1
|
Rivera DE, Poirier K, Moore S, Nicolle O, Morgan E, Longares JF, Singh A, Michaux G, Félix MA, Luallen RJ. Dynamics of gut colonization by commensal and pathogenic bacteria that attach to the intestinal epithelium. NPJ Biofilms Microbiomes 2025; 11:70. [PMID: 40319018 PMCID: PMC12049552 DOI: 10.1038/s41522-025-00696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/04/2025] [Indexed: 05/07/2025] Open
Abstract
Bacterial adherence to the intestinal epithelium plays a role in niche establishment in the gut lumen. Through sampling natural populations of Caenorhabditis, we discovered several bacterial species that adhere to the intestinal epithelium via polar, intimate association, best described as attachment. These bacteria had varying effects on host fitness and physiology, with one species having negative effects, and the others exhibiting neutral effects. These bacteria can actively divide in the gut lumen, either replicating throughout the gut simultaneously or anteroposteriorly. In competition assays, animals pre-colonized with an attaching commensal bacteria reduced colonization by the pathogenic bacteria, but this effect was not seen when animals were colonized by both species simultaneously. Regardless of the colonization paradigm, populations exposed to both bacteria showed a near-identical mitigation of the pathogenic effects. Altogether, these strains illustrate the capacity of microbiome bacteria to attach, replicate, and establish a niche across the entire intestinal lumen.
Collapse
Affiliation(s)
- Dalaena E Rivera
- Department of Biology, San Diego State University, San Diego, USA
| | - Kayla Poirier
- Department of Biology, San Diego State University, San Diego, USA
| | - Samuel Moore
- Department of Biology, San Diego State University, San Diego, USA
| | - Ophélie Nicolle
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) -UMR 6290, F-35000, Rennes, France
| | - Emily Morgan
- Department of Biology, San Diego State University, San Diego, USA
| | | | - Anupama Singh
- Department of Biology, San Diego State University, San Diego, USA
| | - Grégoire Michaux
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) -UMR 6290, F-35000, Rennes, France
| | - Marie-Anne Félix
- Institut de Biologie de l'École Normale Supérieure, Centre National de la Recherche Scientifique, INSERM, École Normale Supérieure, Paris Sciences et Lettres, Paris, France.
| | - Robert J Luallen
- Department of Biology, San Diego State University, San Diego, USA.
| |
Collapse
|
2
|
Wu X, Liu H, Guo YR. Insights into Virus-Host Interactions: Lessons from Caenorhabditis elegans-Orsay Virus Model. Curr Med Sci 2025; 45:169-184. [PMID: 40029496 DOI: 10.1007/s11596-025-00004-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/18/2024] [Accepted: 10/20/2024] [Indexed: 03/05/2025]
Abstract
The study of virus-host interactions has been significantly advanced using model organisms, with nematodes being a prominent example. Caenorhabditis elegans (C. elegans) nematodes have provided valuable insights into the mechanisms of viral infections, host defense strategies, and the development of antiviral therapies. With the discovery of natural viral pathogens of nematodes, Orsay virus, Le Blanc virus, Santeuil virus, and Mělník virus, the exploration of the virus-host interaction model based on nematodes has entered a new era. The virus-host interaction network consists of viruses, hosts, and the antagonistic effects of viruses on host immunity. The nematode virus-host interaction model is a concrete manifestation used to study the complex relationships among these three elements. Previous studies have indicated that during the entire process of nematode infection by viruses, antiviral RNA interference (RNAi) plays a crucial role. Additionally, the host's innate immune responses, such as the antiviral-specific intracellular pathogen response (IPR) and certain signaling pathways homologous to those in humans, are particularly important in the natural immune and antiviral processes of nematodes. These processes are regulated by multiple genes in the host. The reverse genetics system for Orsay virus has been successfully developed to study viral gene function and virus-host interactions. Nematodes serve as simple host models for understanding RNA virus replication, related cellular components, and virus-host interaction mechanisms. These findings will likely contribute to the development of antiviral treatment strategies based on novel targets.
Collapse
Affiliation(s)
- Xun Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Heng Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yusong R Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Zhang XP, Ma X, Liu JL, Liu AL. Exploring the potential use of Caenorhabditis elegans as an animal model for evaluating chemical-induced intestinal dysfunction. Toxicol Appl Pharmacol 2024; 493:117140. [PMID: 39500396 DOI: 10.1016/j.taap.2024.117140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024]
Abstract
Evaluating intestinal toxicity is crucial for identifying and preventing the harmful effects of environmental chemicals. Owing to the limitations of existing models in evaluating intestinal toxicity, the development of alternative models is urgently needed. This study explored the potential use of the nematode Caenorhabditis elegans as a model animal for assessing chemical-induced intestinal dysfunction. Changes in intestinal permeability and nutrient absorption in C. elegans individuals exposed to four intestine-disrupting chemicals (sodium dodecyl sulfate (SDS), dextran sulfate sodium (DSS), lipopolysaccharide (LPS) and ethanol) were examined using dye stain assays, an enzymatic photometric assay, and fluorescent probe uptake assays. Additionally, epigallocatechin-3-gallate (EGCG), an intestine-protecting phytochemical, was chosen to prevent ethanol-induced intestinal damage. The results indicated that SDS, DSS, LPS, and ethanol compromised the intestinal barrier in C. elegans. SDS had no effect on glucose absorption, but LPS, DSS, and ethanol inhibited or tended to inhibit glucose absorption. SDS, DSS, LPS, and ethanol reduced fatty acid absorption. LPS increased peptide absorption at a low dose but decreased it at a high dose; SDS, DSS, and ethanol attenuated peptide absorption. EGCG protected against the disruption of the intestinal barrier that was induced by ethanol treatment. These results suggest that C. elegans is a suitable surrogate model animal for evaluating chemical-induced intestinal dysfunction. These findings also provide new insights into the effects of SDS, DSS, LPS, and ethanol on intestinal function and highlight the potential of EGCG as a natural dietary intervention to protect individuals who use excess alcohol from intestinal injury.
Collapse
Affiliation(s)
- Xiao-Pan Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xuan Ma
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun-Ling Liu
- Wuhan Center for Disease Control and Prevention, Wuhan 430022, China
| | - Ai-Lin Liu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
4
|
Kim S, Lee YR, Yang H, Park CH, Yun CS, Jang BC, Hong Y, Park DS. Potential probiotic Lactiplantibacillus plantarum DS1800 extends lifespan and enhances stress resistance in Caenorhabditis elegans model. Front Physiol 2024; 15:1476096. [PMID: 39502409 PMCID: PMC11534597 DOI: 10.3389/fphys.2024.1476096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Probiotics are live microorganisms that provide health benefits when administered in appropriate amounts by improving or restoring the balance of intestinal microbiota. Various functional probiotic products have been developed due to the growing interest in the health-promoting and anti-aging effects of enhancing the gut microbiome. Lactiplantibacillus plantarum species are known for their potential to extend lifespan. However, this activity is strain or isolation source specific, necessitating the identification of individual strain functionalities. This study used the C. elegans model to screen probiotics for life-extension effects and analyze their functions. The 43 lactic-acid bacteria strains isolated from fermented foods, breast milk, and human feces were subjected to longevity assays, and L. plantarum DS1800 was selected to demonstrate the most effective lifespan extension. The average lifespan of Caenorhabditis elegans fed DS1800 increased by 17.36% compared with those fed Escherichia coli OP50. Further analysis of the expression of key genes related to longevity revealed the high expression of the skinhead-1 (skn-1), antibacterial, and heat stress resistance genes via the p38 MAPK pathway. These expression patterns suggest that DS1800 extends the lifespan of C. elegans by enhancing its stress resistance and protecting it against pathogens. Additionally, DS1800 exhibited excellent intestinal adhesion, with 7.56% adhesion to HT-29 cells. Therefore, L. plantarum DS1800 is effective in extending the lifespan of C. elegans and can be used as a functional probiotic.
Collapse
Affiliation(s)
- Seunghyun Kim
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yu-Ri Lee
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Haneol Yang
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Chan-Hyeok Park
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Chan-Seok Yun
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Byung-Chun Jang
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yeongjin Hong
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Doo-Sang Park
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
5
|
Sousa M, Magalhães R, Ferreira V, Teixeira P. Current methodologies available to evaluate the virulence potential among Listeria monocytogenes clonal complexes. Front Microbiol 2024; 15:1425437. [PMID: 39493856 PMCID: PMC11528214 DOI: 10.3389/fmicb.2024.1425437] [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: 04/29/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Listeria monocytogenes is a foodborne pathogen that causes listeriosis in humans, the severity of which depends on multiple factors, including intrinsic characteristics of the affected individuals and the pathogen itself. Additionally, emerging evidence suggests that epigenetic modifications may also modulate host susceptibility to infection. Therefore, different clinical outcomes can be expected, ranging from self-limiting gastroenteritis to severe central nervous system and maternal-neonatal infections, and bacteremia. Furthermore, L. monocytogenes is a genetically and phenotypically diverse species, resulting in a large variation in virulence potential between strains. Multilocus sequence typing (MLST) has been widely used to categorize the clonal structure of bacterial species and to define clonal complexes (CCs) of genetically related isolates. The combination of MLST and epidemiological data allows to distinguish hypervirulent CCs, which are notably more prevalent in clinical cases and typically associated with severe forms of the disease. Conversely, other CCs, termed hypovirulent, are predominantly isolated from food and food processing environments and are associated with the occurrence of listeriosis in immunosuppressed individuals. Reports of genetic traits associated with this diversity have been described. The Food and Agriculture Organization (FAO) is encouraging the search for virulence biomarkers to rapidly identify the main strains of concern to reduce food waste and economical losses. The aim of this review is to comprehensively collect, describe and discuss the methodologies used to discriminate the virulence potential of L. monocytogenes CCs. From the exploration of in vitro and in vivo models to the study of expression of virulence genes, each approach is critically explored to better understand its applicability and efficiency in distinguishing the virulence potential of the pathogen.
Collapse
Affiliation(s)
| | | | | | - Paula Teixeira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, Portugal
| |
Collapse
|
6
|
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
|
7
|
Kywe C, Lundquist EA, Ackley BD, Lansdon P. The MAB-5/Hox family transcription factor is important for Caenorhabditis elegans innate immune response to Staphylococcus epidermidis infection. G3 (BETHESDA, MD.) 2024; 14:jkae054. [PMID: 38478633 PMCID: PMC11075571 DOI: 10.1093/g3journal/jkae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/17/2024] [Accepted: 03/03/2024] [Indexed: 04/12/2024]
Abstract
Innate immunity functions as a rapid defense against broad classes of pathogenic agents. While the mechanisms of innate immunity in response to antigen exposure are well-studied, how pathogen exposure activates the innate immune responses and the role of genetic variation in immune activity is currently being investigated. Previously, we showed significant survival differences between the N2 and the CB4856 Caenorhabditis elegans isolates in response to Staphylococcus epidermidis infection. One of those differences was expression of the mab-5 Hox family transcription factor, which was induced in N2, but not CB4856, after infection. In this study, we use survival assays and RNA-sequencing to better understand the role of mab-5 in response to S. epidermidis. We found that mab-5 loss-of-function (LOF) mutants were more susceptible to S. epidermidis infection than N2 or mab-5 gain-of-function (GOF) mutants, but not as susceptible as CB4856 animals. We then conducted transcriptome analysis of infected worms and found considerable differences in gene expression profiles when comparing animals with mab-5 LOF to either N2 or mab-5 GOF. N2 and mab-5 GOF animals showed a significant enrichment in expression of immune genes and C-type lectins, whereas mab-5 LOF mutants did not. Overall, gene expression profiling in mab-5 mutants provided insight into MAB-5 regulation of the transcriptomic response of C. elegans to pathogenic bacteria and helps us to understand mechanisms of innate immune activation and the role that transcriptional regulation plays in organismal health.
Collapse
Affiliation(s)
- Christopher Kywe
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Erik A Lundquist
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Brian D Ackley
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Patrick Lansdon
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
8
|
González R, Félix MA. Caenorhabditis elegans immune responses to microsporidia and viruses. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 154:105148. [PMID: 38325500 DOI: 10.1016/j.dci.2024.105148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
The model organism Caenorhabditis elegans is susceptible to infection by obligate intracellular pathogens, specifically microsporidia and viruses. These intracellular pathogens infect intestinal cells, or, for some microsporidia, epidermal cells. Strikingly, intestinal cell infections by viruses or microsporidia trigger a common transcriptional response, activated in part by the ZIP-1 transcription factor. Among the strongest activated genes in this response are ubiquitin-pathway members and members of the pals family, an intriguing gene family with cross-regulations of different members of genomic clusters. Some of the induced genes participate in host defense against the pathogens, for example through ubiquitin-mediated inhibition. Other mechanisms defend the host specifically against viral infections, including antiviral RNA interference and uridylation. These various immune responses are altered by environmental factors and by intraspecific genetic variation of the host. These pathogens were first isolated 15 years ago and much remains to be discovered using C. elegans genetics; also, other intracellular pathogens of C. elegans may yet to be discovered.
Collapse
Affiliation(s)
- Rubén González
- Institut de Biologie de l'École Normale Supérieure, CNRS, INSERM, 75005, Paris, France.
| | - Marie-Anne Félix
- Institut de Biologie de l'École Normale Supérieure, CNRS, INSERM, 75005, Paris, France
| |
Collapse
|
9
|
Otarigho B, Butts AF, Aballay A. Neuronal NPR-15 modulates molecular and behavioral immune responses via the amphid sensory neuron-intestinal axis in C. elegans. eLife 2024; 12:RP90051. [PMID: 38446031 PMCID: PMC10942643 DOI: 10.7554/elife.90051] [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: 03/07/2024] Open
Abstract
The survival of hosts during infections relies on their ability to mount effective molecular and behavioral immune responses. Despite extensive research on these defense strategies in various species, including the model organism Caenorhabditis elegans, the neural mechanisms underlying their interaction remain poorly understood. Previous studies have highlighted the role of neural G-protein-coupled receptors (GPCRs) in regulating both immunity and pathogen avoidance, which is particularly dependent on aerotaxis. To address this knowledge gap, we conducted a screen of mutants in neuropeptide receptor family genes. We found that loss-of-function mutations in npr-15 activated immunity while suppressing pathogen avoidance behavior. Through further analysis, NPR-15 was found to regulate immunity by modulating the activity of key transcription factors, namely GATA/ELT-2 and TFEB/HLH-30. Surprisingly, the lack of pathogen avoidance of npr-15 mutant animals was not influenced by oxygen levels. Moreover, our studies revealed that the amphid sensory neuron ASJ is involved in mediating the immune and behavioral responses orchestrated by NPR-15. Additionally, NPR-15 was found to regulate avoidance behavior via the TRPM (transient receptor potential melastatin) gene, GON-2, which may sense the intestinal distension caused by bacterial colonization to elicit pathogen avoidance. Our study contributes to a broader understanding of host defense strategies and mechanisms underlining the interaction between molecular and behavioral immune responses.
Collapse
Affiliation(s)
- Benson Otarigho
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Anna Frances Butts
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Alejandro Aballay
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealthHoustonUnited States
| |
Collapse
|
10
|
Poirier KM, Luallen RJ, Rivera DE. RNA fluorescence in situ hybridization (FISH) as a method to visualize bacterial colonization in the C. elegans gut. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001044. [PMID: 38481555 PMCID: PMC10935869 DOI: 10.17912/micropub.biology.001044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/05/2024] [Accepted: 02/25/2024] [Indexed: 03/17/2024]
Abstract
Caenorhabditis elegans is an excellent model to study host-microbe interactions as it is easy to visualize bacterial presence in their intestine. However, previous studies have shown that utilizing transgenic, fluorescent protein-expressing bacteria is not a reliable method to distinguish living bacteria from dead bacteria in the lumen of C. elegans . In this study, we compared methods for visualizing bacterial presence within the C. elegans intestine and found that RNA f luorescent i n s itu h ybridization (RNA FISH) could distinguish the difference between intact and dead bacteria. Thus, we propose RNA FISH as the preferred method to visualize live bacterial presence in the intestines of C. elegans prior to fixation.
Collapse
Affiliation(s)
- Kayla M Poirier
- Department of Biology, San Diego State University, San Diego, California, United States
| | - Robert J Luallen
- Department of Biology, San Diego State University, San Diego, California, United States
| | - Dalaena E Rivera
- Department of Biology, San Diego State University, San Diego, California, United States
| |
Collapse
|
11
|
Otarigho B, Butts AF, Aballay A. Neuronal NPR-15 modulates molecular and behavioral immune responses via the amphid sensory neuron-intestinal axis in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.27.550570. [PMID: 37546751 PMCID: PMC10402133 DOI: 10.1101/2023.07.27.550570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The survival of hosts during infections relies on their ability to mount effective molecular and behavioral immune responses. Despite extensive research on these defense strategies in various species, including the model organism Caenorhabditis elegans, the neural mechanisms underlying their interaction remain poorly understood. Previous studies have highlighted the role of neural G protein-coupled receptors (GPCRs) in regulating both immunity and pathogen avoidance, which is particularly dependent on aerotaxis. To address this knowledge gap, we conducted a screen of mutants in neuropeptide receptor family genes. We found that loss-of-function mutations in npr-15 activated immunity while suppressing pathogen avoidance behavior. Through further analysis, NPR-15 was found to regulate immunity by modulating the activity of key transcription factors, namely GATA/ELT-2 and TFEB/HLH-30. Surprisingly, the lack of pathogen avoidance of npr-15 mutant animals was not influenced by oxygen levels. Moreover, our studies revealed that the amphid sensory neuron ASJ is involved in mediating the immune and behavioral responses orchestrated by NPR-15. Additionally, NPR-15 was found to regulate avoidance behavior via the TRPM gene, GON-2, which may sense the intestinal distension caused by bacterial colonization to elicit pathogen avoidance. Our study contributes to a broader understanding of host defense strategies and mechanisms underlining the interaction between molecular and behavioral immune responses.
Collapse
Affiliation(s)
- Benson Otarigho
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anna Frances Butts
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alejandro Aballay
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth Houston, TX
| |
Collapse
|
12
|
Lažetić V, Batachari LE, Russell AB, Troemel ER. Similarities in the induction of the intracellular pathogen response in Caenorhabditis elegans and the type I interferon response in mammals. Bioessays 2023; 45:e2300097. [PMID: 37667453 PMCID: PMC10694843 DOI: 10.1002/bies.202300097] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023]
Abstract
Although the type-I interferon (IFN-I) response is considered vertebrate-specific, recent findings about the Intracellular Pathogen Response (IPR) in nematode Caenorhabditis elegans indicate that there are similarities between these two transcriptional immunological programs. The IPR is induced during infection with natural intracellular fungal and viral pathogens of the intestine and promotes resistance against these pathogens. Similarly, the IFN-I response is induced by viruses and other intracellular pathogens and promotes resistance against infection. Whether the IPR and the IFN-I response evolved in a divergent or convergent manner is an unanswered and exciting question, which could be addressed by further studies of immunity against intracellular pathogens in C. elegans and other simple host organisms. Here we highlight similar roles played by RIG-I-like receptors, purine metabolism enzymes, proteotoxic stressors, and transcription factors to induce the IPR and IFN-I response, as well as the similar consequences of these defense programs on organismal development.
Collapse
Affiliation(s)
- Vladimir Lažetić
- School of Biological SciencesUniversity of California, San DiegoLa JollaCaliforniaUSA
- Department of Biological SciencesThe George Washington UniversityWashingtonDCUSA
| | - Lakshmi E. Batachari
- School of Biological SciencesUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Alistair B. Russell
- School of Biological SciencesUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Emily R. Troemel
- School of Biological SciencesUniversity of California, San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
13
|
Bai S, Song J, Pu H, Yu Y, Song W, Chen Z, Wang M, Campbell-Valois FX, Wong WL, Cai Q, Wan M, Zhang C, Bai Y, Feng X. Chemical Biology Approach to Reveal the Importance of Precise Subcellular Targeting for Intracellular Staphylococcus aureus Eradication. J Am Chem Soc 2023; 145:23372-23384. [PMID: 37838963 DOI: 10.1021/jacs.3c09587] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Intracellular bacterial pathogens, such as Staphylococcus aureus, that may hide in intracellular vacuoles represent the most significant manifestation of bacterial persistence. They are critically associated with chronic infections and antibiotic resistance, as conventional antibiotics are ineffective against such intracellular persisters due to permeability issues and mechanistic reasons. Direct subcellular targeting of S. aureus vacuoles suggests an explicit opportunity for the eradication of these persisters, but a comprehensive understanding of the chemical biology nature and significance of precise S. aureus vacuole targeting remains limited. Here, we report an oligoguanidine-based peptidomimetic that effectively targets and eradicates intracellular S. aureus persisters in the phagolysosome lumen, and this oligomer was utilized to reveal the mechanistic insights linking precise targeting to intracellular antimicrobial efficacy. The oligomer has high cellular uptake via a receptor-mediated endocytosis pathway and colocalizes with S. aureus persisters in phagolysosomes as a result of endosome-lysosome interconversion and lysosome-phagosome fusion. Moreover, the observation of a bacterium's altered susceptibility to the oligomer following a modification in its intracellular localization offers direct evidence of the critical importance of precise intracellular targeting. In addition, eradication of intracellular S. aureus persisters was achieved by the oligomer's membrane/DNA dual-targeting mechanism of action; therefore, its effectiveness is not hampered by the hibernation state of the persisters. Such precise subcellular targeting of S. aureus vacuoles also increases the agent's biocompatibility by minimizing its interaction with other organelles, endowing excellent in vivo bacterial targeting and therapeutic efficacy in animal models.
Collapse
Affiliation(s)
- Silei Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Junfeng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Huangsheng Pu
- College of Advanced Interdisciplinary Studies & Hunan Provincial Key Laboratory of Novel NanoOptoelectronic Information Materials and Devices, National University of Defense Technology, Changsha, Hunan 410073, China
- Nanhu Laser Laboratory, National University of Defense Technology, Changsha 410073, China
| | - Yue Yu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Wenwen Song
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Zhiyong Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Min Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | | | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, Hong Kong SAR, China
| | - Qingyun Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Muyang Wan
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Chunhui Zhang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yugang Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Xinxin Feng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
14
|
Das B, Madhubala D, Mahanta S, Patra A, Puzari U, Khan MR, Mukherjee AK. A Novel Therapeutic Formulation for the Improved Treatment of Indian Red Scorpion ( Mesobuthus tamulus) Venom-Induced Toxicity-Tested in Caenorhabditis elegans and Rodent Models. Toxins (Basel) 2023; 15:504. [PMID: 37624261 PMCID: PMC10467153 DOI: 10.3390/toxins15080504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
Indian Red Scorpion (Mesobuthus tamulus) stings are a neglected public health problem in tropical and sub-tropical countries, including India. The drawbacks of conventional therapies using commercial anti-scorpion antivenom (ASA) and α1-adrenoreceptor antagonists (AAA) have prompted us to search for an adequate formulation to improve treatment against M. tamulus stings. Novel therapeutic drug formulations (TDF) of low doses of commercial ASA, AAA, and ascorbic acid have remarkably improved in neutralising the in vivo toxic effects of M. tamulus venom (MTV) tested in Caenorhabditis elegans and Wistar strain albino rats in vivo models. The neutralisation of MTV-induced production of free radicals, alteration of the mitochondrial transmembrane potential, and upregulated expression of genes involved in apoptosis, detoxification, and stress response in C. elegans by TDF surpassed the same effect shown by individual components of the TDF. Further, TDF efficiently neutralized the MTV-induced increase in blood glucose level within 30 to 60 min post-treatment, organ tissue damage, necrosis, and pulmonary oedema in Wistar rats, indicating its clinical application for effecting treating M. tamulus envenomation. This study demonstrates for the first time that C. elegans can be a model organism for screening the neutralization potency of the drug molecules against a neurotoxic scorpion venom.
Collapse
Affiliation(s)
- Bhabana Das
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; (B.D.); (D.M.); (U.P.)
| | - Dev Madhubala
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; (B.D.); (D.M.); (U.P.)
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Garchuk, Paschim Boragaon, Guwahati 781035, Assam, India; (A.P.); (M.R.K.)
| | - Saurov Mahanta
- National Institute of Electronics and Information Technology (NIELIT), Guwahati 781008, Assam, India;
| | - Aparup Patra
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Garchuk, Paschim Boragaon, Guwahati 781035, Assam, India; (A.P.); (M.R.K.)
| | - Upasana Puzari
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; (B.D.); (D.M.); (U.P.)
| | - Mojibur R. Khan
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Garchuk, Paschim Boragaon, Guwahati 781035, Assam, India; (A.P.); (M.R.K.)
| | - Ashis K. Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; (B.D.); (D.M.); (U.P.)
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Garchuk, Paschim Boragaon, Guwahati 781035, Assam, India; (A.P.); (M.R.K.)
| |
Collapse
|
15
|
Lažetić V, Blanchard MJ, Bui T, Troemel ER. Multiple pals gene modules control a balance between immunity and development in Caenorhabditis elegans. PLoS Pathog 2023; 19:e1011120. [PMID: 37463170 PMCID: PMC10353827 DOI: 10.1371/journal.ppat.1011120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
The immune system continually battles against pathogen-induced pressures, which often leads to the evolutionary expansion of immune gene families in a species-specific manner. For example, the pals gene family expanded to 39 members in the Caenorhabditis elegans genome, in comparison to a single mammalian pals ortholog. Our previous studies have revealed that two members of this family, pals-22 and pals-25, act as antagonistic paralogs to control the Intracellular Pathogen Response (IPR). The IPR is a protective transcriptional response, which is activated upon infection by two molecularly distinct natural intracellular pathogens of C. elegans-the Orsay virus and the fungus Nematocida parisii from the microsporidia phylum. In this study, we identify a previously uncharacterized member of the pals family, pals-17, as a newly described negative regulator of the IPR. pals-17 mutants show constitutive upregulation of IPR gene expression, increased immunity against intracellular pathogens, as well as impaired development and reproduction. We also find that two other previously uncharacterized pals genes, pals-20 and pals-16, are positive regulators of the IPR, acting downstream of pals-17. These positive regulators reverse the effects caused by the loss of pals-17 on IPR gene expression, immunity, and development. We show that the negative IPR regulator protein PALS-17 and the positive IPR regulator protein PALS-20 colocalize inside and at the apical side of intestinal epithelial cells, which are the sites of infection for IPR-inducing pathogens. In summary, our study demonstrates that several pals genes from the expanded pals gene family act as ON/OFF switch modules to regulate a balance between organismal development and immunity against natural intracellular pathogens in C. elegans.
Collapse
Affiliation(s)
- Vladimir Lažetić
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Michael J. Blanchard
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Theresa Bui
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Emily R. Troemel
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
16
|
Wang Y, Zhang L, Yuan X, Wang D. Treatment with paeoniflorin increases lifespan of Pseudomonas aeruginosa infected Caenorhabditis elegans by inhibiting bacterial accumulation in intestinal lumen and biofilm formation. Front Pharmacol 2023; 14:1114219. [PMID: 37050896 PMCID: PMC10083309 DOI: 10.3389/fphar.2023.1114219] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Paeoniflorin is one of the important components in Paeoniaceae plants. In this study, we used Caenorhabditis elegans as a model host and Pseudomonas aeruginosa as a bacterial pathogen to investigate the possible role of paeoniflorin treatment against P. aeruginosa infection in the host and the underlying mechanisms. Posttreatment with 1.25–10 mg/L paeoniflorin could significantly increase the lifespan of P. aeruginosa infected nematodes. After the infection, the P. aeruginosa colony-forming unit (CFU) and P. aeruginosa accumulation in intestinal lumen were also obviously reduced by 1.25–10 mg/L paeoniflorin treatment. The beneficial effects of paeoniflorin treatment in increasing lifespan in P. aeruginosa infected nematodes and in reducing P. aeruginosa accumulation in intestinal lumen could be inhibited by RNAi of pmk-1, egl-1, and bar-1. In addition, paeoniflorin treatment suppressed the inhibition in expressions of pmk-1, egl-1, and bar-1 caused by P. aeruginosa infection in nematodes, suggesting that paeoniflorin could increase lifespan of P. aeruginosa infected nematode by activating PMK-1, EGL-1, and BAR-1. Moreover, although treatment with 1.25–10 mg/L paeoniflorin did not show obvious anti-P. aeruginosa activity, the P. aeruginosa biofilm formation and expressions of related virulence genes (pelA, pelB, phzA, lasB, lasR, rhlA, and rhlC) were significantly inhibited by paeoniflorin treatment. Treatment with 1.25–10 mg/L paeoniflorin could further decrease the levels of related virulence factors of pyocyanin, elastase, and rhamnolipid. In addition, 2.5–10 mg/L paeoniflorin treatment could inhibit the swimming, swarming, and twitching motility of P. aeruginosa, and treatment with 2.5–10 mg/L paeoniflorin reduced the cyclic-di-GMP (c-di-GMP) level. Therefore, paeoniflorin treatment has the potential to extend lifespan of P. aeruginosa infected hosts by reducing bacterial accumulation in intestinal lumen and inhibiting bacterial biofilm formation.
Collapse
|
17
|
Lažetić V, Blanchard MJ, Bui T, Troemel ER. Multiple pals gene modules control a balance between immunity and development in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.15.524171. [PMID: 36711775 PMCID: PMC9882112 DOI: 10.1101/2023.01.15.524171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The immune system continually battles against pathogen-induced pressures, which often leads to the evolutionary expansion of immune gene families in a species-specific manner. For example, the pals gene family expanded to 39 members in the Caenorhabditis elegans genome, in comparison to a single mammalian pals ortholog. Our previous studies have revealed that two members of this family, pals-22 and pals-25 , act as antagonistic paralogs to control the Intracellular Pathogen Response (IPR). The IPR is a protective transcriptional response, which is activated upon infection by two molecularly distinct natural intracellular pathogens of C. elegans - the Orsay virus and the fungus Nematocida parisii from the microsporidia phylum. In this study, we identify a previously uncharacterized member of the pals family, pals-17 , as a newly described negative regulator of the IPR. pals-17 mutants show constitutive upregulation of IPR gene expression, increased immunity against intracellular pathogens, as well as impaired development and reproduction. We also find that two other previously uncharacterized pals genes, pals-20 and pals-16 , are positive regulators of the IPR, acting downstream of pals-17 . These positive regulators reverse the effects caused by the loss of pals-17 on IPR gene expression, immunity and development. We show that the negative IPR regulator protein PALS-17 and the positive IPR regulator protein PALS-20 colocalize inside intestinal epithelial cells, which are the sites of infection for IPR-inducing pathogens. In summary, our study demonstrates that several pals genes from the expanded pals gene family act as ON/OFF switch modules to regulate a balance between organismal development and immunity against natural intracellular pathogens in C. elegans . AUTHOR SUMMARY Immune responses to pathogens induce extensive rewiring of host physiology. In the short term, these changes are generally beneficial as they can promote resistance against infection. However, prolonged activation of immune responses can have serious negative consequences on host health, including impaired organismal development and fitness. Therefore, the balance between activating the immune system and promoting development must be precisely regulated. In this study, we used genetics to identify a gene in the roundworm Caenorhabditis elegans called pals-17 that acts as a repressor of the Intracellular Pathogen Response (IPR), a defense response against viral and microsporidian infections. We also found that pals-17 is required for the normal development of these animals. Furthermore, we identified two other pals genes, pals-20 and pals-16 , as suppressors of pals-17 mutant phenotypes. Finally, we found that PALS-17 and PALS-20 proteins colocalize inside intestinal cells, where viruses and microsporidia invade and replicate in the host. Taken together, our study demonstrates a balance between organismal development and immunity that is regulated by several genetic ON/OFF switch 'modules' in C. elegans .
Collapse
Affiliation(s)
- Vladimir Lažetić
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States
| | - Michael J. Blanchard
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States
| | - Theresa Bui
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States
| | - Emily R. Troemel
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States,Corresponding author
| |
Collapse
|
18
|
Yang C, Diarra MS, Attiq Rehman M, Li L, Yu H, Yin X, Aslam M, Carrillo CD, Yang C, Gong J. Virulence potential of antimicrobial-resistant extraintestinal pathogenic Escherichia coli from retail poultry meat in a Caenorhabditis elegans model. J Food Prot 2023; 86:100008. [PMID: 36916583 DOI: 10.1016/j.jfp.2022.11.001] [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: 03/09/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 12/23/2022]
Abstract
Healthy poultry can be a reservoir for extraintestinal pathogenic Escherichia coli (ExPEC), some of which could be multidrug resistant to antimicrobials. These ExPEC strains could contaminate the environment and/or food chain representing thus, food safety and human health risk. However, few studies have shown the virulence of poultry-source antimicrobial-resistant (AMR) ExPEC in humans. This study characterized AMR ExPEC and investigated the virulence potential of some of their isolates in a Caenorhabditis elegans infection model. A total of 46 E. coli isolates from poultry (chicken, n = 29; turkey, n = 12) retail meats and chicken feces (n = 4), or humans (n = 1) were sequenced and identified as ExPEC. Except eight, all remaining 38 ExPEC isolates were resistant to at least one antibiotic and carried corresponding antimicrobial resistance genes (ARGs). About 27 of the 46 ExPEC isolates were multidrug-resistant (≥3 antibiotic classes). Seven ExPEC isolates from chicken or turkey meats were of serotype O25:H4 and sequence type (ST) 131 which clustered with an isolate from a human urinary tract infection (UTI) case having the same serotype and ST. The C. elegans challenge model using eight of studied ExPEC isolates harboring various ARGs and virulence genes (VGs) showed that regardless of their ARG or VG numbers in tested poultry meat and feces, ExPEC significantly reduced the life span of the nematode (P < 0.05) similarly to a human UTI isolate. This study indicated the pathogenic potential of AMR ExPEC from retail poultry meat or feces, but more studies are warranted to establish their virulence in poultry and human. Furthermore, relationships between specific resistance profiles and/or VGs in these E. coli isolates for their pathogenicity deserve investigations.
Collapse
Affiliation(s)
- Chongwu Yang
- Guelph Research and Development Centre, Agriculture Agri-Food Canada (AAFC), Guelph, Ontario, Canada N1G 5C9; Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada R3T 2N2
| | - Moussa S Diarra
- Guelph Research and Development Centre, Agriculture Agri-Food Canada (AAFC), Guelph, Ontario, Canada N1G 5C9.
| | - Muhammad Attiq Rehman
- Guelph Research and Development Centre, Agriculture Agri-Food Canada (AAFC), Guelph, Ontario, Canada N1G 5C9
| | - Linyan Li
- Guelph Research and Development Centre, Agriculture Agri-Food Canada (AAFC), Guelph, Ontario, Canada N1G 5C9; State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hai Yu
- Guelph Research and Development Centre, Agriculture Agri-Food Canada (AAFC), Guelph, Ontario, Canada N1G 5C9
| | - Xianhua Yin
- Guelph Research and Development Centre, Agriculture Agri-Food Canada (AAFC), Guelph, Ontario, Canada N1G 5C9
| | - Mueen Aslam
- Lacombe Research Centre, AAFC, Lacombe, Alberta, Canada T4L1W1
| | - Catherine D Carrillo
- Canadian Food Inspection Agency (CFIA), Ottawa Laboratory (Carling), Ottawa, Ontario, Canada K1Y 4K7
| | - Chengbo Yang
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada R3T 2N2
| | - Joshua Gong
- Guelph Research and Development Centre, Agriculture Agri-Food Canada (AAFC), Guelph, Ontario, Canada N1G 5C9.
| |
Collapse
|
19
|
Structure of the reduced microsporidian proteasome bound by PI31-like peptides in dormant spores. Nat Commun 2022; 13:6962. [PMID: 36379934 PMCID: PMC9666519 DOI: 10.1038/s41467-022-34691-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 11/02/2022] [Indexed: 11/17/2022] Open
Abstract
Proteasomes play an essential role in the life cycle of intracellular pathogens with extracellular stages by ensuring proteostasis in environments with limited resources. In microsporidia, divergent parasites with extraordinarily streamlined genomes, the proteasome complexity and structure are unknown, which limits our understanding of how these unique pathogens adapt and compact essential eukaryotic complexes. We present cryo-electron microscopy structures of the microsporidian 20S and 26S proteasome isolated from dormant or germinated Vairimorpha necatrix spores. The discovery of PI31-like peptides, known to inhibit proteasome activity, bound simultaneously to all six active sites within the central cavity of the dormant spore proteasome, suggests reduced activity in the environmental stage. In contrast, the absence of the PI31-like peptides and the existence of 26S particles post-germination in the presence of ATP indicates that proteasomes are reactivated in nutrient-rich conditions. Structural and phylogenetic analyses reveal that microsporidian proteasomes have undergone extensive reductive evolution, lost at least two regulatory proteins, and compacted nearly every subunit. The highly derived structure of the microsporidian proteasome, and the minimized version of PI31 presented here, reinforce the feasibility of the development of specific inhibitors and provide insight into the unique evolution and biology of these medically and economically important pathogens.
Collapse
|
20
|
Zhang L, Wang Y, Cao C, Zhu Y, Huang W, Yang Y, Qiu H, Liu S, Wang D. Beneficial effect of Xuebijing against Pseudomonas aeruginosa infection in Caenorhabditis elegans. Front Pharmacol 2022; 13:949608. [PMID: 36120363 PMCID: PMC9470999 DOI: 10.3389/fphar.2022.949608] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/01/2022] [Indexed: 12/14/2022] Open
Abstract
In the clinical intensive care units (ICU), the traditional Chinese medicine (TCM) formulation of Xuebijing has been frequently used for treating sepsis. Nevertheless, the underlying pharmacological mechanisms of Xuebijing remain largely unclear. Caenorhabditis elegans is an important experimental host for bacterial infections. Using C. elegans as an animal model, we here examined the potential of Xuebijing treatment against bacterial infection and the underlying mechanisms. Xuebijing treatment could inhibit the reduction tendency of lifespan caused by Pseudomonas aeruginosa infection. For the cellular mechanisms of this antibacterial infection property, we found that Xuebijing treatment rescued C. elegans lifespan to be against P. aeruginosa infection by inhibiting Pseudomonas colonization in the intestinal lumen. Meanwhile, the increase in the expression of antimicrobial genes induced by Pseudomonas infection was also suppressed by Xuebijing treatment. Moreover, the beneficial effect of Xuebijing against Pseudomonas infection depended on insulin, p38 MAPK, Wnt, DBL-1/TGF-β, ELT-2, and programmed cell death (PCD)-related signals. Although Xuebijing did not show obvious antibacterial activity, Xuebijing (100%) treatment could inhibit the Pseudomonas biofilm formation and decrease the expression of virulence genes (lasA, lasB, rhlA, rhlC, phzA, phzM, phzH, and phzS) and quorum sensing (QS)-related genes (lasI, lasR, rhlI, rhlR, pqsA, and pqsR). Our results support the potential role of Xuebijing treatment against bacterial infection in hosts.
Collapse
Affiliation(s)
- Le Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
| | - Yuxing Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
| | - Chang Cao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
| | - Yike Zhu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
- Department of Critical Care Medicine, Zhongda Hospital, Nanjing, China
| | - Wei Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
- Department of Critical Care Medicine, Zhongda Hospital, Nanjing, China
| | - Yi Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
- Department of Critical Care Medicine, Zhongda Hospital, Nanjing, China
| | - Haibo Qiu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
- Department of Critical Care Medicine, Zhongda Hospital, Nanjing, China
| | - Songqiao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
- Department of Critical Care Medicine, Zhongda Hospital, Nanjing, China
- *Correspondence: Songqiao Liu, ; Dayong Wang,
| | - Dayong Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, China
- *Correspondence: Songqiao Liu, ; Dayong Wang,
| |
Collapse
|
21
|
Yao H, Xu A, Liu J, Wang F, Yao H, Chen J. Evaluation of in vivo antibacterial drug efficacy using Caenorhabditis elegans infected with carbapenem-resistant Klebsiella pneumoniae as a model host. Front Pharmacol 2022; 13:973551. [PMID: 36059996 PMCID: PMC9439659 DOI: 10.3389/fphar.2022.973551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022] Open
Abstract
Objective: This study was developed to assess the in vivo antimicrobial activity of specific drugs using a model system consisting of Caenorhabditis elegans (C. elegans) infected with Carbapenem-resistant Klebsiella pneumoniae (CRKP) in an effort to identify promising drugs for CRKP-infected patient treatment. Methods: A C. elegans-CRKP liquid assay platform was developed and used to conduct limited in vivo screening for antimicrobial agents with potential activity against CRKP. Time curves for 10 different concentrations of tested antimicrobial agents were tested in this model system at 0, 2, 6, 8, and 12 h after treatment. The protective effects of these different antimicrobial agents were compared at different time points. Furthermore, ten CRKP strains samples were isolated from clinical specimens to demonstrate the applicability of the nematode model method, and two typical clinical cases are presented. Results: CRKP bacteria were sufficient to induce C. elegans death in a dose- and time-dependent fashion, while effective antimicrobial agents improved the survival of these nematodes in a dose-dependent manner. Notably, PB and TGC exhibited robust antibacterial protection within 12 h even at low tested concentrations, and clear efficacy remained evident for high doses of CAZ at this same time point as mediators of improved nematode survival. The results of C. elegans model method were well consistent with that using the Kirby-Bauer method in 10 CRKP strains samples, and two typical clinical cases showed applicability, reliability and efficacy of C. elegans model method. Conclusion: Overall, nematode models in drug sensitivity testing have shown advantages in clinical settings. Our results highlight the value of C. elegans model systems as tools for the simultaneous screening of different agents for in vivo antibacterial efficacy and are deserved further study.
Collapse
Affiliation(s)
- Huijuan Yao
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ajing Xu
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingxian Liu
- Clinical Laboratory, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fang Wang
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huimin Yao
- School of Traditional Chinese Medicine, Jilin Agriculture Science and Technology University, Jilin, China
| | - Jihui Chen
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Rivera DE, Lažetić V, Troemel ER, Luallen RJ. RNA Fluorescence in situ Hybridization (FISH) to Visualize Microbial Colonization and Infection in Caenorhabditis elegans Intestines. J Vis Exp 2022:10.3791/63980. [PMID: 35969095 PMCID: PMC9969837 DOI: 10.3791/63980] [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] [Indexed: 10/31/2022] Open
Abstract
The intestines of wild Caenorhabditis nematodes are inhabited by a variety of microorganisms, including gut microbiome bacteria and pathogens, such as microsporidia and viruses. Because of the similarities between Caenorhabditis elegans and mammalian intestinal cells, as well as the power of the C. elegans system, this host has emerged as a model system to study host intestine-microbe interactions in vivo. While it is possible to observe some aspects of these interactions with bright-field microscopy, it is difficult to accurately classify microbes and characterize the extent of colonization or infection without more precise tools. RNA fluorescence in situ hybridization (FISH) can be used as a tool to identify and visualize microbes in nematodes from the wild or to experimentally characterize and quantify infection in nematodes infected with microbes in the lab. FISH probes, labeling the highly abundant small subunit ribosomal RNA, produce a bright signal for bacteria and microsporidian cells. Probes designed to target conserved regions of ribosomal RNA common to many species can detect a broad range of microbes, whereas targeting divergent regions of the ribosomal RNA is useful for narrower detection. Similarly, probes can be designed to label viral RNA. A protocol for RNA FISH staining with either paraformaldehyde (PFA) or acetone fixation is presented. PFA fixation is ideal for nematodes associated with bacteria, microsporidia, and viruses, whereas acetone fixation is necessary for the visualization of microsporida spores. Animals were first washed and fixed in paraformaldehyde or acetone. After fixation, FISH probes were incubated with samples to allow for the hybridization of probes to the desired target. The animals were again washed and then examined on microscope slides or using automated approaches. Overall, this FISH protocol enables detection, identification, and quantification of the microbes that inhabit the C. elegans intestine, including microbes for which there are no genetic tools available.
Collapse
Affiliation(s)
| | - Vladimir Lažetić
- School of Biological Sciences, University of California, San Diego
| | - Emily R. Troemel
- School of Biological Sciences, University of California, San Diego
| | | |
Collapse
|
23
|
Lalsiamthara J, Aballay A. The gut efflux pump MRP-1 exports oxidized glutathione as a danger signal that stimulates behavioral immunity and aversive learning. Commun Biol 2022; 5:422. [PMID: 35513700 PMCID: PMC9072357 DOI: 10.1038/s42003-022-03381-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 04/19/2022] [Indexed: 11/09/2022] Open
Abstract
Innate immune surveillance, which monitors the presence of potentially harmful microorganisms and the perturbations of host physiology that occur in response to infections, is critical to distinguish pathogens from beneficial microbes. Here, we show that multidrug resistance-associated protein-1 (MRP-1) functions in the basolateral membrane of intestinal cells to transport byproducts of cellular redox reactions to control both molecular and behavioral immunity in Caenorhabditis elegans. Pseudomonas aeruginosa infection disrupts glutathione homeostasis, leading to the excess production of the MRP-1 substrate, oxidized glutathione (GSSG). Extracellular GSSG triggers pathogen avoidance behavior and primes naïve C. elegans to induce aversive learning behavior via neural NMDA class glutamate receptor-1 (NMR-1). Our results indicate that MRP-1 transports GSSG, which acts as a danger signal capable of warning C. elegans of changes in intestinal homeostasis, thereby initiating a gut neural signal that elicits an appropriate host defense response. The multidrug resistance-associated protein-1 (MRP-1) functions in the basolateral membrane of intestinal cells to transport byproducts of cellular redox reactions to control both molecular and behavioral immunity in C. elegans.
Collapse
Affiliation(s)
- Jonathan Lalsiamthara
- Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health & Science University Portland, Oregon, OR, 97239, USA
| | - Alejandro Aballay
- Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health & Science University Portland, Oregon, OR, 97239, USA.
| |
Collapse
|
24
|
Lansdon P, Carlson M, Ackley BD. Wild-type Caenorhabditis elegans isolates exhibit distinct gene expression profiles in response to microbial infection. BMC Genomics 2022; 23:229. [PMID: 35321659 PMCID: PMC8943956 DOI: 10.1186/s12864-022-08455-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 02/28/2022] [Indexed: 11/20/2022] Open
Abstract
The soil-dwelling nematode Caenorhabditis elegans serves as a model system to study innate immunity against microbial pathogens. C. elegans have been collected from around the world, where they, presumably, adapted to regional microbial ecologies. Here we use survival assays and RNA-sequencing to better understand how two isolates from disparate climates respond to pathogenic bacteria. We found that, relative to N2 (originally isolated in Bristol, UK), CB4856 (isolated in Hawaii), was more susceptible to the Gram-positive microbe, Staphylococcus epidermidis, but equally susceptible to Staphylococcus aureus as well as two Gram-negative microbes, Providencia rettgeri and Pseudomonas aeruginosa. We performed transcriptome analysis of infected worms and found gene-expression profiles were considerably different in an isolate-specific and microbe-specific manner. We performed GO term analysis to categorize differential gene expression in response to S. epidermidis. In N2, genes that encoded detoxification enzymes and extracellular matrix proteins were significantly enriched, while in CB4856, genes that encoded detoxification enzymes, C-type lectins, and lipid metabolism proteins were enriched, suggesting they have different responses to S. epidermidis, despite being the same species. Overall, discerning gene expression signatures in an isolate by pathogen manner can help us to understand the different possibilities for the evolution of immune responses within organisms.
Collapse
Affiliation(s)
- Patrick Lansdon
- Department of Molecular Biosciences, University of Kansas, 5004 Haworth Hall, 1200 Sunnyside Ave, KS, 66045, Lawrence, USA
| | - Maci Carlson
- Department of Molecular Biosciences, University of Kansas, 5004 Haworth Hall, 1200 Sunnyside Ave, KS, 66045, Lawrence, USA
| | - Brian D Ackley
- Department of Molecular Biosciences, University of Kansas, 5004 Haworth Hall, 1200 Sunnyside Ave, KS, 66045, Lawrence, USA.
| |
Collapse
|
25
|
Manohar P, Loh B, Elangovan N, Loganathan A, Nachimuthu R, Leptihn S. A Multiwell-Plate Caenorhabditis elegans Assay for Assessing the Therapeutic Potential of Bacteriophages against Clinical Pathogens. Microbiol Spectr 2022; 10:e0139321. [PMID: 35171008 PMCID: PMC8849058 DOI: 10.1128/spectrum.01393-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/14/2022] [Indexed: 12/12/2022] Open
Abstract
In order to establish phage therapy as a standard clinical treatment for bacterial infections, testing of every phage to ensure the suitability and safety of the biological compound is required. While some issues have been addressed over recent years, standard and easy-to-use animal models to test phages are still rare. Testing of phages in highly suitable mammalian models such as mice is subjected to strict ethical regulations, while insect larvae such as the Galleria mellonella model suffer from batch-to-batch variations and require manual operator skills to inject bacteria, resulting in unreliable experimental outcomes. A much simpler model is the nematode Caenorhabditis elegans, which feeds on bacteria, a fast growing and easy to handle organism that can be used in high-throughput screening. In this study, two clinical bacterial strains of Escherichia coli, one Klebsiella pneumoniae, and one Enterobacter cloacae strain were tested on the model system together with lytic bacteriophages that we isolated previously. We developed a liquid-based assay, in which the efficiency of phage treatment was evaluated using a scoring system based on microscopy and counting of the nematodes, allowing increasing statistical significance compared to other assays such as larvae or mice. Our work demonstrates the potential to use Caenorhabditis elegans to test the virulence of strains of Klebsiella pneumoniae, Enterobacter cloacae, and EHEC/EPEC as well as the efficacy of bacteriophages to treat or prevent infections, allowing a more reliable evaluation for the clinical therapeutic potential of lytic phages. IMPORTANCE Validating the efficacy and safety of phages prior to clinical application is crucial to see phage therapy in practice. Current animal models include mice and insect larvae, which pose ethical or technical challenges. This study examined the use of the nematode model organism C. elegans as a quick, reliable, and simple alternative for testing phages. The data show that all the four tested bacteriophages can eliminate bacterial pathogens and protect the nematode from infections. Survival rates of the nematodes increased from <20% in the infection group to >90% in the phage treatment group. Even the nematodes with poly-microbial infections recovered during phage cocktail treatment. The use of C. elegans as a simple whole-animal infection model is a rapid and robust way to study the efficacy of phages before testing them on more complex model animals such as mice.
Collapse
Affiliation(s)
- Prasanth Manohar
- Zhejiang University-University of Edinburgh (ZJE) Institute, Zhejiang University, School of Medicine, Haining, Zhejiang, People’s Republic of China
- The Second Affiliated Hospital Zhejiang University (SAHZU), School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Belinda Loh
- Zhejiang University-University of Edinburgh (ZJE) Institute, Zhejiang University, School of Medicine, Haining, Zhejiang, People’s Republic of China
| | - Namasivayam Elangovan
- Department of Biotechnology, School of Bioscience, Periyar University, Salem, Tamil Nadu, India
| | - Archana Loganathan
- Antibiotic Resistance and Phage Therapy Lab, Department of Biomedical Science, School of Biosciences and Technology, Vellore, Tamil Nadu, India
| | - Ramesh Nachimuthu
- Antibiotic Resistance and Phage Therapy Lab, Department of Biomedical Science, School of Biosciences and Technology, Vellore, Tamil Nadu, India
| | - Sebastian Leptihn
- Zhejiang University-University of Edinburgh (ZJE) Institute, Zhejiang University, School of Medicine, Haining, Zhejiang, People’s Republic of China
- Department of Infectious Diseases, Sir Run Department Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- University of Edinburgh Medical School, Biomedical Sciences, College of Medicine & Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
26
|
Woods A, Parker D, Glick MM, Peng Y, Lenoir F, Mulligan E, Yu V, Piizzi G, Lister T, Lilly MD, Dzink-Fox J, Jansen JM, Ryder NS, Dean CR, Smith TM. High-Throughput Screen for Inhibitors of Klebsiella pneumoniae Virulence Using a Tetrahymena pyriformis Co-Culture Surrogate Host Model. ACS OMEGA 2022; 7:5401-5414. [PMID: 35187355 PMCID: PMC8851646 DOI: 10.1021/acsomega.1c06633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/08/2021] [Indexed: 05/31/2023]
Abstract
The continuing emergence of antibacterial resistance reduces the effectiveness of antibiotics and drives an ongoing search for effective replacements. Screening compound libraries for antibacterial activity in standard growth media has been extensively explored and may be showing diminishing returns. Inhibition of bacterial targets that are selectively important under in vivo (infection) conditions and, therefore, would be missed by conventional in vitro screens might be an alternative. Surrogate host models of infection, however, are often not suitable for high-throughput screens. Here, we adapted a medium-throughput Tetrahymena pyriformis surrogate host model that was successfully used to identify inhibitors of a hyperviscous Klebsiella pneumoniae strain to a high-throughput format and screened circa 1.2 million compounds. The screen was robust and identified confirmed hits from different chemical classes with potent inhibition of K. pneumoniae growth in the presence of T. pyriformis that lacked any appreciable direct antibacterial activity. Several of these appeared to inhibit capsule/mucoidy, which are key virulence factors in hypervirulent K. pneumoniae. A weakly antibacterial inhibitor of LpxC (essential for the synthesis of the lipid A moiety of lipopolysaccharides) also appeared to be more active in the presence of T. pyriformis, which is consistent with the role of LPS in virulence as well as viability in K. pneumoniae.
Collapse
Affiliation(s)
- Angela
L. Woods
- Infectious
Diseases, Novartis Institutes for Biomedical
Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - David Parker
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Meir M. Glick
- Chemical
Biology and Therapeutics, Novartis Institutes
for Biomedical Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Yunshan Peng
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Francois Lenoir
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Evan Mulligan
- Chemical
Biology and Therapeutics, Novartis Institutes
for Biomedical Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Vincent Yu
- Chemical
Biology and Therapeutics, Novartis Institutes
for Biomedical Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Grazia Piizzi
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Troy Lister
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Maria-Dawn Lilly
- Infectious
Diseases, Novartis Institutes for Biomedical
Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - JoAnn Dzink-Fox
- Infectious
Diseases, Novartis Institutes for Biomedical
Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Johanna M. Jansen
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research Inc, Emeryville California 94608-2916, United States
| | - Neil S. Ryder
- Infectious
Diseases, Novartis Institutes for Biomedical
Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Charles R. Dean
- Infectious
Diseases, Novartis Institutes for Biomedical
Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Thomas M. Smith
- Chemical
Biology and Therapeutics, Novartis Institutes
for Biomedical Research Inc, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
27
|
Pina-Pérez MC, Úbeda-Manzanaro M, Beyrer M, Martínez A, Rodrigo D. In vivo Assessment of Cold Atmospheric Pressure Plasma Technology on the Bioactivity of Spirulina. Front Microbiol 2022; 12:781871. [PMID: 35140692 PMCID: PMC8819064 DOI: 10.3389/fmicb.2021.781871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
The present study challenges the in vivo assessment of cold atmospheric pressure plasma (CAPP) technology on the bioactive activity (antioxidant/antiaging and antimicrobial potential) of Spirulina powder, using Caenorhabditis elegans as an animal model. Surface microdischarge cold atmospheric pressure plasma (SMD-CAPP) treatment was 3.3 W discharge power for 7 min. C. elegans lifespan and egg laying were used as indicators of antioxidant/antiaging potential of Spirulina (1 mg/mL), when grown with Spirulina CP-treated [E_SCP] and untreated [E_S], compared with a control [E_0] (non-supplemented with Spirulina). According to our results, under both Spirulina supplemented media [E_SCP and E_S] and for the first 17 days, nematodes experienced an increase in lifespan but without significant differences (p > 0.05) between control and Spirulina CP-treated. Regarding the in vivo assay of the antimicrobial potential of Spirulina against Salmonella enterica serovar Typhimurium (infected worms), no significant differences (p > 0.05) were found between the three exposure scenarios (control [S_0]; Spirulina supplemented media [S_S]; CP-treated Spirulina supplemented media [S_SCP]). According to present results, CAPP-treatment do not influence negatively the lifespan of C. elegans but a reduction in the Spirulina antiaging potential was found. No in vivo modifications in antimicrobial activity seem to be linked to CAPP-processed Spirulina.
Collapse
Affiliation(s)
- María Consuelo Pina-Pérez
- Departamento de Microbiología y Ecología, Universitat de València, Burjassot, Valencia, Spain
- Food Engineering Laboratory, Institute of Life Technologies, University of Applied Sciences and Arts Western-Switzerland (HES-SO) Valais-Wallis, Sion, Switzerland
| | - María Úbeda-Manzanaro
- Departamento Conservación y Calidad, Instituto de Agroquímica y Tecnología de Alimentos IATA - Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - Michael Beyrer
- Food Engineering Laboratory, Institute of Life Technologies, University of Applied Sciences and Arts Western-Switzerland (HES-SO) Valais-Wallis, Sion, Switzerland
| | - Antonio Martínez
- Departamento Conservación y Calidad, Instituto de Agroquímica y Tecnología de Alimentos IATA - Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - Dolores Rodrigo
- Departamento Conservación y Calidad, Instituto de Agroquímica y Tecnología de Alimentos IATA - Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
- *Correspondence: Dolores Rodrigo,
| |
Collapse
|
28
|
Yun B, Ryu S, Kang M, Lee J, Yoo J, Kim Y, Oh S. Probiotic Lacticaseibacillus rhamnosus GG Increased Longevity and Resistance Against Foodborne Pathogens in Caenorhabditis elegans by Regulating MicroRNA miR-34. Front Cell Infect Microbiol 2022; 11:819328. [PMID: 35127565 PMCID: PMC8807481 DOI: 10.3389/fcimb.2021.819328] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
In this study, we investigated the relation of probiotic activity of Lacticaseibacillus rhamnosus strain GG (LGG) and expression of microRNA to immune response and longevity in Caenorhabditis elegans host model. First, we evaluated the survival rate of C. elegans due to LGG exposure and bacterial colonization in the intestine. Next, the expression of mRNA and miRNA was analyzed in C. elegans exposure to LGG for 24 h using microarray. After exposure to LGG to C. elegans, colonized LGG was observed in the intestines of C. elegans and induced to extend lifespan. Moreover, persistent LGG in the intestine significantly enhanced the resistance of C. elegans exposed to both pathogenic bacteria and prolonged the lifespan of C. elegans. Transcriptome analysis indicated that LGG affected the expression levels of genes related to the innate immune response and upregulated the abundance of genes in multiple pathways of C. elegans, including Wnt signaling, TGF-beta signaling and mitogen-activated protein kinase (MAPK) pathways. In addition, qRT-PCR analysis confirmed that the expression of antibacterial genes was increased by LGG. Moreover, as the expression of microRNA miR-34 and immune-related pathways increased by exposure to LGG, the lifespan of C. elegans increased. However, in the miR-34 mutant C. elegans, the lifespan by LGG did not increase, so it was determined that miR-34 indirectly affects immune-related pathways. There was no significant difference in the expression of PMK-1 for LGG exposure in miR-34 mutants, suggesting that miR-34 may regulate PMK-1. In conclusion, we suggest that exposure of LGG to C. elegans enhances lifespan and resistance to food-borne pathogen infection by stimulating miR-34 and indirectly promoting PMK-1 activity.
Collapse
Affiliation(s)
- Bohyun Yun
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Sangdon Ryu
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| | - Minkyoung Kang
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Juyeon Lee
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Jiseon Yoo
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, South Korea
- *Correspondence: Younghoon Kim, ; Sangnam Oh,
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, South Korea
- *Correspondence: Younghoon Kim, ; Sangnam Oh,
| |
Collapse
|
29
|
Jespersen N, Monrroy L, Barandun J. Impact of Genome Reduction in Microsporidia. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 114:1-42. [PMID: 35543997 DOI: 10.1007/978-3-030-93306-7_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Microsporidia represent an evolutionary outlier in the tree of life and occupy the extreme edge of the eukaryotic domain with some of their biological features. Many of these unicellular fungi-like organisms have reduced their genomic content to potentially the lowest limit. With some of the most compacted eukaryotic genomes, microsporidia are excellent model organisms to study reductive evolution and its functional consequences. While the growing number of sequenced microsporidian genomes have elucidated genome composition and organization, a recent increase in complementary post-genomic studies has started to shed light on the impacts of genome reduction in these unique pathogens. This chapter will discuss the biological framework enabling genome minimization and will use one of the most ancient and essential macromolecular complexes, the ribosome, to illustrate the effects of extreme genome reduction on a structural, molecular, and cellular level. We outline how reductive evolution in microsporidia has shaped DNA organization, the composition and function of the ribosome, and the complexity of the ribosome biogenesis process. Studying compacted mechanisms, processes, or macromolecular machines in microsporidia illuminates their unique lifestyle and provides valuable insights for comparative eukaryotic structural biology.
Collapse
Affiliation(s)
- Nathan Jespersen
- Department of Molecular Biology, The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Science for Life Laboratory, Umeå University, Umeå, Sweden.
| | - Leonardo Monrroy
- Department of Molecular Biology, The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Science for Life Laboratory, Umeå University, Umeå, Sweden
| | - Jonas Barandun
- Department of Molecular Biology, The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Science for Life Laboratory, Umeå University, Umeå, Sweden.
| |
Collapse
|
30
|
Emerging technologies and infection models in cellular microbiology. Nat Commun 2021; 12:6764. [PMID: 34799563 PMCID: PMC8604907 DOI: 10.1038/s41467-021-26641-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/18/2021] [Indexed: 01/09/2023] Open
Abstract
The field of cellular microbiology, rooted in the co-evolution of microbes and their hosts, studies intracellular pathogens and their manipulation of host cell machinery. In this review, we highlight emerging technologies and infection models that recently promoted opportunities in cellular microbiology. We overview the explosion of microscopy techniques and how they reveal unprecedented detail at the host-pathogen interface. We discuss the incorporation of robotics and artificial intelligence to image-based screening modalities, biochemical mapping approaches, as well as dual RNA-sequencing techniques. Finally, we describe chips, organoids and animal models used to dissect biophysical and in vivo aspects of the infection process. As our knowledge of the infected cell improves, cellular microbiology holds great promise for development of anti-infective strategies with translational applications in human health.
Collapse
|
31
|
Kumar S, Anwer R, Azzi A. Virulence Potential and Treatment Options of Multidrug-Resistant (MDR) Acinetobacter baumannii. Microorganisms 2021; 9:microorganisms9102104. [PMID: 34683425 PMCID: PMC8541637 DOI: 10.3390/microorganisms9102104] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
Acinetobacter baumannii is an opportunistic pathogen which is undoubtedly known for a high rate of morbidity and mortality in hospital-acquired infections. A. baumannii causes life-threatening infections, including; ventilator-associated pneumonia (VAP), meningitis, bacteremia, and wound and urinary tract infections (UTI). In 2017, the World Health Organization listed A. baumannii as a priority-1 pathogen. The prevalence of A. baumannii infections and outbreaks emphasizes the direct need for the use of effective therapeutic agents for treating such infections. Available antimicrobials, such as; carbapenems, tigecycline, and colistins have insufficient effectiveness due to the appearance of multidrug-resistant strains, accentuating the need for alternative and novel therapeutic remedies. To understand and overcome this menace, the knowledge of recent discoveries on the virulence factors of A. baumannii is needed. Herein, we summarized the role of various virulence factors, including; outer membrane proteins, efflux pumps, biofilm, penicillin-binding proteins, and siderophores/iron acquisition systems. We reviewed the recent scientific literature on different A. baumannii virulence factors and the effective antimicrobial agents for the treatment and management of bacterial infections.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India;
| | - Razique Anwer
- Department of Pathology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317-4233, Saudi Arabia;
| | - Arezki Azzi
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317-4233, Saudi Arabia
- Correspondence:
| |
Collapse
|
32
|
Aljasir SF, D'Amico DJ. Probiotic potential of commercial dairy-associated protective cultures: In vitro and in vivo protection against Listeria monocytogenes infection. Food Res Int 2021; 149:110699. [PMID: 34600693 DOI: 10.1016/j.foodres.2021.110699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 01/23/2023]
Abstract
Protective bacterial cultures (PCs) are commercially available to producers to control undesirable microbes in foods, including foodborne pathogens such as Listeria monocytogenes. They are generally recognized as safe for consumption and many are capable of producing bacteriocins. Yet their potential to act as probiotics and confer a health benefit on the host is not known. This study investigated the ability of three commercial PCs to survive human gastrointestinal conditions and exert anti-infective properties against L. monocytogenes. Counts of two PCs of Lactiplantibacillus plantarum remained unchanged after exposure to simulated gastrointestinal conditions, whereas counts of the PC Lactococcus lactis subsp. lactis were reduced by 5.3 log CFU/mL. Cultures of Lactiplantibacillus plantarum and Lactococcus lactis subsp. lactis adhered to human Caco-2 epithelial cells at ∼ 6 log CFU/mL. This pretreatment reduced subsequent L. monocytogenes adhesion and invasion by 1-1.6 log CFU/mL and 3.8-4.9 log CFU/mL, respectively, compared to control. L. monocytogenes-induced cytotoxicity was also reduced from 29.1% in untreated monolayers to ∼ 8% in those treated with PCs. Pretreatment of Caco-2 monolayers with Lactococcus lactis subsp. lactis and one PC of Lactiplantibacillus plantarum reduced L. monocytogenes translocation by ≥ 1.2 log CFU/mL compared to control (≥ 94.5% inhibition). All PCs significantly reduced DextranFITC permeability through Caco-2 monolayers to approximately half that of control. Pretreatment with PCs also reduced L. monocytogenes-induced mortality in Caenorhabditis elegans. These findings demonstrate the potential for commercially produced PCs to exert probiotic effects in the host through protection against L. monocytogenes infection, thus providing an additional benefit to food safety beyond inhibiting pathogen growth, survival, and virulence in foods.
Collapse
Affiliation(s)
- Sulaiman F Aljasir
- Department of Animal Science, University of Connecticut, Agricultural Biotechnology Laboratory, 1390 Storrs Road, U-4163, Storrs, CT 06269-4163, USA.
| | - Dennis J D'Amico
- Department of Animal Science, University of Connecticut, Agricultural Biotechnology Laboratory, 1390 Storrs Road, U-4163, Storrs, CT 06269-4163, USA.
| |
Collapse
|
33
|
Impact of Tuning the Surface Charge Distribution on Colloidal Iron Oxide Nanoparticle Toxicity Investigated in Caenorhabditis elegans. NANOMATERIALS 2021; 11:nano11061551. [PMID: 34208275 PMCID: PMC8230852 DOI: 10.3390/nano11061551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/09/2021] [Indexed: 01/31/2023]
Abstract
Assessing the toxic effect in living organisms remains a major issue for the development of safe nanomedicines and exposure of researchers involved in the synthesis, handling and manipulation of nanoparticles. In this study, we demonstrate that Caenorhabditis elegans could represent an in vivo model alternative to superior mammalians for the collection of several physiological functionality parameters associated to both short-term and long-term effects of colloidally stable nanoparticles even in absence of microbial feeding, usually reported to be necessary to ensure appropriate intake. Contextually, we investigated the impact of surface charge on toxicity of superparamagnetic iron oxide coated with a wrapping polymeric envelop that confers them optimal colloidal stability. By finely tuning the functional group composition of this shallow polymer–obtaining totally anionic, partially pegylated, partially anionic and partially cationic, respectively–we showed that the ideal surface charge organization to optimize safety of colloidal nanoparticles is the one containing both cationic and anionic groups. Our results are in accordance with previous evidence that zwitterionic nanoparticles allow long circulation, favorable distribution in the tumor area and optimal tumor penetration and thus support the hypothesis that zwitterionic iron oxide nanoparticles could be an excellent solution for diagnostic imaging and therapeutic applications in nanooncology.
Collapse
|
34
|
White PS, Arslan D, Kim D, Penley M, Morran L. Host genetic drift and adaptation in the evolution and maintenance of parasite resistance. J Evol Biol 2021; 34:845-851. [PMID: 33783870 DOI: 10.1111/jeb.13785] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 03/22/2021] [Indexed: 01/17/2023]
Abstract
Host-parasite interactions may often be subject to opposing evolutionary forces, which likely influence the evolutionary trajectories of both partners. Natural selection and genetic drift are two major evolutionary forces that act in host and parasite populations. Further, population size is a significant determinant of the relative strengths of these forces. In small populations, drift may undermine the persistence of beneficial alleles, potentially impeding host adaptation to parasites. Here, we investigate two questions: (a) can selection pressure for increased resistance in small, susceptible host populations overcome the effects of drift and (b) can resistance be maintained in small host populations? To answer these questions, we experimentally evolved the host Caenorhabditis elegans against its bacterial parasite, Serratia marcescens, for 13 host generations. We found that strong selection favouring increased host resistance was insufficient to counteract drift in small populations, resulting in persistently high host mortality. Additionally, in small populations of resistant hosts, we found that selection for the maintenance of resistance is not always sufficient to curb the loss of resistance. We compared these results with selection in large host populations. We found that initially resistant, large host populations were able to maintain high levels of resistance. Likewise, initially susceptible, large host populations were able to gain resistance to the parasite. These results show that strong selection pressure for survival is not always sufficient to counteract drift. In consideration of C. elegans natural population dynamics, we suggest that drift may often impede selection in nature.
Collapse
Affiliation(s)
- P Signe White
- Department of Biology, College of Arts and Sciences, Emory University, Atlanta, GA, USA.,Population Biology, Ecology, and Evolution Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Danial Arslan
- Department of Biology, College of Arts and Sciences, Emory University, Atlanta, GA, USA
| | - David Kim
- Department of Biology, College of Arts and Sciences, Emory University, Atlanta, GA, USA
| | - McKenna Penley
- Department of Biology, College of Arts and Sciences, Emory University, Atlanta, GA, USA
| | - Levi Morran
- Department of Biology, College of Arts and Sciences, Emory University, Atlanta, GA, USA
| |
Collapse
|
35
|
Abstract
In its natural habitat, C. elegans encounters a wide variety of microbes, including food, commensals and pathogens. To be able to survive long enough to reproduce, C. elegans has developed a complex array of responses to pathogens. These activities are coordinated on scales that range from individual organelles to the entire organism. Often, the response is triggered within cells, by detection of infection-induced damage, mainly in the intestine or epidermis. C. elegans has, however, a capacity for cell non-autonomous regulation of these responses. This frequently involves the nervous system, integrating pathogen recognition, altering host biology and governing avoidance behavior. Although there are significant differences with the immune system of mammals, some mechanisms used to limit pathogenesis show remarkable phylogenetic conservation. The past 20 years have witnessed an explosion of host-pathogen interaction studies using C. elegans as a model. This review will discuss the broad themes that have emerged and highlight areas that remain to be fully explored.
Collapse
Affiliation(s)
- Céline N Martineau
- Aix Marseille Université, Inserm, CNRS, CIML, Turing Centre for Living Systems, Marseille, France
| | | | - Nathalie Pujol
- Aix Marseille Université, Inserm, CNRS, CIML, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
36
|
Poupet C, Chassard C, Nivoliez A, Bornes S. Caenorhabditis elegans, a Host to Investigate the Probiotic Properties of Beneficial Microorganisms. Front Nutr 2020; 7:135. [PMID: 33425969 PMCID: PMC7786404 DOI: 10.3389/fnut.2020.00135] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Caenorhabditis elegans, a non-parasitic nematode emerges as a relevant and powerful candidate as an in vivo model for microorganisms-microorganisms and microorganisms-host interactions studies. Experiments have demonstrated the probiotic potential of bacteria since they can provide to the worm a longer lifespan, an increased resistance to pathogens and to oxidative or heat stresses. Probiotics are used to prevent or treat microbiota dysbiosis and associated pathologies but the molecular mechanisms underlying their capacities are still unknown. Beyond safety and healthy aspects of probiotics, C. elegans represents a powerful way to design large-scale studies to explore transkingdom interactions and to solve questioning about the molecular aspect of these interactions. Future challenges and opportunities would be to validate C. elegans as an in vivo tool for high-throughput screening of microorganisms for their potential probiotic use on human health and to enlarge the panels of microorganisms studied as well as the human diseases investigated.
Collapse
Affiliation(s)
- Cyril Poupet
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| | | | | | - Stéphanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| |
Collapse
|
37
|
Alam ST, Le TAN, Park JS, Kwon HC, Kang K. Antimicrobial Biophotonic Treatment of Ampicillin-Resistant Pseudomonas aeruginosa with Hypericin and Ampicillin Cotreatment Followed by Orange Light. Pharmaceutics 2019; 11:E641. [PMID: 31805742 PMCID: PMC6956302 DOI: 10.3390/pharmaceutics11120641] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/24/2019] [Accepted: 11/28/2019] [Indexed: 12/30/2022] Open
Abstract
Bacterial antibiotic resistance is an alarming global issue that requires alternative antimicrobial methods to which there is no resistance. Antimicrobial photodynamic therapy (APDT) is a well-known method to combat this problem for many pathogens, especially Gram-positive bacteria and fungi. Hypericin and orange light APDT efficiently kill Staphylococcus aureus, methicillin-resistant Staphylococcus aureus (MRSA), and the yeast Candida albicans. Although Gram-positive bacteria and many fungi are readily killed with APDT, Gram-negative bacteria are difficult to kill due to their different cell wall structures. Pseudomonas aeruginosa is one of the most important opportunistic, life-threatening Gram-negative pathogens. However, it cannot be killed successfully by hypericin and orange light APDT. P. aeruginosa is ampicillin resistant, but we hypothesized that ampicillin could still damage the cell wall, which can promote photosensitizer uptake into Gram-negative cells. Using hypericin and ampicillin cotreatment followed by orange light, a significant reduction (3.4 log) in P. aeruginosa PAO1 was achieved. P. aeruginosa PAO1 inactivation and gut permeability improvement by APDT were successfully shown in a Caenorhabditis elegans model.
Collapse
Affiliation(s)
- Seemi Tasnim Alam
- Natural Products Informatics Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangwon-do 25451, Korea; (S.T.A.); (T.A.N.L.); (J.-S.P.); (H.C.K.)
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Gangwon-do 25451, Korea
| | - Tram Anh Ngoc Le
- Natural Products Informatics Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangwon-do 25451, Korea; (S.T.A.); (T.A.N.L.); (J.-S.P.); (H.C.K.)
| | - Jin-Soo Park
- Natural Products Informatics Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangwon-do 25451, Korea; (S.T.A.); (T.A.N.L.); (J.-S.P.); (H.C.K.)
| | - Hak Cheol Kwon
- Natural Products Informatics Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangwon-do 25451, Korea; (S.T.A.); (T.A.N.L.); (J.-S.P.); (H.C.K.)
| | - Kyungsu Kang
- Natural Products Informatics Research Center, Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangwon-do 25451, Korea; (S.T.A.); (T.A.N.L.); (J.-S.P.); (H.C.K.)
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Gangwon-do 25451, Korea
| |
Collapse
|
38
|
Sarkar S, Heise MT. Mouse Models as Resources for Studying Infectious Diseases. Clin Ther 2019; 41:1912-1922. [PMID: 31540729 PMCID: PMC7112552 DOI: 10.1016/j.clinthera.2019.08.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/05/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Mouse models are important tools both for studying the pathogenesis of infectious diseases and for the preclinical evaluation of vaccines and therapies against a wide variety of human pathogens. The use of genetically defined inbred mouse strains, humanized mice, and gene knockout mice has allowed the research community to explore how pathogens cause disease, define the role of specific host genes in either controlling or promoting disease, and identify potential targets for the prevention or treatment of a wide range of infectious agents. This review discusses several of the most commonly used mouse model systems, as well as new resources such as the Collaborative Cross as models for studying infectious diseases.
Collapse
Affiliation(s)
- Sanjay Sarkar
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark T Heise
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
39
|
Valliammai A, Sethupathy S, Priya A, Selvaraj A, Bhaskar JP, Krishnan V, Pandian SK. 5-Dodecanolide interferes with biofilm formation and reduces the virulence of Methicillin-resistant Staphylococcus aureus (MRSA) through up regulation of agr system. Sci Rep 2019; 9:13744. [PMID: 31551455 PMCID: PMC6760239 DOI: 10.1038/s41598-019-50207-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/30/2019] [Indexed: 12/20/2022] Open
Abstract
Methicillin resistant Staphylococcus aureus (MRSA) is a predominant human pathogen with high morbidity that is listed in the WHO high priority pathogen list. Being a primary cause of persistent human infections, biofilm forming ability of S. aureus plays a pivotal role in the development of antibiotic resistance. Hence, targeting biofilm is an alternative strategy to fight bacterial infections. The present study for the first time demonstrates the non-antibacterial biofilm inhibitory efficacy of 5-Dodecanolide (DD) against ATCC strain and clinical isolates of S. aureus. In addition, DD is able to inhibit adherence of MRSA on human plasma coated Titanium surface. Further, treatment with DD significantly reduced the eDNA synthesis, autoaggregation, staphyloxanthin biosynthesis and ring biofilm formation. Reduction in staphyloxanthin in turn increased the susceptibility of MRSA to healthy human blood and H2O2 exposure. Quantitative PCR analysis revealed the induced expression of agrA and agrC upon DD treatment. This resulted down regulation of genes involved in biofilm formation such as fnbA and fnbB and up regulation of RNAIII, hld, psmα and genes involved in biofilm matrix degradation such as aur and nuc. Inefficacy of DD on the biofilm formation of agr mutant further validated the agr mediated antibiofilm potential of DD. Notably, DD was efficient in reducing the in vivo colonization of MRSA in Caenorhabditis elegans. Results of gene expression studies and physiological assays unveiled the agr mediated antibiofilm efficacy of DD.
Collapse
Affiliation(s)
- Alaguvel Valliammai
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630003, Tamil Nadu, India
| | - Sivasamy Sethupathy
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630003, Tamil Nadu, India
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Arumugam Priya
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630003, Tamil Nadu, India
| | - Anthonymuthu Selvaraj
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, 630003, Tamil Nadu, India
| | | | | | | |
Collapse
|
40
|
Sanz-Puig M, Arana-Lozano A, Pina-Pérez MC, Fernández P, Martínez A, Rodrigo D. Occurrence of Salmonella typhimurium resistance under sublethal/repeated exposure to cauliflower infusion and infection effects on Caernohabditis elegans host test organism. FOOD SCI TECHNOL INT 2019; 26:151-159. [PMID: 31544526 DOI: 10.1177/1082013219873500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Resistant bacteria to antimicrobials are increasingly emerging in medical, food industry and livestock environments. The present research work assesses the capability of Salmonella enterica var Typhimurium to become adapted under the exposure to a natural cauliflower antimicrobial by-product infusion in consecutive repeated exposure cycles. Caenorhabditis elegans was proposed as in vivo host-test organism to compare possible changes in the virulent pattern of the different rounds treated S. enterica var Typhimurium and untreated bacterial cells. According to the obtained results, S. enterica var Typhimurium was able to generate resistance against a repeated exposure to cauliflower by-product infusion 5% (w/v), increasing the resistance with the number of exposed repetitions. Meanwhile, at the first exposure, cauliflower by-product infusion was effective in reducing S. enterica var Typhimurium (≈1 log10 cycle), and S. enterica var Typhimurium became resistant to this natural antimicrobial after the second and third treatment-round and was able to grow (≈1 log10 cycle). In spite of the increased resistance observed for repeatedly treated bacteria, the present study reveals no changes on C. elegans infection effects between resistant and untreated S. enterica var Typhimurium, according to phenotypic parameters evaluation (lifespan duration and egg-laying).
Collapse
Affiliation(s)
- Maria Sanz-Puig
- Instituto de Agroquímica y Tecnología de Alimentos - Consejo Superior de Investigaciones Científicas (IATA-CSIC), València, Spain
| | - Alejandra Arana-Lozano
- Instituto de Agroquímica y Tecnología de Alimentos - Consejo Superior de Investigaciones Científicas (IATA-CSIC), València, Spain
| | | | - Pablo Fernández
- Unidad Asociada al CSIC, Universidad Politécnica de Cartagena, Murcia, Spain
| | - Antonio Martínez
- Instituto de Agroquímica y Tecnología de Alimentos - Consejo Superior de Investigaciones Científicas (IATA-CSIC), València, Spain
| | - Dolores Rodrigo
- Instituto de Agroquímica y Tecnología de Alimentos - Consejo Superior de Investigaciones Científicas (IATA-CSIC), València, Spain
| |
Collapse
|
41
|
Kim JY, Le TAN, Lee SY, Song DG, Hong SC, Cha KH, Lee JW, Pan CH, Kang K. 3,3'-Diindolylmethane Improves Intestinal Permeability Dysfunction in Cultured Human Intestinal Cells and the Model Animal Caenorhabditis elegans. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9277-9285. [PMID: 31353906 DOI: 10.1021/acs.jafc.9b03039] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
3,3'-Diindolylmethane (DIM), a digestive metabolite originating from cruciferous vegetables, has dietary potential for the treatment of various human intestinal diseases. Although intestinal permeability dysfunction is closely related to the initiation and progression of human intestinal inflammatory diseases (IBDs), the effect of DIM on intestinal permeability is unclear. We evaluated the effect of DIM on the intestinal permeability of human intestinal cell monolayers and the animal model Caenorhabditis elegans, which were treated with IL-1β and Pseudomonas aeruginosa, respectively, to mimic IBD conditions. DIM substantially restored the intestinal permeability of differentiated Caco-2 cells by enhancing the expression of tight junction proteins (including occludin and ZO-1). Compared to the IL-1β single treatment (551.0 ± 49.0 Ω·cm2), DIM (10 μM) significantly increased the transepithelial electrical resistance (TEER) of Caco-2 cell monolayers (919.0 ± 66.4 Ω·cm2, p < 0.001). DIM also ameliorated the impaired intestinal permeability and extended the lifespan of C. elegans fed P. aeruginosa. The mean lifespan of DIM-treated worms (10.8 ± 1.3 days) was higher than that of control-treated worms (9.7 ± 1.1 days, p < 0.01). Thus, DIM is a potential nutraceutical candidate for the treatment of leaky gut syndrome by improving intestinal permeability.
Collapse
Affiliation(s)
- Joo Yeon Kim
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Tram Anh Ngoc Le
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - So Young Lee
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Dae-Geun Song
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Sung-Chul Hong
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Jae Wook Lee
- Natural Products Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology (UST) , Seoul 02792 , Republic of Korea
| | - Cheol-Ho Pan
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology (UST) , Seoul 02792 , Republic of Korea
| | - Kyungsu Kang
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology (UST) , Seoul 02792 , Republic of Korea
| |
Collapse
|
42
|
Casadei E, Salinas I. Comparative models for human nasal infections and immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 92:212-222. [PMID: 30513304 PMCID: PMC7102639 DOI: 10.1016/j.dci.2018.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 05/09/2023]
Abstract
The human olfactory system is a mucosal surface and a major portal of entry for respiratory and neurotropic pathogens into the body. Understanding how the human nasopharynx-associated lymphoid tissue (NALT) halts the progression of pathogens into the lower respiratory tract or the central nervous system is key for developing effective cures. Although traditionally mice have been used as the gold-standard model for the study of human nasal diseases, mouse models present important caveats due to major anatomical and functional differences of the human and murine olfactory system and NALT. We summarize the NALT anatomy of different animal groups that have thus far been used to study host-pathogen interactions at the olfactory mucosa and to test nasal vaccines. The goal of this review is to highlight the strengths and limitations of each animal model of nasal immunity and to identify the areas of research that require further investigation to advance human health.
Collapse
Affiliation(s)
- Elisa Casadei
- University of New Mexico, Department of Biology, Center for Evolutionary and Theoretical Immunology (CETI), Albuquerque, NM, USA.
| | - Irene Salinas
- University of New Mexico, Department of Biology, Center for Evolutionary and Theoretical Immunology (CETI), Albuquerque, NM, USA
| |
Collapse
|
43
|
Sublethal Effects of Ionic and Nanogold on the Nematode Caenorhabditis elegans. J Toxicol 2018; 2018:6218193. [PMID: 30515209 PMCID: PMC6236908 DOI: 10.1155/2018/6218193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/10/2018] [Indexed: 12/31/2022] Open
Abstract
The nematode Caenorhabditis elegans is used as an ecotoxicological model species in both aqueous medium and solid substrates. It is easy and of low cost to maintain in the laboratory and it produces hundreds of offspring within a short period of time. It also has a small body size (1 mm), making it possible for in vivo assays to be conducted in 12-well plates. Engineered nanomaterials (ENMs) are a class of emerging pollutants. Nanogold (nAu) is used in many consumer products and in vivo drug delivery. These materials can be released into the aquatic environment during production or discarding of consumer products. As nAu is insoluble in water, the sediment would become the final depository for the materials. It has become increasingly important to use sediment dwelling organisms to screen for possible toxicity of these ENMs. In this study C. elegans was exposed to a range of concentrations of nAu and ionic gold in M9-media, acting as a substitute for pore water. After 96-hour growth, fertility and reproduction were determined. Internal structure damage and internalisation of particles in C. elegans were determined by using SEM and CytoViva® Darkfield Imaging. From these images the nanomaterials are distributed around the oocytes in the reproductive organs, as well as the pharynx. Results obtained indicate that nAu affects reproduction more than growth due to internal gonad damage, albeit at very high exposure concentrations, indicating no toxicity at environmentally relevant concentrations. Ionic Au is more toxic than nAu and effects fertility and reproduction due to ion release. These results give more information regarding the toxicity and in vivo uptake of nAu and form part of an environmental risk assessment of ENMs.
Collapse
|
44
|
Kim Y, Choudhry QN, Chatterjee N, Choi J. Immune and xenobiotic response crosstalk to chemical exposure by PA01 infection in the nematode Caenorhabditis elegans. CHEMOSPHERE 2018; 210:1082-1090. [PMID: 30208533 DOI: 10.1016/j.chemosphere.2018.07.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 06/08/2023]
Abstract
Most organisms simultaneously face various chemical and biological stresses in the environment. Herein, we investigated how pathogen infection modifies an organism's response to chemical exposure. To explore this phenomenon, we conducted a toxicity study combined with pathogen infection by using the nematode Caenorhabditis elegans, the pathogen Pseudomonas aeruginosa, and various environmental chemicals. C. elegans preinfected with PA01, when subsequently exposed to chemicals, became sensitized to the toxicity of nonylphenol (NP) and cadmium (Cd), whereas they became tolerant to the toxicity of silver nanoparticles (AgNPs); this led us to conduct a mechanistic study focusing on AgNP exposure. A gene expression profiling study revealed that most of the immune response genes activated by PA01 infection remained activated after subsequent exposure to AgNPs, thereby suggesting that the acquired tolerance of C. elegans to AgNP exposure may be due to boosted immunity resulting from PA01 preinfection. Further, a functional genetic analysis revealed that the immune response pathway (i.e., PMK-1/p38 MAPK) was involved in defense against AgNP exposure in PA01-preinfected C. elegans, thus suggesting immune and stress response crosstalk to xenobiotic exposure. This study will aid in the elucidation of how pathogen infection impacts the way the defense system responds to subsequent xenobiotic exposure.
Collapse
Affiliation(s)
- Youngho Kim
- School of Environmental Engineering, University of Seoul, 163 Siripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea
| | - Qaisra Naheed Choudhry
- School of Environmental Engineering, University of Seoul, 163 Siripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea
| | - Nivedita Chatterjee
- School of Environmental Engineering, University of Seoul, 163 Siripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, 163 Siripdaero, Dongdaemun-gu, Seoul, 02504, Republic of Korea.
| |
Collapse
|
45
|
Khan F, Jain S, Oloketuyi SF. Bacteria and bacterial products: Foe and friends to Caenorhabditis elegans. Microbiol Res 2018; 215:102-113. [DOI: 10.1016/j.micres.2018.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/11/2018] [Accepted: 06/24/2018] [Indexed: 02/07/2023]
|
46
|
Real-time monitoring of immune responses under pathogen invasion and drug interference by integrated microfluidic device coupled with worm-based biosensor. Biosens Bioelectron 2018; 110:233-238. [DOI: 10.1016/j.bios.2018.03.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/09/2018] [Accepted: 03/27/2018] [Indexed: 01/17/2023]
|
47
|
Infection Models: Novel Potato Blight-like Pathogens in Worms. Curr Biol 2018; 28:R273-R275. [DOI: 10.1016/j.cub.2018.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
48
|
Muthamil S, Devi VA, Balasubramaniam B, Balamurugan K, Pandian SK. Green synthesized silver nanoparticles demonstrating enhanced in vitro and in vivo antibiofilm activity against Candida
spp. J Basic Microbiol 2018; 58:343-357. [DOI: 10.1002/jobm.201700529] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/08/2018] [Accepted: 01/20/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Subramanian Muthamil
- Department of Biotechnology; Science Campus; Alagappa University; Karaikudi Tamil Nadu India
| | - Vivekanandham Amsa Devi
- Department of Biotechnology; Science Campus; Alagappa University; Karaikudi Tamil Nadu India
| | | | | | | |
Collapse
|
49
|
GIBSON AMANDAK, MORRAN LEVIT. A Model for Evolutionary Ecology of Disease: The Case for Caenorhabditis Nematodes and Their Natural Parasites. J Nematol 2018. [DOI: 10.21307/jofnem-2017-083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
50
|
GERBABA TEKLUK, GREEN-HARRISON LUKE, BURET ANDREG. Modeling Host-Microbiome Interactions in Caenorhabditis elegans. J Nematol 2018. [DOI: 10.21307/jofnem-2017-082] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|