1
|
Seira Curto J, Dominguez Martinez A, Perez Collell G, Barniol Simon E, Romero Ruiz M, Franco Bordés B, Sotillo Sotillo P, Villegas Hernandez S, Fernandez MR, Sanchez de Groot N. Exogenous prion-like proteins and their potential to trigger cognitive dysfunction. Mol Syst Biol 2025:10.1038/s44320-025-00114-4. [PMID: 40425815 DOI: 10.1038/s44320-025-00114-4] [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: 05/03/2024] [Revised: 04/16/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
The gut is exposed to a wide range of proteins, including ingested proteins and those produced by the resident microbiota. While ingested prion-like proteins can propagate across species, their implications for disease development remain largely unknown. Here, we apply a multidisciplinary approach to examine the relationship between the biophysical properties of exogenous prion-like proteins and the phenotypic consequences of ingesting them. Through computational analysis of gut bacterial proteins, we identified an enrichment of prion-like sequences in Helicobacter pylori. Based on these findings, we rationally designed a set of synthetic prion-like sequences that form amyloid fibrils, interfere with amyloid-beta-peptide aggregation, and trigger prion propagation when introduced in the yeast Sup35 model. When C. elegans were fed bacteria expressing these prion-like proteins, they lost associative memory and exhibited increased lipid oxidation. These data suggest a link between memory impairment, the conformational state of aggregates, and oxidative stress. Overall, this work supports gut microbiota as a reservoir of exogenous prion-like sequences, especially H. pylori, and the gut as an entry point for molecules capable of triggering cognitive dysfunction.
Collapse
Affiliation(s)
- Jofre Seira Curto
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Adan Dominguez Martinez
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
| | - Genis Perez Collell
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Estrella Barniol Simon
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marina Romero Ruiz
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Berta Franco Bordés
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Paula Sotillo Sotillo
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sandra Villegas Hernandez
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Rosario Fernandez
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Natalia Sanchez de Groot
- Unitat de Bioquímica, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
2
|
Lee M, Lee J, Kim D, Min H, Shim YH. Caffeine-Induced Upregulation of pas-1 and pas-3 Enhances Intestinal Integrity by Reducing Vitellogenin in Aged Caenorhabditis elegans Model. Nutrients 2024; 16:4298. [PMID: 39770921 PMCID: PMC11677849 DOI: 10.3390/nu16244298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Intestinal aging is characterized by declining protein homeostasis via reduced proteasome activity, which are hallmarks of age-related diseases. Our previous study showed that caffeine intake improved intestinal integrity with age by reducing vitellogenin (VIT, yolk protein) in C. elegans. In this study, we investigated the regulatory mechanisms by which caffeine intake improves intestinal integrity and reduces vitellogenin (VIT) production in aged Caenorhabditis elegans. Methods: We performed RNA-seq analysis, and qRT-PCR to validate and confirm the RNA-seq results. Transgenic worms with VIT-2::GFP and VIT-6::GFP were used for measuring VIT production. dsRNAi was conducted to elucidate the roles of pas-1 and pas-3 genes. Results:pas-1 and pas-3, a C. elegans ortholog of human PASM4, was upregulated by caffeine intake. They reduced VIT production by repressing unc-62, a transcriptional activator of vit expression. Interestingly, vit-2 was required for pas-1 and pas-3 expression, and RNAi of pas-1 and pas-3 promoted intestinal atrophy and colonization, suggesting a balancing mechanism for VIT levels in intestinal health. Additionally, lifespan was extended by caffeine intake (2 ± 0.05 days), however, this effect was not observed by pas-1 but not pas-3 RNAi, suggesting that the mode of action for an anti-aging effect of caffeine through pas-1 and pas-3 is distinctive. The lifespan extended by pas-1 was mediated by SKN-1 activation. Conclusions: Caffeine intake enhances intestinal health through proteasome activity and extends lifespan in aged C. elegans by upregulating pas-1 and pas-3. These findings suggest that caffeine consumption mitigates age-related proteasome impairment and maintains intestinal integrity during aging.
Collapse
Affiliation(s)
- Mijin Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (M.L.); (H.M.)
| | - Jea Lee
- Department of Systems Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (J.L.); (D.K.)
| | - Dongyeon Kim
- Department of Systems Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (J.L.); (D.K.)
| | - Hyemin Min
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (M.L.); (H.M.)
| | - Yhong-Hee Shim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (M.L.); (H.M.)
- Department of Systems Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (J.L.); (D.K.)
| |
Collapse
|
3
|
Kalganova AI, Eliseev IE, Smirnov IV, Terekhov SS. Platforms for the Search for New Antimicrobial Agents Using In Vivo C. elegans Models. Acta Naturae 2024; 16:15-26. [PMID: 39877009 PMCID: PMC11771841 DOI: 10.32607/actanaturae.27348] [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: 12/11/2023] [Accepted: 11/19/2024] [Indexed: 01/31/2025] Open
Abstract
Despite the achievements brought about by high-throughput screening technologies, there is still a lack of effective platforms to be used to search for new antimicrobial drugs. The antimicrobial activity of compounds continues, for the most part, to be assessed mainly using in vitro pathogen cultures, a situation which does not make easy a detailed investigation of the molecular mechanisms underlying host-pathogen interactions. In vivo testing of promising compounds using chordate models is labor-intensive and expensive and, therefore, is used in preclinical studies of selected drug candidates but not in primary screening. This approach does not facilitate the selection of compounds with low organ toxicity and is not suitable for the identification of therapeutic compounds that affect virulence factors. The use of microscopic nematode C. elegans to model human infections is a promising approach that enables one to investigate the host-pathogen interaction and identify anti-infective compounds with new mechanisms of action.
Collapse
Affiliation(s)
- A. I. Kalganova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| | - I. E. Eliseev
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| | - I. V. Smirnov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Endocrinology Research Center, Moscow, 117292 Russian Federation
| | - S. S. Terekhov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| |
Collapse
|
4
|
Walker AC, Bhargava R, Bucher MJ, Argote YM, Brust AS, Czyż DM. Identification of proteotoxic and proteoprotective bacteria that non-specifically affect proteins associated with neurodegenerative diseases. iScience 2024; 27:110828. [PMID: 39310761 PMCID: PMC11414702 DOI: 10.1016/j.isci.2024.110828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/05/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
There are no cures for neurodegenerative protein conformational diseases (PCDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Emerging evidence suggests the gut microbiota plays a role in their pathogenesis, though the influences of specific bacteria on disease-associated proteins remain elusive. Here, we reveal the effects of 229 human bacterial isolates on the aggregation and toxicity of Aβ1-42, α-synuclein, and polyglutamine tracts in Caenorhabditis elegans expressing these culprit proteins. Our findings demonstrate that bacterial effects on host protein aggregation are consistent across different culprit proteins, suggesting that microbes affect protein stability by modulating host proteostasis rather than selectively targeting disease-associated proteins. Furthermore, we found that feeding C. elegans proteoprotective Prevotella corporis activates the heat shock response, revealing an unexpected discovery of a microbial influence on host proteostasis. Insight into how individual bacteria affect PCD proteins could open new strategies for prevention and treatment by altering the abundance of microbes.
Collapse
Affiliation(s)
- Alyssa C. Walker
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Rohan Bhargava
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Michael J. Bucher
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Yoan M. Argote
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Amanda S. Brust
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Daniel M. Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
5
|
Cordeiro LM, Soares FAA, Arantes LP. Evaluating polyglutamine protein aggregation and toxicity in transgenic Caenorhabditis elegans models of Huntington's disease. Methods Cell Biol 2024; 192:115-130. [PMID: 39863386 DOI: 10.1016/bs.mcb.2024.06.002] [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: 01/27/2025]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by a repeat of the cytosine-adenine-guanine trinucleotide (CAG) in the huntingtin gene (HTT). This results in the translation of a mutant huntingtin (mHTT) protein with an abnormally long polyglutamine (polyQ) repeat. The pathology of HD leads to neuronal cell loss, motor abnormalities, and dementia. Currently, the pathogenesis of HD remains incompletely understood, and available treatments only address symptoms. Caenorhabditis elegans has been used as a model for neurodegenerative diseases, enabling the exploration of the molecular, cellular, and physiological mechanisms underlying HD pathogenesis. It also facilitates the investigation of potential therapeutic targets and interventions. Here, we describe common experiments employed to assess polyQ aggregation and toxicity in transgenic C. elegans models of HD, utilizing fluorescent markers to detect protein aggregation and neuron degeneration, in addition to specific behavioral assays (thrash frequency, nose touch response, and octanol response).
Collapse
Affiliation(s)
- Larissa Marafiga Cordeiro
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Santa Maria, RS, Brazil
| | - Félix Alexandre Antunes Soares
- Federal University of Santa Maria, Center for Natural and Exact Sciences, Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Camobi, Santa Maria, RS, Brazil
| | | |
Collapse
|
6
|
Wu CY, Davis S, Saudagar N, Shah S, Zhao W, Stern A, Martel J, Ojcius D, Yang HC. Caenorhabditis elegans as a Convenient Animal Model for Microbiome Studies. Int J Mol Sci 2024; 25:6670. [PMID: 38928375 PMCID: PMC11203780 DOI: 10.3390/ijms25126670] [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: 04/18/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Microbes constitute the most prevalent life form on Earth, yet their remarkable diversity remains mostly unrecognized. Microbial diversity in vertebrate models presents a significant challenge for investigating host-microbiome interactions. The model organism Caenorhabditis elegans has many advantages for delineating the effects of host genetics on microbial composition. In the wild, the C. elegans gut contains various microbial species, while in the laboratory it is usually a host for a single bacterial species. There is a potential host-microbe interaction between microbial metabolites, drugs, and C. elegans phenotypes. This mini-review aims to summarize the current understanding regarding the microbiome in C. elegans. Examples using C. elegans to study host-microbe-metabolite interactions are discussed.
Collapse
Affiliation(s)
- Cheng-Yeu Wu
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan 33302, Taiwan; (C.-Y.W.); (J.M.)
| | - Scott Davis
- Department of Endodontics, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103, USA;
| | - Neekita Saudagar
- Doctor of Dental Surgery Program, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103, USA; (N.S.); (S.S.); (W.Z.)
| | - Shrey Shah
- Doctor of Dental Surgery Program, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103, USA; (N.S.); (S.S.); (W.Z.)
| | - William Zhao
- Doctor of Dental Surgery Program, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103, USA; (N.S.); (S.S.); (W.Z.)
| | - Arnold Stern
- Grossman School of Medicine, New York University, New York, NY 10016, USA;
| | - Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan 33302, Taiwan; (C.-Y.W.); (J.M.)
| | - David Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan 33302, Taiwan; (C.-Y.W.); (J.M.)
- Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103, USA
| | - Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu 30041, Taiwan
| |
Collapse
|
7
|
Pu X, Qi B. Lysosomal dysfunction by inactivation of V-ATPase drives innate immune response in C. elegans. Cell Rep 2024; 43:114138. [PMID: 38678555 DOI: 10.1016/j.celrep.2024.114138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/10/2024] [Accepted: 04/08/2024] [Indexed: 05/01/2024] Open
Abstract
Pathogens target vacuolar ATPase (V-ATPase) to inhibit lysosomal acidification or lysosomal fusion, causing lysosomal dysfunction. However, it remains unknown whether cells can detect dysfunctional lysosomes and initiate an immune response. In this study, we discover that dysfunction of lysosomes caused by inactivation of V-ATPase enhances innate immunity against bacterial infections. We find that lysosomal V-ATPase interacts with DVE-1, whose nuclear localization serves as a proxy for the induction of mitochondrial unfolded protein response (UPRmt). The inactivation of V-ATPase promotes the nuclear localization of DVE-1, activating UPRmt and inducing downstream immune response genes. Furthermore, pathogen resistance conferred by inactivation of V-ATPase requires dve-1 and its downstream immune effectors. Interestingly, animals grow slower after vha RNAi, suggesting that the vha-RNAi-induced immune response costs the most energy through activation of DVE-1, which trades off with growth. This study reveals how dysfunctional lysosomes can trigger an immune response, emphasizing the importance of conserving energy during immune defense.
Collapse
Affiliation(s)
- Xuepiao Pu
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Bin Qi
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China.
| |
Collapse
|
8
|
Zimmermann J, Piecyk A, Sieber M, Petersen C, Johnke J, Moitinho-Silva L, Künzel S, Bluhm L, Traulsen A, Kaleta C, Schulenburg H. Gut-associated functions are favored during microbiome assembly across a major part of C. elegans life. mBio 2024; 15:e0001224. [PMID: 38634692 PMCID: PMC11077962 DOI: 10.1128/mbio.00012-24] [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: 01/02/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
The microbiome expresses a variety of functions that influence host biology. The range of functions depends on the microbiome's composition, which can change during the host's lifetime due to neutral assembly processes, host-mediated selection, and environmental conditions. To date, the exact dynamics of microbiome assembly, the underlying determinants, and the effects on host-associated functions remain poorly understood. Here, we used the nematode Caenorhabditis elegans and a defined community of fully sequenced, naturally associated bacteria to study microbiome dynamics and functions across a major part of the worm's lifetime of hosts under controlled experimental conditions. Bacterial community composition initially shows strongly declining levels of stochasticity, which increases during later time points, suggesting selective effects in younger animals as opposed to more random processes in older animals. The adult microbiome is enriched in genera Ochrobactrum and Enterobacter compared to the direct substrate and a host-free control environment. Using pathway analysis, metabolic, and ecological modeling, we further find that the lifetime assembly dynamics increase competitive strategies and gut-associated functions in the host-associated microbiome, indicating that the colonizing bacteria benefit the worm. Overall, our study introduces a framework for studying microbiome assembly dynamics based on stochastic, ecological, and metabolic models, yielding new insights into the processes that determine host-associated microbiome composition and function. IMPORTANCE The microbiome plays a crucial role in host biology. Its functions depend on the microbiome composition that can change during a host's lifetime. To date, the dynamics of microbiome assembly and the resulting functions still need to be better understood. This study introduces a new approach to characterize the functional consequences of microbiome assembly by modeling both the relevance of stochastic processes and metabolic characteristics of microbial community changes. The approach was applied to experimental time-series data obtained for the microbiome of the nematode Caenorhabditis elegans across the major part of its lifetime. Stochastic processes played a minor role, whereas beneficial bacteria as well as gut-associated functions enriched in hosts. This indicates that the host might actively shape the composition of its microbiome. Overall, this study provides a framework for studying microbiome assembly dynamics and yields new insights into C. elegans microbiome functions.
Collapse
Affiliation(s)
- Johannes Zimmermann
- Research Group Evolutionary Ecology and Genetics, Zoological Institute, Kiel University, Kiel, Germany
- Max Planck Fellow Group Antibiotic Resistance Evolution, Max Planck Institute for Evolutionary Biology, Ploen, Germany
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Agnes Piecyk
- Research Group Evolutionary Ecology and Genetics, Zoological Institute, Kiel University, Kiel, Germany
| | - Michael Sieber
- Department for Evolutionary Theory, Max Planck Institute for Evolutionary Biology, Ploen, Germany
| | - Carola Petersen
- Research Group Evolutionary Ecology and Genetics, Zoological Institute, Kiel University, Kiel, Germany
| | - Julia Johnke
- Research Group Evolutionary Ecology and Genetics, Zoological Institute, Kiel University, Kiel, Germany
| | - Lucas Moitinho-Silva
- />Institute of Clinical Molecular Biology, Christian-Albrechts University, Kiel, Germany
| | - Sven Künzel
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, Ploen, Germany
| | - Lena Bluhm
- Research Group Evolutionary Ecology and Genetics, Zoological Institute, Kiel University, Kiel, Germany
| | - Arne Traulsen
- Department for Evolutionary Theory, Max Planck Institute for Evolutionary Biology, Ploen, Germany
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Hinrich Schulenburg
- Research Group Evolutionary Ecology and Genetics, Zoological Institute, Kiel University, Kiel, Germany
- Max Planck Fellow Group Antibiotic Resistance Evolution, Max Planck Institute for Evolutionary Biology, Ploen, Germany
| |
Collapse
|
9
|
Singh A, Luallen RJ. Understanding the factors regulating host-microbiome interactions using Caenorhabditis elegans. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230059. [PMID: 38497260 PMCID: PMC10945399 DOI: 10.1098/rstb.2023.0059] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 01/01/2024] [Indexed: 03/19/2024] Open
Abstract
The Human Microbiome Project was a research programme that successfully identified associations between microbial species and healthy or diseased individuals. However, a major challenge identified was the absence of model systems for studying host-microbiome interactions, which would increase our capacity to uncover molecular interactions, understand organ-specificity and discover new microbiome-altering health interventions. Caenorhabditis elegans has been a pioneering model organism for over 70 years but was largely studied in the absence of a microbiome. Recently, ecological sampling of wild nematodes has uncovered a large amount of natural genetic diversity as well as a slew of associated microbiota. The field has now explored the interactions of C. elegans with its associated gut microbiome, a defined and non-random microbial community, highlighting its suitability for dissecting host-microbiome interactions. This core microbiome is being used to study the impact of host genetics, age and stressors on microbiome composition. Furthermore, single microbiome species are being used to dissect molecular interactions between microbes and the animal gut. Being amenable to health altering genetic and non-genetic interventions, C. elegans has emerged as a promising system to generate and test new hypotheses regarding host-microbiome interactions, with the potential to uncover novel paradigms relevant to other systems. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- Anupama Singh
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Robert J. Luallen
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| |
Collapse
|
10
|
Gorelik MG, Yakhnin H, Pannuri A, Walker AC, Pourciau C, Czyz D, Romeo T, Babitzke P. Multitier regulation of the E. coli extreme acid stress response by CsrA. J Bacteriol 2024; 206:e0035423. [PMID: 38319100 PMCID: PMC11210196 DOI: 10.1128/jb.00354-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
CsrA is an RNA-binding protein that regulates processes critical for growth and survival, including central carbon metabolism, motility, biofilm formation, stress responses, and expression of virulence factors in pathogens. Transcriptomics studies in Escherichia coli suggested that CsrA repressed genes involved in surviving extremely acidic conditions. Here, we examine the effects of disrupting CsrA-dependent regulation on the expression of genes and circuitry for acid stress survival and demonstrate CsrA-mediated repression at multiple levels. We show that this repression is critical for managing the trade-off between growth and survival; overexpression of acid stress genes caused by csrA disruption enhances survival under extreme acidity but is detrimental for growth under mildly acidic conditions. In vitro studies confirmed that CsrA binds specifically to mRNAs of structural and regulatory genes for acid stress survival, causing translational repression. We also found that translation of the top-tier acid stress regulator, evgA, is coupled to that of a small leader peptide, evgL, which is repressed by CsrA. Unlike dedicated acid stress response genes, csrA and its sRNA antagonists, csrB and csrC, did not exhibit a substantial response to acid shock. Furthermore, disruption of CsrA regulation of acid stress genes impacted host-microbe interactions in Caenorhabditis elegans, alleviating GABA deficiencies. This study expands the known regulon of CsrA to genes of the extreme acid stress response of E. coli and highlights a new facet of the global role played by CsrA in balancing the opposing physiological demands of stress resistance with the capacity for growth and modulating host interactions.IMPORTANCETo colonize/infect the mammalian intestinal tract, bacteria must survive exposure to the extreme acidity of the stomach. E. coli does this by expressing proteins that neutralize cytoplasmic acidity and cope with molecular damage caused by low pH. Because of the metabolic cost of these processes, genes for surviving acid stress are tightly regulated. Here, we show that CsrA negatively regulates the cascade of expression responsible for the acid stress response. Increased expression of acid response genes due to csrA disruption improved survival at extremely low pH but inhibited growth under mildly acidic conditions. Our findings define a new layer of regulation in the acid stress response of E. coli and a novel physiological function for CsrA.
Collapse
Affiliation(s)
- Mark G. Gorelik
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida, USA
| | - Helen Yakhnin
- Department of Biochemistry and Molecular Biology, Center for RNA Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Archana Pannuri
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida, USA
| | - Alyssa C. Walker
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida, USA
| | - Christine Pourciau
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida, USA
- Department of Biochemistry and Molecular Biology, Center for RNA Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Daniel Czyz
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida, USA
| | - Tony Romeo
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida, USA
| | - Paul Babitzke
- Department of Biochemistry and Molecular Biology, Center for RNA Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
11
|
Wesseltoft JB, Danielsen CD, Andersen AM, de Jonge N, Olsen A, Rohde PD, Kristensen TN. Feeding Drosophila gut microbiomes from young and old flies modifies the microbiome. Sci Rep 2024; 14:7799. [PMID: 38565609 PMCID: PMC10987527 DOI: 10.1038/s41598-024-58500-1] [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: 01/10/2024] [Accepted: 03/30/2024] [Indexed: 04/04/2024] Open
Abstract
It is becoming increasingly evident that the myriad of microbes in the gut, within cells and attached to body parts (or roots of plants), play crucial roles for the host. Although this has been known for decades, recent developments in molecular biology allow for expanded insight into the abundance and function of these microbes. Here we used the vinegar fly, Drosophila melanogaster, to investigate fitness measures across the lifetime of flies fed a suspension of gut microbes harvested from young or old flies, respectively. Our hypothesis was that flies constitutively enriched with a 'Young microbiome' would live longer and be more agile at old age (i.e. have increased healthspan) compared to flies enriched with an 'Old microbiome'. Three major take home messages came out of our study: (1) the gut microbiomes of young and old flies differ markedly; (2) feeding flies with Young and Old microbiomes altered the microbiome of recipient flies and (3) the two different microbial diets did not have any effect on locomotor activity nor lifespan of the recipient flies, contradicting our working hypothesis. Combined, these results provide novel insight into the interplay between hosts and their microbiomes and clearly highlight that the phenotypic effects of gut transplants and probiotics can be complex and unpredictable.
Collapse
Affiliation(s)
| | | | | | - Nadieh de Jonge
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Anders Olsen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Palle Duun Rohde
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
12
|
Zou X, Zou G, Zou X, Wang K, Chen Z. Gut microbiota and its metabolites in Alzheimer's disease: from pathogenesis to treatment. PeerJ 2024; 12:e17061. [PMID: 38495755 PMCID: PMC10944166 DOI: 10.7717/peerj.17061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/15/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction An increasing number of studies have demonstrated that altered microbial diversity and function (such as metabolites), or ecological disorders, regulate bowel-brain axis involvement in the pathophysiologic processes in Alzheimer's disease (AD). The dysregulation of microbes and their metabolites can be a double-edged sword in AD, presenting the possibility of microbiome-based treatment options. This review describes the link between ecological imbalances and AD, the interactions between AD treatment modalities and the microbiota, and the potential of interventions such as prebiotics, probiotics, synbiotics, fecal microbiota transplantation, and dietary interventions as complementary therapeutic strategies targeting AD pathogenesis and progression. Survey methodology Articles from PubMed and china.com on intestinal flora and AD were summarized to analyze the data and conclusions carefully to ensure the comprehensiveness, completeness, and accuracy of this review. Conclusions Regulating the gut flora ecological balance upregulates neurotrophic factor expression, regulates the microbiota-gut-brain (MGB) axis, and suppresses the inflammatory responses. Based on emerging research, this review explored novel directions for future AD research and clinical interventions, injecting new vitality into microbiota research development.
Collapse
Affiliation(s)
- Xinfu Zou
- Subject of Integrated Chinese and Western Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Guoqiang Zou
- Subject of Traditional Chinese Medicine, Shandong University Of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinyan Zou
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
| | - Kangfeng Wang
- Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zetao Chen
- Subject of Integrated Chinese and Western Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
13
|
Israelson H, Vedsted-Jakobsen A, Zhu L, Gagnaire A, von Münchow A, Polakovicova N, Valente AH, Raza A, Andersen-Civil AIS, Olsen JE, Myhill LJ, Geldhof P, Williams AR. Diet composition drives tissue-specific intensity of murine enteric infections. mBio 2024; 15:e0260323. [PMID: 38179939 PMCID: PMC10865784 DOI: 10.1128/mbio.02603-23] [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: 09/25/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Diet composition plays a large role in regulating gut health and enteric infection. In particular, synthetic "Western-style" diets may predispose to disease, while whole-grain diets containing high levels of crude fiber are thought to promote gut health. Here, we show that, in contrast to this paradigm, mice fed with unrefined chow are significantly more susceptible to infection with Trichuris muris, a caecum-dwelling nematode, than mice fed with refined, semi-synthetic diets (SSDs). Moreover, mice fed with SSD supplemented with inulin, a fermentable fiber, developed chronic T. muris burdens, whereas mice fed with SSD efficiently cleared the infection. Diet composition significantly impacted infection-induced changes in the host gut microbiome. Mice infected with the bacterium Citrobacter rodentium were also more susceptible to pathogen colonization when fed with either chow or inulin-enriched SSD. However, transcriptomic analysis of tissues from mice fed with either SSD or inulin-enriched SSD revealed that, in contrast to T. muris, increased C. rodentium infection appeared to be independent of the host immune response. Accordingly, exogenous treatment with interleukin (IL)-25 reduced T. muris burdens in inulin-fed mice, whereas IL-22 treatment was unable to restore resistance to C. rodentium colonization. Diet-mediated effects on pathogen burden were more pronounced for large intestine-dwelling pathogens, as effects on small the intestinal helminth (Heligmosomoides polygyrus) were less evident, and protozoan (Giardia muris) infection burdens were equivalent in mice fed with chow, inulin-enriched SSD, or SSD, despite higher cyst excretion in chow-fed mice. Collectively, our results point to a tissue- and pathogen-restricted effect of dietary fiber levels on enteric infection intensity.IMPORTANCEEnteric infections induce dysbiosis and inflammation and are a major public health burden. As the gut environment is strongly shaped by diet, the role of different dietary components in promoting resistance to infection is of interest. While diets rich in fiber or whole grain are normally associated with improved gut health, we show here that these components predispose the host to higher levels of pathogen infection. Thus, our results have significance for interpreting how different dietary interventions may impact on gastrointestinal infections. Moreover, our results may shed light on our understanding of how gut flora and mucosal immune function is influenced by the food that we eat.
Collapse
Affiliation(s)
- Helene Israelson
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Amalie Vedsted-Jakobsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ling Zhu
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Aurelie Gagnaire
- Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Laboratory of Parasitology, Ghent University, Merelbeke, Belgium
| | - Alexandra von Münchow
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Nina Polakovicova
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Angela H. Valente
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ali Raza
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Audrey I. S. Andersen-Civil
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - John E. Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Laura J. Myhill
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Peter Geldhof
- Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Laboratory of Parasitology, Ghent University, Merelbeke, Belgium
| | - Andrew R. Williams
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
14
|
Lai TT, Tsai YH, Liou CW, Fan CH, Hou YT, Yao TH, Chuang HL, Wu WL. The gut microbiota modulate locomotion via vagus-dependent glucagon-like peptide-1 signaling. NPJ Biofilms Microbiomes 2024; 10:2. [PMID: 38228675 DOI: 10.1038/s41522-024-00477-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024] Open
Abstract
Locomotor activity is an innate behavior that can be triggered by gut-motivated conditions, such as appetite and metabolic condition. Various nutrient-sensing receptors distributed in the vagal terminal in the gut are crucial for signal transduction from the gut to the brain. The levels of gut hormones are closely associated with the colonization status of the gut microbiota, suggesting a complicated interaction among gut bacteria, gut hormones, and the brain. However, the detailed mechanism underlying gut microbiota-mediated endocrine signaling in the modulation of locomotion is still unclear. Herein, we show that broad-spectrum antibiotic cocktail (ABX)-treated mice displayed hypolocomotion and elevated levels of the gut hormone glucagon-like peptide-1 (GLP-1). Blockade of the GLP-1 receptor and subdiaphragmatic vagal transmission rescued the deficient locomotor phenotype in ABX-treated mice. Activation of the GLP-1 receptor and vagal projecting brain regions led to hypolocomotion. Finally, selective antibiotic treatment dramatically increased serum GLP-1 levels and decreased locomotion. Colonizing Lactobacillus reuteri and Bacteroides thetaiotaomicron in microbiota-deficient mice suppressed GLP-1 levels and restored the hypolocomotor phenotype. Our findings identify a mechanism by which specific gut microbes mediate host motor behavior via the enteroendocrine and vagal-dependent neural pathways.
Collapse
Affiliation(s)
- Tzu-Ting Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Hsuan Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Chia-Wei Liou
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Tian Hou
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Tzu-Hsuan Yao
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, 115202, Taiwan
| | - Wei-Li Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
| |
Collapse
|
15
|
Zaplana T, Miele S, Tolonen AC. Lachnospiraceae are emerging industrial biocatalysts and biotherapeutics. Front Bioeng Biotechnol 2024; 11:1324396. [PMID: 38239921 PMCID: PMC10794557 DOI: 10.3389/fbioe.2023.1324396] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024] Open
Abstract
The Lachnospiraceae is a family of anaerobic bacteria in the class Clostridia with potential to advance the bio-economy and intestinal therapeutics. Some species of Lachnospiraceae metabolize abundant, low-cost feedstocks such as lignocellulose and carbon dioxide into value-added chemicals. Others are among the dominant species of the human colon and animal rumen, where they ferment dietary fiber to promote healthy gut and immune function. Here, we summarize recent studies of the physiology, cultivation, and genetics of Lachnospiraceae, highlighting their wide substrate utilization and metabolic products with industrial applications. We examine studies of these bacteria as Live Biotherapeutic Products (LBPs), focusing on in vivo disease models and clinical studies using them to treat infection, inflammation, metabolic syndrome, and cancer. We discuss key research areas including elucidation of intra-specific diversity and genetic modification of candidate strains that will facilitate the exploitation of Lachnospiraceae in industry and medicine.
Collapse
Affiliation(s)
| | | | - Andrew C. Tolonen
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry, Université Paris-Saclay, Evry, France
| |
Collapse
|
16
|
Khoshnan A. Gut Microbiota as a Modifier of Huntington's Disease Pathogenesis. J Huntingtons Dis 2024; 13:133-147. [PMID: 38728199 PMCID: PMC11307070 DOI: 10.3233/jhd-240012] [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] [Accepted: 04/07/2024] [Indexed: 05/12/2024]
Abstract
Huntingtin (HTT) protein is expressed in most cell lineages, and the toxicity of mutant HTT in multiple organs may contribute to the neurological and psychiatric symptoms observed in Huntington's disease (HD). The proteostasis and neurotoxicity of mutant HTT are influenced by the intracellular milieu and responses to environmental signals. Recent research has highlighted a prominent role of gut microbiota in brain and immune system development, aging, and the progression of neurological disorders. Several studies suggest that mutant HTT might disrupt the homeostasis of gut microbiota (known as dysbiosis) and impact the pathogenesis of HD. Dysbiosis has been observed in HD patients, and in animal models of the disease it coincides with mutant HTT aggregation, abnormal behaviors, and reduced lifespan. This review article aims to highlight the potential toxicity of mutant HTT in organs and pathways within the microbiota-gut-immune-central nervous system (CNS) axis. Understanding the functions of Wild-Type (WT) HTT and the toxicity of mutant HTT in these organs and the associated networks may elucidate novel pathogenic pathways, identify biomarkers and peripheral therapeutic targets for HD.
Collapse
Affiliation(s)
- Ali Khoshnan
- Keck School of Medicine, Physiology and Neuroscience, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Bishoyi AK, Lakra A, Mandhata CP, Sahoo CR, Padhy RN. Prospective Phycocompounds for Developing Therapeutics for Urinary Tract Infection. Curr Microbiol 2023; 81:35. [PMID: 38063889 DOI: 10.1007/s00284-023-03535-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 10/23/2023] [Indexed: 12/18/2023]
Abstract
Antibiotic resistance of bacteria is causing clinical and public health concerns that are challenging to treat. Infections are becoming more common in the present era, and patients admitted to hospitals often have drug-resistant bacteria that can spread nosocomial infections. Urinary tract infections (UTIs) are among the most common infectious diseases affecting all age groups. There has been an increase in the proportion of bacteria that are resistant to multiple drugs. Herein is a comprehensive update on UTI-associated diseases: cystitis, urethritis, acute urethral syndrome, pyelonephritis, and recurrent UTIs. Further emphasis on the global statistical incidence and recent advancement of the role of natural products in treating notorious infections are described. This updated compendium will inspire the development of novel phycocompounds as the prospective antibacterial candidate.
Collapse
Affiliation(s)
- Ajit Kumar Bishoyi
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan Deemed to Be University, Bhubaneswar, Odisha, 751003, India
| | - Anjali Lakra
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan Deemed to Be University, Bhubaneswar, Odisha, 751003, India
| | - Chinmayee Priyadarsani Mandhata
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan Deemed to Be University, Bhubaneswar, Odisha, 751003, India
| | - Chita Ranjan Sahoo
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan Deemed to Be University, Bhubaneswar, Odisha, 751003, India.
| | - Rabindra Nath Padhy
- Central Research Laboratory, Institute of Medical Sciences & Sum Hospital, Siksha 'O' Anusandhan Deemed to Be University, Bhubaneswar, Odisha, 751003, India.
| |
Collapse
|
18
|
Walker AC, Bhargava R, Bucher M, Brust AS, Czy DM. Identification of proteotoxic and proteoprotective bacteria that non-specifically affect proteins associated with neurodegenerative diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563685. [PMID: 37961318 PMCID: PMC10634778 DOI: 10.1101/2023.10.24.563685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Neurodegenerative protein conformational diseases (PCDs), such as Alzheimer's, Parkinson's, and Huntington's, are a leading cause of death and disability worldwide and have no known cures or effective treatments. Emerging evidence suggests a role for the gut microbiota in the pathogenesis of neurodegenerative PCDs; however, the influence of specific bacteria on the culprit proteins associated with each of these diseases remains elusive, primarily due to the complexity of the microbiota. In the present study, we employed a single-strain screening approach to identify human bacterial isolates that enhance or suppress the aggregation of culprit proteins and the associated toxicity in Caenorhabditis elegans expressing Aβ1-42, α-synuclein, and polyglutamine tracts. Here, we reveal the first comprehensive analysis of the human microbiome for its effect on proteins associated with neurodegenerative diseases. Our results suggest that bacteria affect the aggregation of metastable proteins by modulating host proteostasis rather than selectively targeting specific disease-associated proteins. These results reveal bacteria that potentially influence the pathogenesis of PCDs and open new promising prevention and treatment opportunities by altering the abundance of beneficial and detrimental microbes.
Collapse
Affiliation(s)
- Alyssa C Walker
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Rohan Bhargava
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Michael Bucher
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Amanda S Brust
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Daniel M Czy
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
19
|
Walker A, Czyz DM. Oh my gut! Is the microbial origin of neurodegenerative diseases real? Infect Immun 2023; 91:e0043722. [PMID: 37750713 PMCID: PMC10580905 DOI: 10.1128/iai.00437-22] [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] [Indexed: 09/27/2023] Open
Abstract
There is no cure or effective treatment for neurodegenerative protein conformational diseases (PCDs), such as Alzheimer's or Parkinson's diseases, mainly because the etiology of these diseases remains elusive. Recent data suggest that unique changes in the gut microbial composition are associated with these ailments; however, our current understanding of the bacterial role in the pathogenesis of PCDs is hindered by the complexity of the microbial communities associated with specific microbiomes, such as the gut, oral, or vaginal microbiota. The composition of these specific microbiomes is regarded as a unique fingerprint affected by factors such as infections, diet, lifestyle, and antibiotics. All of these factors also affect the severity of neurodegenerative diseases. The majority of studies that reveal microbial contribution are correlational, and various models, including worm, fly, and mouse, are being utilized to decipher the role of individual microbes that may affect disease onset and progression. Recent evidence from across model organisms and humans shows a positive correlation between the presence of gram-negative enteropathogenic bacteria and the pathogenesis of PCDs. While these correlational studies do not provide a mechanistic explanation, they do reveal contributing bacterial species and provide an important basis for further investigation. One of the lurking concerns related to the microbial contribution to PCDs is the increasing prevalence of antibiotic resistance and poor antibiotic stewardship, which ultimately select for proteotoxic bacteria, especially the gram-negative species that are known for intrinsic resistance. In this review, we summarize what is known about individual microbial contribution to PCDs and the potential impact of increasing antimicrobial resistance.
Collapse
Affiliation(s)
- Alyssa Walker
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Daniel M. Czyz
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
20
|
Abstract
After decades of research, our knowledge of the complexity of cancer mechanisms, elegantly summarized as 'hallmarks of cancer', is expanding, as are the therapeutic opportunities that this knowledge brings. However, cancer still needs intense research to diminish its tremendous impact. In this context, the use of simple model organisms such as Caenorhabditis elegans, in which the genetics of the apoptotic pathway was discovered, can facilitate the investigation of several cancer hallmarks. Amenable for genetic and drug screens, convenient for fast and efficient genome editing, and aligned with the 3Rs ('Replacement, Reduction and Refinement') principles for ethical animal research, C. elegans plays a significant role in unravelling the intricate network of cancer mechanisms and presents a promising option in clinical diagnosis and drug discovery.
Collapse
Affiliation(s)
- Julián Cerón
- Modeling Human Diseases in C. elegans Group – Genes, Disease and Therapy Program, Bellvitge Biomedical Research Institute – IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
21
|
SAITO R, SATO N, OKINO Y, WANG DS, SEO G. Bacillus subtilis TO-A extends the lifespan of Caenorhabditis elegans. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 42:124-130. [PMID: 37016687 PMCID: PMC10067327 DOI: 10.12938/bmfh.2022-057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/12/2022] [Indexed: 06/19/2023]
Abstract
Clostridium butyricum TO-A, Enterococcus faecium T-110, and Bacillus subtilis TO-A are sold as oral probiotic preparations and reportedly exhibit many beneficial effects on the health of hosts, including humans and livestock. In this study, we compared the ability of these clinically applied probiotic bacteria with Escherichia coli OP50 in extending the lifespan of Caenorhabditis elegans. To compare the C. elegans lifespan-extending effects of the three bacteria, experiments were performed using a nematode growth medium containing a small amount of trypticase soy agar. The maximum lifespans of worms fed C. butyricum TO-A, E. faecium T-110, or B. subtilis TO-A increased by 11, 12, and 26%, respectively, compared with worms fed E. coli OP50. In addition, we conducted a metabolomic analysis of methanol extracts of B. subtilis TO-A cells, which exhibited the strongest lifespan-extending effect on C. elegans among the probiotic bacteria tested in this study. As a result, 59 candidate substances involved in extending the lifespan of C. elegans were identified in B. subtilis TO-A cells.
Collapse
Affiliation(s)
- Ryuichi SAITO
- Bioscience R&D Department, TOA Biopharma Co., Ltd., 606
Kondoh-cho, Tatebayashi, Gunma 374-0042, Japan
| | - Naoki SATO
- Bioscience R&D Department, TOA Biopharma Co., Ltd., 606
Kondoh-cho, Tatebayashi, Gunma 374-0042, Japan
| | - Yoichi OKINO
- Bioscience R&D Department, TOA Biopharma Co., Ltd., 606
Kondoh-cho, Tatebayashi, Gunma 374-0042, Japan
| | - Dian-Sheng WANG
- Bioscience R&D Department, TOA Biopharma Co., Ltd., 606
Kondoh-cho, Tatebayashi, Gunma 374-0042, Japan
| | - Genichiro SEO
- Bioscience R&D Department, TOA Biopharma Co., Ltd., 606
Kondoh-cho, Tatebayashi, Gunma 374-0042, Japan
| |
Collapse
|
22
|
Feng M, Gao B, Garcia LR, Sun Q. Microbiota-derived metabolites in regulating the development and physiology of Caenorhabditis elegans. Front Microbiol 2023; 14:1035582. [PMID: 36925470 PMCID: PMC10011103 DOI: 10.3389/fmicb.2023.1035582] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/09/2023] [Indexed: 03/08/2023] Open
Abstract
Microbiota consist of microorganisms that provide essential health benefits and contribute to the animal's physiological homeostasis. Microbiota-derived metabolites are crucial mediators in regulating host development, system homeostasis, and overall fitness. In this review, by focusing on the animal model Caenorhabditis elegans, we summarize key microbial metabolites and their molecular mechanisms that affect animal development. We also provide, from a bacterial perspective, an overview of host-microbiota interaction networks used for maintaining host physiological homeostasis. Moreover, we discuss applicable methodologies for profiling new bacterial metabolites that modulate host developmental signaling pathways. Microbiota-derived metabolites have the potential to be diagnostic biomarkers for diseases, as well as promising targets for engineering therapeutic interventions against animal developmental or health-related defects.
Collapse
Affiliation(s)
- Min Feng
- Department of Chemical Engineering, Texas A&M University, College Station, TX, United States
| | - Baizhen Gao
- Department of Chemical Engineering, Texas A&M University, College Station, TX, United States
| | - L Rene Garcia
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Qing Sun
- Department of Chemical Engineering, Texas A&M University, College Station, TX, United States
| |
Collapse
|
23
|
Abdugheni R, Wang W, Wang Y, Du M, Liu F, Zhou N, Jiang C, Wang C, Wu L, Ma J, Liu C, Liu S. Metabolite profiling of human-originated Lachnospiraceae at the strain level. IMETA 2022; 1:e58. [PMID: 38867908 PMCID: PMC10989990 DOI: 10.1002/imt2.58] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 06/14/2024]
Abstract
The human gastrointestinal (GI) tract harbors diverse microbes, and the family Lachnospiraceae is one of the most abundant and widely occurring bacterial groups in the human GI tract. Beneficial and adverse effects of the Lachnospiraceae on host health were reported, but the diversities at species/strain levels as well as their metabolites of Lachnospiraceae have been, so far, not well documented. In the present study, we report on the collection of 77 human-originated Lachnospiraceae species (please refer hLchsp, https://hgmb.nmdc.cn/subject/lachnospiraceae) and the in vitro metabolite profiles of 110 Lachnospiraceae strains (https://hgmb.nmdc.cn/subject/lachnospiraceae/metabolites). The Lachnospiraceae strains in hLchsp produced 242 metabolites of 17 categories. The larger categories were alcohols (89), ketones (35), pyrazines (29), short (C2-C5), and long (C > 5) chain acids (31), phenols (14), aldehydes (14), and other 30 compounds. Among them, 22 metabolites were aromatic compounds. The well-known beneficial gut microbial metabolite, butyric acid, was generally produced by many Lachnospiraceae strains, and Agathobacter rectalis strain Lach-101 and Coprococcus comes strain NSJ-173 were the top 2 butyric acid producers, as 331.5 and 310.9 mg/L of butyric acids were produced in vitro, respectively. Further analysis of the publicly available cohort-based volatile-metabolomic data sets of human feces revealed that over 30% of the prevailing volatile metabolites were covered by Lachnospiraceae metabolites identified in this study. This study provides Lachnospiraceae strain resources together with their metabolic profiles for future studies on host-microbe interactions and developments of novel probiotics or biotherapies.
Collapse
Affiliation(s)
- Rashidin Abdugheni
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Desert and Oasis EcologyXinjiang Institute of Ecology and Geography, Chinese Academy of SciencesUrumqiChina
| | - Wen‐Zhao Wang
- State Key Laboratory of MycologyInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Yu‐Jing Wang
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Meng‐Xuan Du
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Feng‐Lan Liu
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- College of Life SciencesHebei UniversityBaodingChina
| | - Nan Zhou
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Cheng‐Ying Jiang
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Chang‐Yu Wang
- Colleg of Life SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Linhuan Wu
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Juncai Ma
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Chang Liu
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Shuang‐Jiang Liu
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| |
Collapse
|
24
|
Gao J, Sadiq FA, Zheng Y, Zhao J, He G, Sang Y. Biofilm-based delivery approaches and specific enrichment strategies of probiotics in the human gut. Gut Microbes 2022; 14:2126274. [PMID: 36175161 PMCID: PMC9542427 DOI: 10.1080/19490976.2022.2126274] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The use of probiotics has been one of the effective strategies to restructure perturbed human gut microbiota following a disease or metabolic disorder. One of the biggest challenges associated with the use of probiotic-based gut modulation strategies is to keep the probiotic cells viable and stable during the gastrointestinal transit. Biofilm-based probiotics delivery approaches have emerged as fascinating modes of probiotic delivery in which probiotics show significantly greater tolerance and biotherapeutic potential, and interestingly probiotic biofilms can be developed on food-grade surfaces too, which is ideal for the growth and proliferation of bacterial cells for incorporation into food matrices. In addition, biofilms can be further encapsulated with food-grade materials or with bacterial self-produced biofilms. This review presents a newly emerging and unprecedently discussed techniques for the safe delivery of probiotics based on biofilms and further discusses newly emerging prebiotic materials which target specific gut microbiota groups for growth and proliferation.
Collapse
Affiliation(s)
- Jie Gao
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Faizan Ahmed Sadiq
- Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Technology & Food Sciences Unit, Melle, Belgium
| | - Yixin Zheng
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Jinrong Zhao
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Guoqing He
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China,CONTACT Guoqing He College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Yaxin Sang
- Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China,Yaxin Sang Collge of Food Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
25
|
Suzuki Y, Kikuchi K, Numayama-Tsuruta K, Ishikawa T. Reciprocating intestinal flows enhance glucose uptake in C. elegans. Sci Rep 2022; 12:15310. [PMID: 36130988 PMCID: PMC9492717 DOI: 10.1038/s41598-022-18968-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022] Open
Abstract
Despite its physiological and pathological importance, the mechanical relationship between glucose uptake in the intestine and intestinal flows is unclear. In the intestine of the nematode Caenorhabditis elegans, the defecation motor program (DMP) causes reciprocating intestinal flows. Although the DMP is frequently activated in the intestines, its physiological function is unknown. We evaluated the mechanical signature of enhanced glucose uptake by the DMP in worms. Glucose uptake tended to increase with increasing flow velocity during the DMP because of mechanical mixing and transport. However, the increase in input energy required for the DMP was low compared with the calorie intake. The findings suggest that animals with gastrointestinal motility exploit the reciprocating intestinal flows caused by peristalsis to promote nutrient absorption by intestinal cells.
Collapse
Affiliation(s)
- Yuki Suzuki
- Graduate School of Engineering, Department of Finemechanics, Tohoku University, 6-6-01 Aramaki, Aoba, Sendai, Miyagi, 980-8579, Japan
| | - Kenji Kikuchi
- Graduate School of Engineering, Department of Finemechanics, Tohoku University, 6-6-01 Aramaki, Aoba, Sendai, Miyagi, 980-8579, Japan. .,Graduate School of Biomedical Engineering, Tohoku University, 6-6-01 Aramaki, Aoba, Sendai, Miyagi, 980-8579, Japan.
| | - Keiko Numayama-Tsuruta
- Graduate School of Biomedical Engineering, Tohoku University, 6-6-01 Aramaki, Aoba, Sendai, Miyagi, 980-8579, Japan
| | - Takuji Ishikawa
- Graduate School of Engineering, Department of Finemechanics, Tohoku University, 6-6-01 Aramaki, Aoba, Sendai, Miyagi, 980-8579, Japan.,Graduate School of Biomedical Engineering, Tohoku University, 6-6-01 Aramaki, Aoba, Sendai, Miyagi, 980-8579, Japan
| |
Collapse
|
26
|
Matthewman C, Narin A, Huston H, Hopkins CE. Systems to model the personalized aspects of microbiome health and gut dysbiosis. Mol Aspects Med 2022; 91:101115. [PMID: 36104261 DOI: 10.1016/j.mam.2022.101115] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/03/2022] [Indexed: 01/17/2023]
Abstract
The human gut microbiome is a complex and dynamic microbial entity that interacts with the environment and other parts of the body including the brain, heart, liver, and immune system. These multisystem interactions are highly conserved from invertebrates to humans, however the complexity and diversity of human microbiota compositions often yield a context that is unique to each individual. Yet commonalities remain across species, where a healthy gut microbiome will be rich in symbiotic commensal biota while an unhealthy gut microbiota will be experiencing abnormal blooms of pathobiont bacteria. In this review we discuss how omics technologies can be applied in a personalized approach to understand the microbial crosstalk and microbial-host interactions that affect the delicate balance between eubiosis and dysbiosis in an individual gut microbiome. We further highlight the strengths of model organisms in identifying and characterizing these conserved synergistic and/or pathogenic host-microbe interactions. And finally, we touch upon the growing area of personalized therapeutic interventions targeting gut microbiome.
Collapse
|
27
|
Kim MR, Cho SY, Lee HJ, Kim JY, Nguyen UTT, Ha NM, Choi KY, Cha KH, Kim JH, Kim WK, Kang K. Schisandrin C improves leaky gut conditions in intestinal cell monolayer, organoid, and nematode models by increasing tight junction protein expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 103:154209. [PMID: 35689901 DOI: 10.1016/j.phymed.2022.154209] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/25/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Leaky gut symptoms and inflammatory bowel disease (IBD) are associated with damaged intestinal mucosa, intestinal permeability dysfunction by epithelial cell cytoskeleton contraction, disrupted intercellular tight junction (TJ) protein expression, and abnormal immune responses and are intractable diseases. PURPOSE We evaluated the effects of schisandrin C, a dibenzocyclooctadiene lignan from Schisandra chinensis, on intestinal inflammation and permeability dysfunction in gut mimetic systems: cultured intestinal cells, intestinal organoids, and a Caenorhabditis elegans model. METHODS Schisandrin C was selected from 9 lignan compounds from S. chinensis based on its anti-inflammatory effects in HT-29 human intestinal cells. IL-1β and Pseudomonas aeruginosa supernatants were used to disrupt intestinal barrier formation in vitro and in C. elegans, respectively. The effects of schisandrin C on transepithelial electrical resistance (TEER) and intestinal permeability were evaluated in intestinal cell monolayers, and its effect on intestinal permeability dysfunction was tested in mouse intestinal organoids and C. elegans by measuring fluorescein isothiocyanate (FITC)-dextran efflux. The effect of schisandrin C on TJ protein expression was investigated by western blotting and fluorescence microscopy. The signaling pathway underlying these effects was also elucidated. RESULTS Schisandrin C ameliorated intestinal permeability dysfunction in three IBD model systems and enhanced epithelial barrier formation via upregulation of ZO-1 and occludin in intestinal cell monolayers and intestinal organoids. In Caco-2 cells, schisandrin C restored IL-1β-mediated increases in MLCK and p-MLC expression, in turn blocking cytoskeletal contraction and subsequent intestinal permeabilization. Schisandrin C inhibited NF-ĸB and p38 MAPK signaling, which regulates MLCK expression and structural reorganization of the TJ complex in Caco-2 cells. Schisandrin C significantly improved abnormal FITC-dextran permeabilization in both intestinal organoids and C. elegans. CONCLUSION Schisandrin C significantly improves abnormal intestinal permeability and regulates the expression of TJ proteins, long MLCK, p-MLC, and inflammation-related proteins, which are closely related to leaky gut symptoms and IBD development. Therefore, schisandrin C is a candidate to treat leaky gut symptoms and IBDs.
Collapse
Affiliation(s)
- Mi Ri Kim
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea; Department of Aquatic Life Medicine, Gangneung-Wonju National University, Gangneung, Gangwon-do, 25457, South Korea
| | - Su-Yeon Cho
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Gangneung, Gangwon-do, 25451, South Korea
| | - Hee Ju Lee
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea
| | - Joo Yeon Kim
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea
| | - Uyen Tran Tu Nguyen
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea
| | - Ngoc Minh Ha
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Gangneung, Gangwon-do, 25451, South Korea
| | - Ki Young Choi
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Gangneung, Gangwon-do, 25451, South Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea
| | - Jeong-Ho Kim
- Department of Aquatic Life Medicine, Gangneung-Wonju National University, Gangneung, Gangwon-do, 25457, South Korea
| | - Won Kyu Kim
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea.
| | - Kyungsu Kang
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, 25451, South Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Gangneung, Gangwon-do, 25451, South Korea.
| |
Collapse
|
28
|
Chongtham A, Yoo JH, Chin TM, Akingbesote ND, Huda A, Marsh JL, Khoshnan A. Gut Bacteria Regulate the Pathogenesis of Huntington's Disease in Drosophila Model. Front Neurosci 2022; 16:902205. [PMID: 35757549 PMCID: PMC9215115 DOI: 10.3389/fnins.2022.902205] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022] Open
Abstract
Changes in the composition of gut microbiota are implicated in the pathogenesis of several neurodegenerative disorders. Here, we investigated whether gut bacteria affect the progression of Huntington’s disease (HD) in transgenic Drosophila melanogaster (fruit fly) models expressing full-length or N-terminal fragments of human mutant huntingtin (HTT) protein. We find that elimination of commensal gut bacteria by antibiotics reduces the aggregation of amyloidogenic N-terminal fragments of HTT and delays the development of motor defects. Conversely, colonization of HD flies with Escherichia coli (E. coli), a known pathobiont of human gut with links to neurodegeneration and other morbidities, accelerates HTT aggregation, aggravates immobility, and shortens lifespan. Similar to antibiotics, treatment of HD flies with small compounds such as luteolin, a flavone, or crocin a beta-carotenoid, ameliorates disease phenotypes, and promotes survival. Crocin prevents colonization of E. coli in the gut and alters the levels of commensal bacteria, which may be linked to its protective effects. The opposing effects of E. coli and crocin on HTT aggregation, motor defects, and survival in transgenic Drosophila models support the involvement of gut-brain networks in the pathogenesis of HD.
Collapse
Affiliation(s)
- Anjalika Chongtham
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - Jung Hyun Yoo
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - Theodore M Chin
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - Ngozi D Akingbesote
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - Ainul Huda
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| | - J Lawrence Marsh
- Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Ali Khoshnan
- Biology and Bioengineering, California Institute of Technology (Caltech), Pasadena, CA, United States
| |
Collapse
|
29
|
Walker AC, Bhargava R, Brust AS, Owji AA, Czyż DM. Time-off-pick Assay to Measure Caenorhabditis elegans Motility. Bio Protoc 2022; 12:e4436. [PMID: 35864904 PMCID: PMC9257836 DOI: 10.21769/bioprotoc.4436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 12/29/2022] Open
Abstract
Caenorhabditis elegans is a simple metazoan that is often used as a model organism to study various human ailments with impaired motility phenotypes, including protein conformational diseases. Numerous motility assays that measure neuro-muscular function have been employed using C. elegans . Here, we describe "time-off-pick" (TOP), a novel assay for assessing motility in C. elegans . TOP is conducted by sliding an eyebrow hair under the mid-section of the worm and counting the number of seconds it takes for the worm to crawl completely off. The time it takes for the worm to crawl off the eyebrow hair is proportional to the severity of its motility defect. Other readouts of motility include crawling or swimming phenotypes, and although widely established, have some limitations. For example, worms that are roller mutants are less suitable for crawling or swimming assays. We demonstrated that our novel TOP assay is sensitive to age-dependent changes in motility, thus, providing another more inclusive method to assess motor function in C. elegans . Graphical abstract: Conceptual overview of the "time-off-pick" (TOP) assay. Various C. elegans models exhibit age-dependent defects in motility. The time it takes for a worm to crawl off of an eyebrow pick that is slid under its mid-section is measured in TOP seconds. A greater TOP is indicative of a greater motility defect. Eventually, worms with phenotypes that lead to paralysis will not be able to leave the pick.
Collapse
Affiliation(s)
- Alyssa C. Walker
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Rohan Bhargava
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Amanda S. Brust
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Ali A. Owji
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Daniel M. Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
,
*For correspondence:
| |
Collapse
|
30
|
Walker AC, Bhargava R, Dove AS, Brust AS, Owji AA, Czyż DM. Bacteria-Derived Protein Aggregates Contribute to the Disruption of Host Proteostasis. Int J Mol Sci 2022; 23:4807. [PMID: 35563197 PMCID: PMC9103901 DOI: 10.3390/ijms23094807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/14/2022] [Accepted: 04/24/2022] [Indexed: 12/10/2022] Open
Abstract
Neurodegenerative protein conformational diseases are characterized by the misfolding and aggregation of metastable proteins encoded within the host genome. The host is also home to thousands of proteins encoded within exogenous genomes harbored by bacteria, fungi, and viruses. Yet, their contributions to host protein-folding homeostasis, or proteostasis, remain elusive. Recent studies, including our previous work, suggest that bacterial products contribute to the toxic aggregation of endogenous host proteins. We refer to these products as bacteria-derived protein aggregates (BDPAs). Furthermore, antibiotics were recently associated with an increased risk for neurodegenerative diseases, including Parkinson's disease and amyotrophic lateral sclerosis-possibly by virtue of altering the composition of the human gut microbiota. Other studies have shown a negative correlation between disease progression and antibiotic administration, supporting their protective effect against neurodegenerative diseases. These contradicting studies emphasize the complexity of the human gut microbiota, the gut-brain axis, and the effect of antibiotics. Here, we further our understanding of bacteria's effect on host protein folding using the model Caenorhabditis elegans. We employed genetic and chemical methods to demonstrate that the proteotoxic effect of bacteria on host protein folding correlates with the presence of BDPAs. Furthermore, the abundance and proteotoxicity of BDPAs are influenced by gentamicin, an aminoglycoside antibiotic that induces protein misfolding, and by butyrate, a short-chain fatty acid that we previously found to affect host protein aggregation and the associated toxicity. Collectively, these results increase our understanding of host-bacteria interactions in the context of protein conformational diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel M. Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA; (A.C.W.); (R.B.); (A.S.D.); (A.S.B.); (A.A.O.)
| |
Collapse
|
31
|
Czyż DM. Exploiting Caenorhabditis elegans to discover human gut microbiota-mediated intervention strategies in protein conformational diseases. Neural Regen Res 2022; 17:2203-2204. [PMID: 35259831 PMCID: PMC9083164 DOI: 10.4103/1673-5374.335788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Daniel M Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| |
Collapse
|
32
|
Mousa WK, Chehadeh F, Husband S. Recent Advances in Understanding the Structure and Function of the Human Microbiome. Front Microbiol 2022; 13:825338. [PMID: 35185849 PMCID: PMC8851206 DOI: 10.3389/fmicb.2022.825338] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Trillions of microbes live within our bodies in a deep symbiotic relationship. Microbial populations vary across body sites, driven by differences in the environment, immunological factors, and interactions between microbial species. Major advances in genome sequencing enable a better understanding of microbiome composition. However, most of the microbial taxa and species of the human microbiome are still unknown. Without revealing the identity of these microbes as a first step, we cannot appreciate their role in human health and diseases. A shift in the microbial balance, termed dysbiosis, is linked to a broad range of diseases from simple colitis and indigestion to cancer and dementia. The last decade has witnessed an explosion in microbiome research that led to a better understanding of the microbiome structure and function. This understanding leads to potential opportunities to develop next-generation microbiome-based drugs and diagnostic biomarkers. However, our understanding is limited given the highly personalized nature of the microbiome and its complex and multidirectional interactions with the host. In this review, we discuss: (1) our current knowledge of microbiome structure and factors that shape the microbial composition, (2) recent associations between microbiome dysbiosis and diseases, and (3) opportunities of new microbiome-based therapeutics. We analyze common themes, promises, gaps, and challenges of the microbiome research.
Collapse
Affiliation(s)
- Walaa K. Mousa
- College of Pharmacy, Al Ain University of Science and Technology, Al Ain, United Arab Emirates
- Department of Biology, Whitman College, Walla Walla, WA, United States
- College of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Fadia Chehadeh
- Department of Biology, Whitman College, Walla Walla, WA, United States
| | - Shannon Husband
- Department of Biology, Whitman College, Walla Walla, WA, United States
| |
Collapse
|
33
|
De Pablo-Fernandez E, Gebeyehu GG, Flain L, Slater R, Frau A, Ijaz UZ, Warner T, Probert C. The faecal metabolome and mycobiome in Parkinson's disease. Parkinsonism Relat Disord 2022; 95:65-69. [PMID: 35045378 DOI: 10.1016/j.parkreldis.2022.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/07/2021] [Accepted: 01/07/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Gut fungal composition and its metabolites have not been assessed simultaneously in Parkinson's disease (PD) despite their potential pathogenic contribution. OBJECTIVE To evaluate the faecal metabolome and mycobiome in PD by assessing volatile organic compounds (VOCs) and fungal rRNA. METHODS Faecal VOCs from 35 PD patients and two control groups (n = 35; n = 15) were assessed using gas chromatography and mass spectrometry. DNA was extracted from 44 samples: 18S rRNA gene amplicons were prepared and sequenced. Metabolomics, mycobiome and integrated analyses were performed. RESULTS Several VOCs were more abundant and short chain fatty acids were less abundant in PD. Hanseniaspora, Kazachstania, uncultured Tremellaceae and Penicillium genera were more abundant, and Saccharomyces less abundant in PD (FDR<0.0007). Torulaspora was associated with PD and two VOCs. CONCLUSION PD patients had a distinct metabolome and mycobiome suggesting that fungal dysbiosis may contribute to PD pathogenesis.
Collapse
Affiliation(s)
- Eduardo De Pablo-Fernandez
- Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, London, United Kingdom.
| | - Gerum Gashaw Gebeyehu
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, The Henry Wellcome Laboratory, Nuffield Building, Crown Street, L69 3GE, Liverpool, United Kingdom.
| | - Luke Flain
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, The Henry Wellcome Laboratory, Nuffield Building, Crown Street, L69 3GE, Liverpool, United Kingdom
| | - Rachael Slater
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, The Henry Wellcome Laboratory, Nuffield Building, Crown Street, L69 3GE, Liverpool, United Kingdom
| | - Alessandra Frau
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, The Henry Wellcome Laboratory, Nuffield Building, Crown Street, L69 3GE, Liverpool, United Kingdom
| | - Umer Zeeshan Ijaz
- James Watt School of Engineering, University of Glasgow, Rankine Building, Oakfield Avenue, G12 8QQ, Glasgow, United Kingdom
| | - Thomas Warner
- Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, London, United Kingdom
| | - Chris Probert
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, The Henry Wellcome Laboratory, Nuffield Building, Crown Street, L69 3GE, Liverpool, United Kingdom
| |
Collapse
|
34
|
Walker AC, Bhargava R, Vaziriyan-Sani AS, Brust AS, Czyz DM. Quantification of Bacterial Loads in Caenorhabditis elegans. Bio Protoc 2022; 12:e4291. [PMID: 35127981 PMCID: PMC8799673 DOI: 10.21769/bioprotoc.4291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/01/2021] [Accepted: 11/08/2021] [Indexed: 12/24/2024] Open
Abstract
Caenorhabditis elegans is a ubiquitous free-living nematode that feeds on bacteria. The organism was introduced into a laboratory setting in the 1970s and has since gained popularity as a model to study host-bacteria interactions. One advantage of using C. elegans is that its intestine can be colonized by the bacteria on which it feeds. Quantifying the bacterial load within C. elegans is an important and easily obtainable metric when investigating host-bacteria interactions. Although quantification of bacteria harbored in C. elegans via whole-worm lysis is not a novel assay, there is great variation between existing methods. To lyse C. elegans, many protocols rely on the use of a hand-held homogenizer, which could introduce systematic error and subsequent variation between researchers performing the same experiment. Here, we describe a method of lysing the intestines of C. elegans to quantify the bacterial load within the intestine. Our method has been optimized for removing exogenous bacteria while maintaining worm paralysis, to ensure no bactericidal agents are swallowed, which could kill bacteria within the intestine and affect results. We utilize and compare the efficiency of two different homogenization tools: a battery-powered hand-held homogenizer, and a benchtop electric homogenizer, where the latter minimizes variability. Thus, our protocol has been optimized to reduce systematic error and decrease the potential for variability among experimenters. Graphic abstract: Simplified overview of the procedure used to quantify the bacterial load within C. elegans. The two different methods are herein described for worm lysis: "Option 1" is a hand-held homogenizer, and "Option 2" is a benchtop homogenizer.
Collapse
Affiliation(s)
- Alyssa C. Walker
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Rohan Bhargava
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | | | - Amanda S. Brust
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| | - Daniel M. Czyz
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
35
|
Moreta MPG, Burgos-Alonso N, Torrecilla M, Marco-Contelles J, Bruzos-Cidón C. Efficacy of Acetylcholinesterase Inhibitors on Cognitive Function in Alzheimer's Disease. Review of Reviews. Biomedicines 2021; 9:biomedicines9111689. [PMID: 34829917 PMCID: PMC8615650 DOI: 10.3390/biomedicines9111689] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/03/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia over the age of 65. It is estimated that 115.4 million people will be affected by AD by 2050. Acetylcholinesterase inhibitors (AChEI) are the only available and approved treatment for AD. The aim of the present study was to analyse the evidence on the efficacy of the AChEI in the treatment of cognitive symptoms of Alzheimer’s disease. For that purpose, a review of review of the systematic reviews (SRs) on this topic was carried out by Web of Science, PubMed, and The Cochrane Library, among others, were searched until 24 September 2021. Thirteen of the 1773 articles evaluated the efficacy of AChEI on cognitive function and/or general condition and/or behavioural disturbances of patients with mild to moderate AD. Methodological quality and risk of bias were rated using the ROBIS scale. The quality of the identified studies was high for nine of them, unclear for two, and finally only in two of the 13 studies did we detect low quality. Overall, AChEI showed very low efficacy in improving cognition in patients with mild to moderate AD. Better results were obtained in improving global state, with donepezil being the most effective treatment. No improvements in behavioural disturbances were found. Few high-quality reviews provide clear evidence of the effects of AChEI on cognition, global change, behaviour, and mortality. The data suggest that AChEI stabilize or slow cognitive deterioration, improving global status. In addition, data indicate that the use of AChEI decreases mortality in patients with mild to moderate AD. However, there is no evidence that they improve patient behaviour. Donepezil is the best therapeutic alternative at a dose of 10 mg/day.
Collapse
Affiliation(s)
- Marta Pérez-Gómez Moreta
- Preventive Medicine and Public Health Department, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
- Correspondence: or ; Tel.: +34-607-304-793
| | - Natalia Burgos-Alonso
- Preventive Medicine and Public Health Department, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
| | - María Torrecilla
- Pharmacology Department, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry, Institute of Organic Chemistry (CSIC), Juan de la Cierva, 3, 28006 Madrid, Spain;
| | - Cristina Bruzos-Cidón
- Nursing I Department, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
| |
Collapse
|
36
|
Tinkov AA, Martins AC, Avila DS, Gritsenko VA, Skalny AV, Santamaria A, Lee E, Bowman AB, Aschner M. Gut Microbiota as a Potential Player in Mn-Induced Neurotoxicity. Biomolecules 2021; 11:1292. [PMID: 34572505 PMCID: PMC8469589 DOI: 10.3390/biom11091292] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [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: 08/16/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022] Open
Abstract
Manganese (Mn) is an essential metal, which at high exposures causes neurotoxic effects and neurodegeneration. The neurotoxic effects of Mn are mediated by neuroinflammation, oxidative and endoplasmic reticulum stress, mitochondrial dysfunction, and other mechanisms. Recent findings have demonstrated the potential impact of Mn overexposure on gut microbiota dysbiosis, which is known to contribute to neurodegeneration via secretion of neuroactive and proinflammatory metabolites. Therefore, in this review, we discuss the existing data on the impact of Mn exposure on gut microbiota biodiversity, bacterial metabolite production, and gut wall permeability regulating systemic levels. Recent data have demonstrated that Mn exposure may affect gut microbiota biodiversity by altering the abundance of Shiegella, Ruminococcus, Dorea, Fusicatenibacter, Roseburia, Parabacteroides, Bacteroidetes, Firmicutes, Ruminococcaceae, Streptococcaceae, and other bacterial phyla. A Mn-induced increase in Bacteroidetes abundance and a reduced Firmicutes/Bacteroidetes ratio may increase lipopolysaccharide levels. Moreover, in addition to increased systemic lipopolysaccharide (LPS) levels, Mn is capable of potentiating LPS neurotoxicity. Due to the high metabolic activity of intestinal microflora, Mn-induced perturbations in gut microbiota result in a significant alteration in the gut metabolome that has the potential to at least partially mediate the biological effects of Mn overexposure. At the same time, a recent study demonstrated that healthy microbiome transplantation alleviates Mn-induced neurotoxicity, which is indicative of the significant role of gut microflora in the cascade of Mn-mediated neurotoxicity. High doses of Mn may cause enterocyte toxicity and affect gut wall integrity through disruption of tight junctions. The resulting increase in gut wall permeability further promotes increased translocation of LPS and neuroactive bacterial metabolites to the systemic blood flow, ultimately gaining access to the brain and leading to neuroinflammation and neurotransmitter imbalance. Therefore, the existing data lead us to hypothesize that gut microbiota should be considered as a potential target of Mn toxicity, although more detailed studies are required to characterize the interplay between Mn exposure and the gut, as well as its role in the pathogenesis of neurodegeneration and other diseases.
Collapse
Affiliation(s)
- Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia;
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Daiana Silva Avila
- Laboratory of Biochemistry and Toxicoology in Caenorhabditis elegans, Universidade Federal do Pampa, Campus Uruguaiana, BR-472 Km 592, Uruguaiana 97500-970, RS, Brazil;
| | - Victor A. Gritsenko
- Institute of Cellular and Intracellular Symbiosis, Ural Branch of the Russian Academy of Sciences, Pionerskaya st 11, 460000 Orenburg, Russia;
| | - Anatoly V. Skalny
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
- Laboratory of Medical Elementology, KG Razumovsky Moscow State University of Technologies and Management, 109004 Moscow, Russia
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico;
| | - Eunsook Lee
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Michael Aschner
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia;
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| |
Collapse
|