions like copper, manganese and iron. Bush and Tanzi proposed 2008 in the 'metal hypothesis of Alzheimer's disease' that a breakdown in metal homeostasis is the main cause of NDs, and drugs restoring metal homeostasis are promising novel therapeutic strategies. We report here that metallothionein (MT), an endogenous metal detoxifying protein, is increased in young amyloid ß (Aß) expressing Caenorhabditis elegans, whereas it is not in wild type strains. Further MT induction collapsed in 8 days old transgenic worms, indicating the age dependency of disease outbreak, and sharing intriguing parallels to diminished MT levels in human brains of AD. A medium throughput screening assay method was established to search for compounds increasing the MT level. Compounds known to induce MT release like progesterone, ZnSO4, quercetin, dexamethasone and apomorphine were active in models of AD and PD. Thioflavin T, clioquinol and emodin are promising leads in AD and PD research, whose mode of action has not been fully established yet. In this study, we could show that the reduction of Aß and α-synuclein toxicity in transgenic C. elegans models correlated with the prolongation of MT induction time and that knockdown of MT with RNA interference resulted in a loss of bioactivity.
Collapse
Affiliation(s)
- Dagmar Pretsch
- Oxford Antibiotic Group GmbH, Konrad-Lorenz-Straße 24, 3430, Tulln, Austria.
| | - Judith Maria Rollinger
- Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Axel Schmid
- Department of Neuroscience and Developmental Biology, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Miroslav Genov
- Oxford Antibiotic Group GmbH, Konrad-Lorenz-Straße 24, 3430, Tulln, Austria
| | - Teresa Wöhrer
- Oxford Antibiotic Group GmbH, Konrad-Lorenz-Straße 24, 3430, Tulln, Austria
| | - Liselotte Krenn
- Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Mark Moloney
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK
| | - Ameya Kasture
- Department of Neuroscience and Developmental Biology, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Thomas Hummel
- Department of Neuroscience and Developmental Biology, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Alexander Pretsch
- Oxford Antibiotic Group GmbH, Konrad-Lorenz-Straße 24, 3430, Tulln, Austria
| |
Collapse
|
48
|
Healthspan pathway maps in C. elegans and humans highlight transcription, proliferation/biosynthesis and lipids. Aging (Albany NY) 2020; 12:12534-12581. [PMID: 32634117 PMCID: PMC7377848 DOI: 10.18632/aging.103514] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/04/2020] [Indexed: 12/17/2022]
Abstract
The molecular basis of aging and of aging-associated diseases is being unraveled at an increasing pace. An extended healthspan, and not merely an extension of lifespan, has become the aim of medical practice. Here, we define health based on the absence of diseases and dysfunctions. Based on an extensive review of the literature, in particular for humans and C. elegans, we compile a list of features of health and of the genes associated with them. These genes may or may not be associated with survival/lifespan. In turn, survival/lifespan genes that are not known to be directly associated with health are not considered. Clusters of these genes based on molecular interaction data give rise to maps of healthspan pathways for humans and for C. elegans. Overlaying healthspan-related gene expression data onto the healthspan pathway maps, we observe the downregulation of (pro-inflammatory) Notch signaling in humans and of proliferation in C. elegans. We identify transcription, proliferation/biosynthesis and lipids as a common theme on the annotation level, and proliferation-related kinases on the gene/protein level. Our literature-based data corpus, including visualization, should be seen as a pilot investigation of the molecular underpinnings of health in two different species. Web address: http://pathways.h2020awe.eu.
Collapse
|
49
|
Lu M, Li H, Li Y, Lu Y, Wang H, Wang X. Exploring the Toxicology of Depleted Uranium with Caenorhabditis elegans. ACS OMEGA 2020; 5:12119-12125. [PMID: 32548391 PMCID: PMC7271045 DOI: 10.1021/acsomega.0c00380] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/11/2020] [Indexed: 06/11/2023]
Abstract
Depleted uranium (DU) is an emerging heavy metal pollutant with considerable environmental and occupational concerns. Its radiotoxicity is known to be low. However, its chemical toxicity should not be ignored. In order to explore the chemical toxicity of DU, the effects of uranyl nitrate, prepared from DU, on the model organism Caenorhabditis elegans were investigated. Chronic exposure to DU did not affect the lifespan or reproduction of the worm. DU had little effect on the physiological processes of C. elegans. Additionally, DU treatment did not make C. elegans more susceptible to UV, heat, or oxidative stress. Interestingly, chronic exposure of DU decreased the in vivo reactive oxygen species-scavenging ability through inhibiting the expression of antioxidant genes ctl-1, ctl-2, ctl-3, gst-7, and gst-10. Chronic but not acute exposure of DU induced a statistically significant degeneration of the dopaminergic (DAergic) neurons of treated worms and promoted the increase of α-synuclein aggregation and DAergic neurotoxicity. These findings may raise the public concerns regarding DU as an etiologic agent of Parkinson's disease and underline its potential neurotoxicity.
Collapse
Affiliation(s)
- Meiling Lu
- State Key Laboratory
for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences of
Guangxi Normal University, Guilin 541004, China
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of
Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yunfei Li
- Department
of Pharmaceutical Engineering, College of Humanities & Information, Changchun University of Technology, Changchun 130122, China
| | - Yuyuan Lu
- State Key Laboratory of Polymer Physics
and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Hengshan Wang
- State Key Laboratory
for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences of
Guangxi Normal University, Guilin 541004, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of
Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
50
|
Di Rosa G, Brunetti G, Scuto M, Trovato Salinaro A, Calabrese EJ, Crea R, Schmitz-Linneweber C, Calabrese V, Saul N. Healthspan Enhancement by Olive Polyphenols in C. elegans Wild Type and Parkinson's Models. Int J Mol Sci 2020; 21:E3893. [PMID: 32486023 PMCID: PMC7312680 DOI: 10.3390/ijms21113893] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the second most prevalent late-age onset neurodegenerative disorder, affecting 1% of the population after the age of about 60 years old and 4% of those over 80 years old, causing motor impairments and cognitive dysfunction. Increasing evidence indicates that Mediterranean diet (MD) exerts beneficial effects in maintaining health, especially during ageing and by the prevention of neurodegenerative disorders. In this regard, olive oil and its biophenolic constituents like hydroxytyrosol (HT) have received growing attention in the past years. Thus, in the current study we test the health-promoting effects of two hydroxytyrosol preparations, pure HT and Hidrox® (HD), which is hydroxytyrosol in its "natural" environment, in the established invertebrate model organism Caenorhabditis elegans. HD exposure led to much stronger beneficial locomotion effects in wild type worms compared to HT in the same concentration. Consistent to this finding, in OW13 worms, a PD-model characterized by α-synuclein expression in muscles, HD exhibited a significant higher effect on α-synuclein accumulation and swim performance than HT, an effect partly confirmed also in swim assays with the UA44 strain, which features α-synuclein expression in DA-neurons. Interestingly, beneficial effects of HD and HT treatment with similar strength were detected in the lifespan and autofluorescence of wild-type nematodes, in the neuronal health of UA44 worms as well as in the locomotion of rotenone-induced PD-model. Thus, the hypothesis that HD features higher healthspan-promoting abilities than HT was at least partly confirmed. Our study demonstrates that HD polyphenolic extract treatment has the potential to partly prevent or even treat ageing-related neurodegenerative diseases and ageing itself. Future investigations including mammalian models and human clinical trials are needed to uncover the full potential of these olive compounds.
Collapse
Affiliation(s)
- Gabriele Di Rosa
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.D.R.); (G.B.); (M.S.); (A.T.S.)
| | - Giovanni Brunetti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.D.R.); (G.B.); (M.S.); (A.T.S.)
| | - Maria Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.D.R.); (G.B.); (M.S.); (A.T.S.)
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.D.R.); (G.B.); (M.S.); (A.T.S.)
| | - Edward J. Calabrese
- Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA 01003, USA;
| | - Roberto Crea
- Oliphenol LLC., 26225 Eden Landing Road, Unit C, Hayward, CA 94545, USA;
| | - Christian Schmitz-Linneweber
- Faculty of Life Sciences, Institute of Biology, Molecular Genetics Group, Humboldt University of Berlin, Philippstr. 13, House 22, 10115 Berlin, Germany; (C.S.-L.); (N.S.)
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy; (G.D.R.); (G.B.); (M.S.); (A.T.S.)
| | - Nadine Saul
- Faculty of Life Sciences, Institute of Biology, Molecular Genetics Group, Humboldt University of Berlin, Philippstr. 13, House 22, 10115 Berlin, Germany; (C.S.-L.); (N.S.)
| |
Collapse
|