1
|
Margarita V, Lodde V, Rappelli P, Doro L, Montella A, Fiori PL, Campesi I. The different innate immune response to infections in males and females emerges before birth. Life Sci 2025; 369:123521. [PMID: 40044031 DOI: 10.1016/j.lfs.2025.123521] [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: 11/08/2024] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 03/14/2025]
Abstract
AIM Sex-specific immune responses to intra-amniotic infections remain poorly understood despite their key role in preterm birth. METHODS We infected male and female amniotic fluid cells (AFCs) with M. hominis to explore the potential impact of sex-specific innate immune responses, evaluating the gene and protein expression levels of IL-1β, IL-6, IL-8, TNF-α, TLR2 and TLR4, the production of nitrites, and the levels of selected miRNAs. KEY FINDINGS The gene expression of IL-1β, IL-6, IL-8, TNF-α, and TLR2 were elevated in infected females AFCs, while only IL-6, IL-8 and TLR4 expression were up-regulated in infected males. Protein levels of IL-6, TNF-α and TLR2 were up-regulated exclusively in infected females. Furthermore, infected female AFCs produced higher levels of nitrites. MiRNA expression revealed an up-regulation of miR-29a-3p in infected females, and miR-223-3p in infected males, with miR-29b-3p showing up-regulation in both sexes upon infection. SIGNIFICANCE The response to intrauterine infections differs between males and females. Female foetuses may possess a greater capacity to manage the infection and inflammation, underscoring the importance of personalized prenatal care.
Collapse
Affiliation(s)
| | - Valeria Lodde
- Department of Biomedical Science, University of Sassari, Sassari, Italy
| | - Paola Rappelli
- Department of Biomedical Science, University of Sassari, Sassari, Italy
| | - Laura Doro
- Department of Biomedical Science, University of Sassari, Sassari, Italy
| | - Andrea Montella
- Department of Biomedical Science, University of Sassari, Sassari, Italy
| | - Pier Luigi Fiori
- Department of Biomedical Science, University of Sassari, Sassari, Italy
| | - Ilaria Campesi
- Department of Biomedical Science, University of Sassari, Sassari, Italy; Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy.
| |
Collapse
|
2
|
da Cunha e Silva FA, da Silva BR, de Barros LR, Beraldo-Neto E, Maleski ALA, Alberto-Silva C. Snake Venom Peptide Fractions from Bothrops jararaca and Daboia siamensis Exhibit Differential Neuroprotective Effects in Oxidative Stress-Induced Zebrafish Models. Pharmaceuticals (Basel) 2025; 18:678. [PMID: 40430497 PMCID: PMC12115354 DOI: 10.3390/ph18050678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 04/25/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction: Snake venoms are rich sources of bioactive peptides with therapeutic potential, particularly against neurodegenerative diseases linked to oxidative stress. While the peptide fraction (<10 kDa) from Bothrops jararaca venom has shown in vitro neuroprotection, analogous fractions from related species remain unexplored in vivo. Methods: This study comparatively evaluated the neuroprotective effects of two peptide fractions (pf) from Daboia siamensis (pf-Ds) and B. jararaca (pf-Bj) against H2O2-induced oxidative stress using in vitro (PC12 cells) and in vivo (zebrafish, Danio rerio) models. Results: In vitro, pf-Ds (1 µg mL-1) did not protect PC12 cells against H2O2-induced cytotoxicity, unlike previously reported effects of pf-Bj. In vivo, neither pf-Ds nor pf-Bj (1-20 µg mL-1) induced significant developmental toxicity in zebrafish larvae up to 120 h post-fertilization (hpf). The neuroprotective effects of both pf were evaluated using two experimental models: (I) Larvae at 96 hpf were exposed to either pf-Ds or pf-Bj (10 µg mL-1) for 4 h, followed by co-exposure to H2O2 (0.2 mmol L-1) for an additional 10 h to induce oxidative stress (4-20 h model); (II) Embryos at 4 hpf were treated with pf-Ds or pf-Bj (10 µg mL-1) continuously until 96 hpf, after which they were exposed to H2O2 (0.2 mmol L-1) for another 24 h (96-120 h model). In a short-term treatment model, neither fraction reversed H2O2-induced deficits in metabolism or locomotor activity. However, in a prolonged treatment model, pf-Bj significantly reversed the H2O2-induced locomotor impairment, whereas pf-Ds did not confer protection. Conclusions: These findings demonstrate, for the first time, the in vivo neuroprotective potential of pf-Bj against oxidative stress-induced behavioral deficits in zebrafish, contingent on the treatment regimen. The differential effects between pf-Ds and pf-Bj highlight species-specific venom composition and underscore the value of zebrafish for evaluating venom-derived peptides.
Collapse
Affiliation(s)
- Felipe Assumpção da Cunha e Silva
- Experimental Morphophysiology Laboratory, Natural and Humanities Sciences Center (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (F.A.d.C.e.S.); (B.R.d.S.); (L.R.d.B.); (A.L.A.M.)
| | - Brenda Rufino da Silva
- Experimental Morphophysiology Laboratory, Natural and Humanities Sciences Center (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (F.A.d.C.e.S.); (B.R.d.S.); (L.R.d.B.); (A.L.A.M.)
| | - Leticia Ribeiro de Barros
- Experimental Morphophysiology Laboratory, Natural and Humanities Sciences Center (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (F.A.d.C.e.S.); (B.R.d.S.); (L.R.d.B.); (A.L.A.M.)
| | - Emidio Beraldo-Neto
- Biochemistry Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil;
| | - Adolfo Luis Almeida Maleski
- Experimental Morphophysiology Laboratory, Natural and Humanities Sciences Center (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (F.A.d.C.e.S.); (B.R.d.S.); (L.R.d.B.); (A.L.A.M.)
| | - Carlos Alberto-Silva
- Experimental Morphophysiology Laboratory, Natural and Humanities Sciences Center (CCNH), Universidade Federal do ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (F.A.d.C.e.S.); (B.R.d.S.); (L.R.d.B.); (A.L.A.M.)
| |
Collapse
|
3
|
Lachnit T, Ulrich L, Willmer FM, Hasenbein T, Steiner LX, Wolters M, Herbst EM, Deines P. Nutrition-induced changes in the microbiota can cause dysbiosis and disease development. mBio 2025; 16:e0384324. [PMID: 39998180 PMCID: PMC11980362 DOI: 10.1128/mbio.03843-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Eukaryotic organisms are associated with complex microbial communities. Changes within these communities have been implicated in disease development. Nonetheless, it remains unclear whether these changes are a cause or a consequence of disease. Here, we report a causal link between environment-induced shifts in the microbiota and disease development. Using the model organism Hydra, we observed changes in microbial composition when transferring laboratory-grown Hydra to natural lake environments. These shifts were caused not only by new colonizers, through the process of community coalescence (merging of previously separate microbial communities), but also by lake water nutrients. Moreover, selective manipulation of the nutrient environment induced compound-specific shifts in the microbiota followed by disease development. Finally, L-arginine supplementation alone caused a transition in Pseudomonas from symbiotic to pathogenic, leading to an upregulation of immune response genes, tissue degradation, and host death. These findings challenge the notion that the host-associated microbiota is exclusively controlled by the host, highlighting the dynamic interplay between host epithelial environment, microbial colonizer pool, and nutrient conditions of the surrounding water. Furthermore, our results show that overfeeding of the microbiota allows for uncontrolled microbial growth and versatile interactions with the host. Environmental conditions may thus render symbionts a potential hazard to their hosts, blurring the divide between pathogenic and non-pathogenic microbes.IMPORTANCEThis study highlights the critical need to understand the dynamic interplay between host-associated microbiota and environmental factors to obtain a holistic view on organismal health. Our results demonstrate that ecosystem-wide microbial trafficking (community coalescence) and environmental nutrient conditions reshape microbial communities with profound implications for host health. By exploring nutrient-driven changes in microbial composition, our research finds experimental support for the "overfeeding hypothesis," which states that overfeeding alters the functionality of the host microbiota such that an overabundance in nutrients can facilitate disease development, transforming non-pathogenic microbes into pathogens. These findings emphasize the critical role of metabolic interactions driving microbial pathogenicity. Furthermore, our research provides empirical evidence for the "pathogenic potential" concept, challenging traditional distinctions between pathogenic and non-pathogenic microbes and supporting the idea that any microbe can become pathogenic under certain conditions.
Collapse
Affiliation(s)
- Tim Lachnit
- Zoological Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Laura Ulrich
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Fiete M. Willmer
- Zoological Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Tim Hasenbein
- Institute of Pharmacology and Toxicology, Technical University of Munich, München, Germany
| | - Leon X. Steiner
- RU Marine Symbioses, RD3 Marine Ecology, GEOMAR Helmholtz Centre for Ocean Research, Kiel, Germany
| | - Maria Wolters
- Fakultät Nachhaltigkeit, Leuphana Universität Lüneburg, Lüneburg, Germany
| | - Eva M. Herbst
- Experimental Orthopedics and Trauma Surgery, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Peter Deines
- Zoological Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
4
|
Chi J, Patterson JS, Jin Y, Kim KJ, Lalime N, Hawley D, Lewis F, Li L, Wang X, Campen MJ, Cui JY, Gu H. Metabolic Reprogramming in Gut Microbiota Exposed to Polystyrene Microplastics. Biomedicines 2025; 13:446. [PMID: 40002859 PMCID: PMC11853289 DOI: 10.3390/biomedicines13020446] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Microplastics (MPs) are small plastic fragments with diameters less than 5 mm in size and are prevalent in everyday essentials and consumables. Large global plastic production has now led to a flooding of MPs in our natural environment. Due to their detrimental impacts on the planet's ecosystems and potentially our health, MPs have emerged as a significant public health concern. In this pilot study, we hypothesize that MPs exposure will negatively affect gut microbiota composition and function, in which metabolic reprogramming plays an important role. Methods: Using in vitro experiments, three bacterial strains (Escherichia coli MG1655, Nissle 1917, and Lactobacillus rhamnosus) were selected to investigate the impacts of MPs exposure. The bacterial strains were individually cultured in an anaerobic chamber and exposed to 1 µm polystyrene MPs at various concentrations (0, 10, 20, 50, 100, and 500 µg/mL) in the culture medium. Results: MPs exposure reduced the growth of all three bacterial strains in a dose-dependent manner. Liquid chromatography mass spectrometry (LC-MS)-based untargeted metabolomics revealed significant differences in multiple metabolic pathways, such as sulfur metabolism and amino sugar and nucleotide sugar metabolism. In addition, we extracted gut microbiota from C57BL/6 mice, and 16S rRNA sequencing results showed a significant upregulation of Lactobacillales and a significant reduction in Erysipelotrichales due to MPs exposure. Furthermore, targeted and untargeted metabolomics corroborated the in vitro results and revealed alterations in microbial tryptophan metabolism and energy producing pathways, such as glycolysis/gluconeogenesis and the pentose phosphate pathway. Conclusions: These findings provide evidence that MPs exposure causes comprehensive changes to healthy gut microbiota, which may also provide insights into the mechanistic effects of MPs exposure in humans.
Collapse
Affiliation(s)
- Jinhua Chi
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA;
| | - Jeffrey S. Patterson
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
| | - Yan Jin
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA;
| | - Kyle Joohyung Kim
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA; (K.J.K.); (J.Y.C.)
| | - Nicole Lalime
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA;
| | - Daniella Hawley
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (D.H.); (X.W.)
| | - Freeman Lewis
- Environmental Health Sciences, Florida International University, Miami, FL 33199, USA;
| | - Lingjun Li
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
| | - Xuan Wang
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA; (D.H.); (X.W.)
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences, Albuquerque, NM 87106, USA;
| | - Julia Yue Cui
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA; (K.J.K.); (J.Y.C.)
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (J.C.); (J.S.P.); (L.L.)
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA;
| |
Collapse
|
5
|
Costantini C, Brancorsini S, Grignani F, Romani L, Bellet MM. Circadian metabolic adaptations to infections. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230473. [PMID: 39842481 PMCID: PMC11753887 DOI: 10.1098/rstb.2023.0473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/22/2024] [Accepted: 05/16/2024] [Indexed: 01/24/2025] Open
Abstract
Circadian clocks are biological oscillators that evolved to coordinate rhythms in behaviour and physiology around the 24-hour day. In mammalian tissues, circadian rhythms and metabolism are highly intertwined. The clock machinery controls rhythmic levels of circulating hormones and metabolites, as well as rate-limiting enzymes catalysing biosynthesis or degradation of macromolecules in metabolic tissues, such control being exerted both at the transcriptional and post-transcriptional level. During infections, major metabolic adaptation occurs in mammalian hosts, at the level of both the single immune cell and the whole organism. Under these circumstances, the rhythmic metabolic needs of the host intersect with those of two other players: the pathogen and the microbiota. These three components cooperate or compete to meet their own metabolic demands across the 24 hours. Here, we review findings describing the circadian regulation of the host response to infection, the circadian metabolic adaptations occurring during host-microbiota-pathogen interactions and how such regulation can influence the immune response of the host and, ultimately, its own survival.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Stefano Brancorsini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Francesco Grignani
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Marina Maria Bellet
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| |
Collapse
|
6
|
Scribani Rossi C, Molina-Henares MA, Espinosa-Urgel M, Rinaldo S. Exploring the Metabolic Response of Pseudomonas putida to L-arginine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1476:31-47. [PMID: 38429473 DOI: 10.1007/5584_2024_797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Beyond their role as protein-building units, amino acids are modulators of multiple behaviours in different microorganisms. In the root-colonizing beneficial bacterium Pseudomonas putida (recently proposed to be reclassified as alloputida) KT2440, current evidence suggests that arginine functions both as a metabolic indicator and as an environmental signal molecule, modulating processes such as chemotactic responses, siderophore-mediated iron uptake or the levels of the intracellular second messenger cyclic diguanylate (c-di-GMP). Using microcalorimetry and extracellular flux analysis, in this work we have studied the metabolic adaptation of P. putida KT2440 to the presence of L-arginine in the growth medium, and the influence of mutations related to arginine metabolism. Arginine causes rapid changes in the respiratory activity of P. putida, particularly magnified in a mutant lacking the transcriptional regulator ArgR. The metabolic activity of mutants affected in arginine transport and metabolism is also altered during biofilm formation in the presence of the amino acid. The results obtained here further support the role of arginine as a metabolic signal in P. putida and the relevance of ArgR in the adaptation to the amino acid. They also serve as proof of concept on the use of calorimetric and extracellular flux techniques to analyse metabolic responses in bacteria and the impact of different mutant backgrounds on such responses.
Collapse
Affiliation(s)
- Chiara Scribani Rossi
- Laboratory affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - María Antonia Molina-Henares
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidin, CSIC, Granada, Spain
| | - Manuel Espinosa-Urgel
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidin, CSIC, Granada, Spain.
| | - Serena Rinaldo
- Laboratory affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
7
|
Mallik SR, Joshi K, Radhakrishnan GK. The arginine/ornithine binding protein ArgT plays an essential role in Brucella neotomae/ Brucella melitensis to prevent intracellular killing and contribute to chronic persistence in the host. Virulence 2024; 15:2421983. [PMID: 39463062 PMCID: PMC11540086 DOI: 10.1080/21505594.2024.2421983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 09/13/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
Brucella species are facultative intracellular bacterial pathogens that cause the contagious zoonotic disease, brucellosis. Brucella spp. infect a wide range of animals, including livestock, wild animals, and marine mammals. Compared with other invasive bacterial pathogens, partial information is available on the virulence factors of Brucella that enable them to survive in the host. Here, we performed transposon-based random mutagenesis of B. neotomae and identified the arginine/ornithine binding protein, ArgT, as one of the crucial virulence determinants of Brucella. Deleting ArgT from B. neotomae or B. melitensis resulted in its attenuation in macrophages, which was restored upon complementation with an ArgT expression plasmid. We observed that macrophages infected with ΔArgT-B. neotomae produced elevated levels of NO due to the inability of these mutants to deplete the host intracellular arginine through their importer. Furthermore, defective survival of ΔArgT B. neotomae and B. melitensis was observed in the infected mice, which correlated with enhanced NO production in the mice. Our studies revealed that ArgT plays a vital role in preventing intracellular killing and contributes to the chronic persistence of B. neotomae/B. melitensis in the host. This study highlights the essential role of arginine in clearing intracellular infections and the subversion of this host defense mechanism by intracellular pathogens for their chronic persistence.
Collapse
Affiliation(s)
- Sushree Rekha Mallik
- Laboratory of Immunology and Microbial Pathogenesis, BRIC-National Institute of Animal Biotechnology (BRIC-NIAB), Hyderabad, Telangana, India
- BRIC-Regional Centre for Biotechnology (BRIC-RCB), Faridabad, Haryana, India
| | - Kiranmai Joshi
- Laboratory of Immunology and Microbial Pathogenesis, BRIC-National Institute of Animal Biotechnology (BRIC-NIAB), Hyderabad, Telangana, India
- BRIC-Regional Centre for Biotechnology (BRIC-RCB), Faridabad, Haryana, India
| | - Girish K. Radhakrishnan
- Laboratory of Immunology and Microbial Pathogenesis, BRIC-National Institute of Animal Biotechnology (BRIC-NIAB), Hyderabad, Telangana, India
| |
Collapse
|
8
|
Ring BE, Shepard GE, Khadka S, Holmes CL, Bachman MA, Mike LA. Arginine Regulates the Mucoid Phenotype of Hypervirulent Klebsiella pneumoniae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624485. [PMID: 39605402 PMCID: PMC11601523 DOI: 10.1101/2024.11.20.624485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Hypervirulent Klebsiella pneumoniae is associated with severe community-acquired infections. Hypervirulent K. pneumoniae colonies typically exhibit a mucoid phenotype. K. pneumoniae mucoidy is influenced by a complex combination of environmental factors and genetic mechanisms. Mucoidy results from altered capsular polysaccharide chain length, yet the specific environmental cues regulating this phenotype and their impact on pathogenesis remain unclear. This study demonstrates that casamino acids enhance the mucoidy phenotype but do not affect total capsular polysaccharide levels. Through targeted screening of each amino acid present in casamino acids, we identified that arginine is necessary and sufficient to stimulate the mucoid phenotype without altering capsule abundance. Furthermore, arginine activates the rmpADC promoter, increasing rmpD transcript levels, which in turn modulates capsular polysaccharide chain length and diversity. The arginine regulator, ArgR, plays a pivotal role in this regulatory cascade since deleting argR decreases mucoidy and increases capsular polysaccharide chain length diversity. Additionally, the ∆argR mutant displays increased macrophage association and has a substantial competitive defect in the lungs of mice, suggesting a link between arginine-dependent gene regulation, immune evasion and in vivo fitness. We discovered that arginine-dependent regulation of mucoidy is conserved in four additional hypervirulent K. pneumoniae isolates likely via a conserved ARG binding box present in rmp promoters. Our findings support a model in which arginine activates ArgR and increases mucoidy in hypervirulent K. pneumoniae. As a result, it is possible that arginine-dependent regulation of mucoidy allows hypervirulent K. pneumoniae to adapt the cell surface across different niches. This study underscores the significance of arginine as a regulatory signal in bacterial virulence.
Collapse
Affiliation(s)
- Brooke E. Ring
- Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Grace E. Shepard
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Saroj Khadka
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Caitlyn L. Holmes
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Michael A. Bachman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Laura A. Mike
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Ye X, Ma C, Guo W, Guo Y, Li DD, Zhou S, Hu Q, Hong Y, Xie Z, Wang L. Metabolomic analysis reveals potential role of immunometabolism dysregulation in recurrent pregnancy loss. Front Endocrinol (Lausanne) 2024; 15:1476774. [PMID: 39444455 PMCID: PMC11496058 DOI: 10.3389/fendo.2024.1476774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Background Recurrent pregnancy loss (RPL) affects women's reproductive health seriously, with immune dysfunction playing a key role in its cause, yet the exact mechanisms remain elusive. We aim to investigate potential mechanisms and identify biomarkers linked to RPL. Methods Immune cytokine testing and metabolomic profiling were conducted on the serum of 34 RPL patients and 30 healthy individuals. The metabolic pathways of the differential metabolites were analyzed, and specific metabolites were validated through targeted profiling. Potential biomarkers were identified, and the relationships between immune cytokines and differential metabolites were explored. Results In the RPL group, serum interleukin-6 and interleukin-10 levels were significantly higher, while interleukin-2 and interferon-γ were significantly lower. A total of 296 differential metabolites were detected by untargeted metabolomic profiling between the RPL and control groups, with most linked to amino acid metabolism. Targeted metabolomic profiling of amino acid metabolism revealed upregulation of indole-3-acetic acid, tyrosine, glycine, isoleucine, tryptophan, lysine, aspartic acid, arginine, leucine, threonine, glutamic acid, cystine, and phenylpyruvic acid (PPA) in the RPL group. Moreover, PPA and 5-hydroxy-L-tryptophan showed great potential in predicting RPL in a diagnostic model. Cystine and tyrosine were associated with immune cytokines in correlation analysis. Conclusion The study highlights the role of amino acid metabolism in RPL pathogenesis, suggesting that PPA and 5-HTP may be potential predictive indicators, while cystine and tyrosine may potentially regulate immune responses related to RPL. Further investigation into the molecular mechanisms underlying these findings could potentially result in the creation of novel diagnostic and therapeutic approaches for RPL.
Collapse
Affiliation(s)
- Xiaofeng Ye
- Reproductive Medicine Centre, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Chong Ma
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Wenqi Guo
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yan Guo
- Department of Gynecology and Obstetrics, Huiyang District Maternal and Child Health Care Hospital, Huizhou, China
| | - Dong-dong Li
- Department of Gynecology and Obstetrics, Graduate College of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Sihang Zhou
- Reproductive Medicine Centre, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Health Science Center, Shenzhen University, Shenzhen, China
| | - Qingyu Hu
- Reproductive Medicine Centre, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Health Science Center, Shenzhen University, Shenzhen, China
| | - Yanjun Hong
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Liping Wang
- Reproductive Medicine Centre, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
10
|
Bijle MN, Abdalla MM, Yiu C. The effect of arginine on the growth of probiotics. J Dent 2024; 149:105272. [PMID: 39074576 DOI: 10.1016/j.jdent.2024.105272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024] Open
Abstract
OBJECTIVE(S) The study objective was to examine the effect of arginine (Arg) supplementation on the growth of probiotics. METHODS Lacticaseibacillus rhamnosus GG, Lactiplantibacillus plantarum, and Lactobacillus acidophilus were identified as potential probiotics. L. rhamnosus GG and L. plantarum were selected for further experimentation. The probiotics were co-treated with 0.9 % NaCl (negative control), 0.5 % Arg, and 1.0 % Arg in a 1:1 ratio for 24 h at 5 % CO2, 37 °C. The probiotics were tested for growth profiles, spectroscopic turbidity assay, metabolic assays (XTT and WST-8), live/dead cell assessment using confocal laser scanning microscopy (CLSM), and colony forming units (CFU). RESULTS The growth profiles of L. rhamnosus GG and L. plantarum were found to be similar, whereas L. acidophilus showed minimal or no transition from the initial lag phase. In the turbidity assay, the end-point absorbance for L. rhamnosus GG with 1.0 % Arg was significantly lower than 0.9 % NaCl and 0.5 % Arg (p < 0.05). For metabolic assays and CFU, increasing concentrations of Arg increased the viable cells for L. rhamnosus GG (p < 0.05), but decreased viability for L. plantarum (p < 0.05). Metabolic assays with dual-species bacterial suspensions indicated that Arg co-treatment inhibited viable proportions compared to control (p < 0.05). The dead cell proportion was significantly lower than live cell proportion for all tested interventions and probiotics (p < 0.05). CONCLUSION Increasing concentrations of Arg promote the growth of L. rhamnosus GG, while conversely inhibiting the growth of L. plantarum. Therefore, the effect of prebiotic Arg on probiotics is concentration-dependent, leading to a selective promotion or inhibition of growth. CLINICAL SIGNIFICANCE The present study results show that Arg supplementation can selectively enhance the growth of L. rhamnosus GG while inhibit the growth of L. plantarum. This underscores the need to consider strain-specific responses in probiotic formulations when developing Arg-based synbiotics for modulating biofilms and creating ecologically homeostatic biofilm microenvironments.
Collapse
Affiliation(s)
| | - Mohamed Mahmoud Abdalla
- Faculty of Dentistry, The University of Hong Kong, Hong Kong; Dental Biomaterials, Faculty of Dental Medicine Al-Azhar University, Cairo, Egypt
| | - Cynthia Yiu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| |
Collapse
|
11
|
Hu X, Liu X, Feng D, Xu T, Li H, Hu C, Wang Z, Liu X, Yin P, Shi X, Shang D, Xu G. Polarization of Macrophages in Tumor Microenvironment Using High-Throughput Single-Cell Metabolomics. Anal Chem 2024; 96:14935-14943. [PMID: 39221578 DOI: 10.1021/acs.analchem.4c02989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Macrophages consist of a heterogeneous population of functionally distinct cells that participate in many physiological and pathological processes. They exhibit prominent plasticity by changing their different functional phenotypes represented by proinflammatory (M1) and anti-inflammatory (M2) in response to different environmental stimuli. Emerging evidence illustrates the importance of intracellular metabolic pathways in macrophage polarizations and functions. In the tumor microenvironment (TME), macrophages tend to M2 polarization, which promotes tumor growth and leads to adverse physiological effects. Due to the lack of highly specific antigens in M1 and M2 macrophages, significant challenges present in isolating these subtypes from clinical samples or in vitro coculture models of tumor-immune cells. In reverse, the single-cell technique provides the possibility to investigate the factors influencing macrophage polarization in the TME. In this research, we employed inertial microfluidic chip-mass spectrometry (IMC-MS) to conduct single-cell metabolomics analysis of macrophages polarized into the two major phenotypes, respectively, and 213 metabolites were identified in total. Subsequently, differential metabolites between macrophage phenotypes were analyzed using volcano plots and binary logistic regression models. Glutamine was pinpointed as a key metabolite for the M1 and M2 phenotypes. Experimental results from both monoculture and coculture cell models demonstrated that M1 polarization is more reliant on the presence of glutamine in the culture environment than M2 polarization. Glutamine deficiency resulted in failed M1 polarization, while its absence had a less pronounced effect on M2 polarization. Replenishing an appropriate amount of glutamine during the intermediate stages of coculture models significantly enhanced the proportion of M1 polarization and suppressed the growth of tumor cells. This research elucidated glutamine as a key factor influencing macrophage polarization in the TME via single-cell metabolomics based on IMC-MS, offering promising insights and targets for tumor therapies.
Collapse
Affiliation(s)
- Xuesen Hu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xinlin Liu
- The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Disheng Feng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Tianrun Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Hang Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Chunxiu Hu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Zhizhou Wang
- The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Peiyuan Yin
- The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xianzhe Shi
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Dong Shang
- The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
12
|
Xiong Z, Li Y, Zhang X, Zhang S, Li K, Zheng N, Zhao S, Wang J. Effects of biochanin A on lactational performance, nitrogen metabolism, and blood metabolites in dairy cows. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:441-449. [PMID: 39309972 PMCID: PMC11416632 DOI: 10.1016/j.aninu.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/24/2024] [Accepted: 05/06/2024] [Indexed: 09/25/2024]
Abstract
Optimizing nitrogen utilization efficiency and mitigating nitrogen losses in cows plays a pivotal role in fostering economic sustainability within contemporary agricultural systems. Biochanin A (BCA), a natural component in red clover, has the potential to improve nitrogen metabolism in dairy cows. The primary objective of this study was to probe the impact of biochanin A supplementation on lactational performance, nitrogen metabolism, and blood metabolites in dairy cows. A complete randomized block design experiment was conducted over 28 d, involving 36 multiparous Holstein cows (comparable milk yield = 37.1 ± 2.90 kg, BW = 642 ± 70.0 kg, days in milk = 92 ± 8.0 d, and parity = 2.4 ± 0.50), which were allocated to three treatment groups: the Control group (with 0 g/d BCA), the Low group (with 10 g/d per cow BCA), and the High group (with 40 g/d per cow BCA). Biochanin A supplementation improved the lactational performance of cows by increasing milk yield by 6.3% (P = 0.007) and feed efficiency by 12.7% (P = 0.009). Total intestinal apparent digestibility was unaffected by BCA supplementation (P > 0.05), but microbial nitrogen was increased by 30.0% (P = 0.002) for promoting nitrogen utilization efficiency by 20.7% (P = 0.004). Milk competent yields (protein, lactose, and non-fat milk solid) were increased with increasing BCA supplementation (P < 0.05). Urea nitrogen levels in plasma and milk were both decreased by BCA supplementation (P < 0.05). Blood routine parameters and plasma biochemical parameters both received no effect by BCA supplementation (P > 0.05). BCA did not affect body health of dairy cows. Additionally, none of the plasma endocrine hormones were affected (P > 0.05). A total of 95 significantly different metabolites were screened from the plasma metabolites of cows in the BCA-added and non-added groups. After performing an enrichment analysis of the metabolic pathways associated with the different metabolites, six specific pathways were identified: bile acid biosynthesis, aspartate metabolism, pyrimidine metabolism, arginine and proline metabolism, the urea cycle, and ammonia recycling. The inclusion of BCA is suggested to enhance milk yield and modulate nitrogen metabolism by influencing relevant metabolites within the metabolic pathways.
Collapse
Affiliation(s)
- Zhanbo Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanjun Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaoyin Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shiqi Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Kexin Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shengguo Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
13
|
Alberto-Silva C, da Silva BR, da Silva JCA, da Cunha e Silva FA, Kodama RT, da Silva WD, Costa MS, Portaro FCV. Small Structural Differences in Proline-Rich Decapeptides Have Specific Effects on Oxidative Stress-Induced Neurotoxicity and L-Arginine Generation by Arginosuccinate Synthase. Pharmaceuticals (Basel) 2024; 17:931. [PMID: 39065782 PMCID: PMC11279908 DOI: 10.3390/ph17070931] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION The proline-rich decapeptide 10c (Bj-PRO-10c; ENWPHPQIPP) from the Bothrops jararaca snake modulates argininosuccinate synthetase (AsS) activity to stimulate L-arginine metabolite production and neuroprotection in the SH-SY5Y cell line. The relationships between structure, interactions with AsS, and neuroprotection are little known. We evaluated the neuroprotective effects of Bj-PRO-10c and three other PROs (Bn-PRO-10a, METHODS Cell integrity, metabolic activity, reactive oxygen species (ROS) production, and arginase activity were examined after 4 h of PRO pre-treatment and 20 h of H2O2-induced damage. RESULTS Only Bn-PRO-10a-MK and Bn-PRO-10c restored cell integrity and arginase function under oxidative stress settings, but they did not reduce ROS or cell metabolism. The MK dipeptide in Bn-PRO-10a-MK and valine (V8) in Bn-PRO-10c are important to these effects when compared to Bn-PRO-10a. Bj-PRO-10c is not neuroprotective in PC12 cells, perhaps because of their limited NMDA-type glutamate receptor activity. The PROs interaction analysis on AsS activation can be rated as follows: Bj-PRO-10c > Bn-PRO-10c > Bn-PRO-10a-MK > Bn-PRO-10a. The structure of PROs and their correlations with enzyme activity revealed that histidine (H5) and glutamine (Q7) in Bj-PRO-10c potentiated their affinity for AsS. CONCLUSIONS Our investigation provides the first insights into the structure and molecular interactions of PROs with AsS, which could possibly further their neuropharmacological applications.
Collapse
Affiliation(s)
- Carlos Alberto-Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (B.R.d.S.); (J.C.A.d.S.); (F.A.d.C.e.S.)
| | - Brenda Rufino da Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (B.R.d.S.); (J.C.A.d.S.); (F.A.d.C.e.S.)
| | - Julio Cezar Araujo da Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (B.R.d.S.); (J.C.A.d.S.); (F.A.d.C.e.S.)
| | - Felipe Assumpção da Cunha e Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo 09606-070, SP, Brazil; (B.R.d.S.); (J.C.A.d.S.); (F.A.d.C.e.S.)
| | - Roberto Tadashi Kodama
- Structure and Functions of Biomolecules Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (R.T.K.); (F.C.V.P.)
| | - Wilmar Dias da Silva
- Laboratory of Immunochemistry, Butantan Institute, São Paulo 05503-900, SP, Brazil;
| | - Maricilia Silva Costa
- Instituto de Pesquisa & Desenvolvimento—IP&D, Universidade do Vale do Paraíba—UNIVAP, Av. Shishima Hifumi, 2911, São José dos Campos 12244-390, SP, Brazil;
| | - Fernanda Calheta Vieira Portaro
- Structure and Functions of Biomolecules Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (R.T.K.); (F.C.V.P.)
| |
Collapse
|
14
|
Thakkar N, Gajera G, Mehta D, Kothari V. Silversol ® (a Colloidal Nanosilver Formulation) Inhibits Growth of Antibiotic-Resistant Staphylococcus aureus by Disrupting Its Physiology in Multiple Ways. Pharmaceutics 2024; 16:726. [PMID: 38931848 PMCID: PMC11206351 DOI: 10.3390/pharmaceutics16060726] [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: 05/02/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Antibiotic-resistant strains of Staphylococcus aureus are being viewed as a serious threat by various public health agencies. Identifying novel targets in this important pathogen is crucial to the development of new effective antibacterial formulations. We investigated the antibacterial effect of a colloidal nanosilver formulation, Silversol®, against an antibiotic-resistant strain of S. aureus using appropriate in vitro assays. Moreover, we deciphered the molecular mechanisms underlying this formulation's anti-S. aureus activity using whole transcriptome analysis. Lower concentrations of the test formulation exerted a bacteriostatic effect against this pathogen, and higher concentrations exerted a bactericidal effect. Silversol® at sub-lethal concentration was found to disturb multiple physiological traits of S. aureus such as growth, antibiotic susceptibility, membrane permeability, efflux, protein synthesis and export, biofilm and exopolysaccharide production, etc. Transcriptome data revealed that the genes coding for transcriptional regulators, efflux machinery, transferases, β-lactam resistance, oxidoreductases, metal homeostasis, virulence factors, and arginine biosynthesis are expressed differently under the influence of the test formulation. Genes (argG and argH) involved in arginine biosynthesis emerged among the major targets of Silversol®'s antibacterial activity against S. aureus.
Collapse
Affiliation(s)
- Nidhi Thakkar
- Institute of Science, Nirma University, Ahmedabad 382481, India; (N.T.); (G.G.)
| | - Gemini Gajera
- Institute of Science, Nirma University, Ahmedabad 382481, India; (N.T.); (G.G.)
| | - Dilip Mehta
- Viridis BioPharma Pvt. Ltd., Mumbai 400043, India;
| | - Vijay Kothari
- Institute of Science, Nirma University, Ahmedabad 382481, India; (N.T.); (G.G.)
| |
Collapse
|
15
|
Cheng Q, Han Y, Xiao Y, Li Z, Qin A, Ji S, Kan B, Liang W. The ArgR-Regulated ADI Pathway Facilitates the Survival of Vibrio fluvialis under Acidic Conditions. Int J Mol Sci 2024; 25:5679. [PMID: 38891866 PMCID: PMC11172107 DOI: 10.3390/ijms25115679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Vibrio fluvialis is an emerging foodborne pathogenic bacterium that can cause severe cholera-like diarrhea and various extraintestinal infections, posing challenges to public health and food safety worldwide. The arginine deiminase (ADI) pathway plays an important role in bacterial environmental adaptation and pathogenicity. However, the biological functions and regulatory mechanisms of the pathway in V. fluvialis remain unclear. In this study, we demonstrate that L-arginine upregulates the expression of the ADI gene cluster and promotes the growth of V. fluvialis. The ADI gene cluster, which we proved to be comprised of two operons, arcD and arcACB, significantly enhances the survival of V. fluvialis in acidic environments both in vitro (in culture medium and in macrophage) and in vivo (in mice). The mRNA level and reporter gene fusion analyses revealed that ArgR, a transcriptional factor, is necessary for the activation of both arcD and arcACB transcriptions. Bioinformatic analysis predicted the existence of multiple potential ArgR binding sites at the arcD and arcACB promoter regions that were further confirmed by electrophoretic mobility shift assay, DNase I footprinting, or point mutation analyses. Together, our study provides insights into the important role of the ArgR-ADI pathway in the survival of V. fluvialis under acidic conditions and the detailed molecular mechanism. These findings will deepen our understanding of how environmental changes and gene expression interact to facilitate bacterial adaptations and virulence.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Biao Kan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Weili Liang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
16
|
Willmann K, Moita LF. Physiologic disruption and metabolic reprogramming in infection and sepsis. Cell Metab 2024; 36:927-946. [PMID: 38513649 DOI: 10.1016/j.cmet.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024]
Abstract
Effective responses against severe systemic infection require coordination between two complementary defense strategies that minimize the negative impact of infection on the host: resistance, aimed at pathogen elimination, and disease tolerance, which limits tissue damage and preserves organ function. Resistance and disease tolerance mostly rely on divergent metabolic programs that may not operate simultaneously in time and space. Due to evolutionary reasons, the host initially prioritizes the elimination of the pathogen, leading to dominant resistance mechanisms at the potential expense of disease tolerance, which can contribute to organ failure. Here, we summarize our current understanding of the role of physiological perturbations resulting from infection in immune response dynamics and the metabolic program requirements associated with resistance and disease tolerance mechanisms. We then discuss how insight into the interplay of these mechanisms could inform future research aimed at improving sepsis outcomes and the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Katharina Willmann
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Luis F Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal; Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
17
|
Shen Y, Wang H, Xie H, Zhang J, Ma Q, Wang S, Yuan P, Xue H, Hong H, Fan S, Xu W, Xie Z. l-arginine promotes angio-osteogenesis to enhance oxidative stress-inhibited bone formation by ameliorating mitophagy. J Orthop Translat 2024; 46:53-64. [PMID: 38808262 PMCID: PMC11131000 DOI: 10.1016/j.jot.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/03/2024] [Accepted: 03/04/2024] [Indexed: 05/30/2024] Open
Abstract
Background Osteoporosis is one of the most common bone diseases in middle-aged and elderly populations worldwide. The development of new drugs to treat the disease is a key focus of research. Current treatments for osteoporosis are mainly directed at promoting osteoblasts and inhibiting osteoclasts. However, there is currently no ideal approach for osteoporosis treatment. l-arginine is a semi-essential amino acid involved in a number of cellular processes, including nitric production, protein biosynthesis, and immune responses. We previously reported that l-arginine-derived compounds can play a regulatory role in bone homeostasis. Purpose To investigate the specific effect of l-arginine on bone homeostasis. Methods Mildly aged and ovariectomized mouse models were used to study the effects of l-arginine on osteogenesis and angiogenesis, assessed by micro-computed tomography and immunostaining of bone tissue. The effect of l-arginine on osteogenesis, angiogenesis, and adipogenesis was further studied in vitro using osteoblasts obtained from cranial cap bone, endothelial cells, and an adipogenic cell line. Specific methods to assess these processes included lipid staining, cell migration, tube-forming, and wound-healing assays. Protein and mRNA expression was determined for select biomarkers. Results We found that l-arginine attenuated bone loss and promoted osteogenesis and angiogenesis. l-arginine increased the activity of vascular endothelial cells, whereas it inhibited adipogenesis in vitro. In addition, we found that l-arginine altered the expression of PINK1/Parkin and Bnip3 in the mitochondria of osteoblast-lineage and endothelial cells, thereby promoting mitophagy and protecting cells from ROS. Similarly, l-arginine treatment effectively ameliorated osteoporosis in an ovariectomized mouse model. Conclusion l-arginine promotes angio-osteogenesis, and inhibits adipogenesis, effects mediated by the PINK1/Parkin- and Bnip3-mediated mitophagy. The Translational Potential of this Article L-arginine supplementation may be an effective adjunct therapy in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Yang Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Haoming Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hongwei Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jiateng Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Qingliang Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shiyu Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Putao Yuan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hong Xue
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Huaxing Hong
- Department of Orthopaedics, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Wenbin Xu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Ziang Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
18
|
Liu X, Li J, Zhang Z, He Y, Wang M, Zhao Y, Lin S, Liu T, Liao Y, Zhang N, Yuan K, Ling Y, Liu Z, Chen X, Chen Z, Chen R, Wang X, Gu B. Acetylation of xenogeneic silencer H-NS regulates biofilm development through the nitrogen homeostasis regulator in Shewanella. Nucleic Acids Res 2024; 52:2886-2903. [PMID: 38142446 PMCID: PMC11014242 DOI: 10.1093/nar/gkad1219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/26/2023] Open
Abstract
Adjusting intracellular metabolic pathways and adopting suitable live state such as biofilms, are crucial for bacteria to survive environmental changes. Although substantial progress has been made in understanding how the histone-like nucleoid-structuring (H-NS) protein modulates the expression of the genes involved in biofilm formation, the precise modification that the H-NS protein undergoes to alter its DNA binding activity is still largely uncharacterized. This study revealed that acetylation of H-NS at Lys19 inhibits biofilm development in Shewanella oneidensis MR-1 by downregulating the expression of glutamine synthetase, a critical enzyme in glutamine synthesis. We further found that nitrogen starvation, a likely condition in biofilm development, induces deacetylation of H-NS and the trimerization of nitrogen assimilation regulator GlnB. The acetylated H-NS strain exhibits significantly lower cellular glutamine concentration, emphasizing the requirement of H-NS deacetylation in Shewanella biofilm development. Moreover, we discovered in vivo that the activation of glutamine biosynthesis pathway and the concurrent suppression of the arginine synthesis pathway during both pellicle and attached biofilms development, further suggesting the importance of fine tune nitrogen assimilation by H-NS acetylation in Shewanella. In summary, posttranslational modification of H-NS endows Shewanella with the ability to respond to environmental needs by adjusting the intracellular metabolism pathways.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
| | - Jun Li
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Zhixuan Zhang
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510080, China
| | - Yizhou He
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
| | - Mingfang Wang
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Yunhu Zhao
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Shituan Lin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianlang Liu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiwen Liao
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Ni Zhang
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Kaixuan Yuan
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Yong Ling
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Ziyao Liu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaozhong Chen
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| | - Zhe Chen
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ran Chen
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
| | - Xiaoxue Wang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), No. 1119, Haibin Road, Nansha District, Guangzhou 511458, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Gu
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, China
| |
Collapse
|
19
|
Peyvandi S, Lan Q, Chabloz A, Prével F, La Torre YH, Ives A, Tacchini-Cottier F. The β-Carboline Harmine Has a Protective Immunomodulatory Role in Nonhealing Cutaneous Leishmaniasis. J Invest Dermatol 2024; 144:862-873.e4. [PMID: 37852357 DOI: 10.1016/j.jid.2023.09.280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
Cutaneous leishmaniasis affects 1 million people worldwide annually. Although conventional treatments primarily target the parasite, there is growing interest in host immune modulation. In this study, we investigated the impact of synthetic β-carboline harmine (ACB1801), previously shown to be immunoregulatory in cancer, on the pathology caused by a drug-resistant Leishmania major strain causing persistent cutaneous lesions. Exposure to ACB1801 in vitro had a modest impact on parasite burden within host macrophages. Moreover, it significantly increased major histocompatibility complex II and costimulatory molecule expression on infected dendritic cells, suggesting an enhanced immune response. In vivo, ACB1801 monotherapy led to a substantial reduction in lesion development and parasite burden in infected C57BL/6 mice, comparable with efficacy of amphotericin B. Transcriptomics analysis further supported ACB1801 immunomodulatory effects, revealing an enrichment of TNF-α, IFN-γ, and major histocompatibility complex II antigen presentation signatures in the draining lymph nodes of treated mice. Flow cytometry analysis confirmed an increased frequency (1.5×) of protective CD4+IFN-γ+TNF-α+ T cells and a decreased frequency (2×) in suppressive IL-10+FoxP3- T cells at the site of infection and in draining lymph nodes. In addition, ACB1801 downregulated the aryl hydrocarbon receptor signaling, known to enhance immunosuppressive cytokines. Thus, these results suggest a potential use for ACB1801 alone or in combination therapy for cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Sanam Peyvandi
- Department of Immunobiology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Qiang Lan
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | | | - Florence Prével
- Department of Immunobiology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | - Yazmin Hauyon La Torre
- Department of Immunobiology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland
| | | | - Fabienne Tacchini-Cottier
- Department of Immunobiology, Faculty of Biology and Medicine, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
20
|
Alberto-Silva C, Pantaleão HQ, da Silva BR, da Silva JCA, Echeverry MB. Activation of M1 muscarinic acetylcholine receptors by proline-rich oligopeptide 7a (<EDGPIPP) from Bothrops jararaca snake venom rescues oxidative stress-induced neurotoxicity in PC12 cells. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20230043. [PMID: 38362565 PMCID: PMC10868729 DOI: 10.1590/1678-9199-jvatitd-2023-0043] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 12/22/2023] [Indexed: 02/17/2024] Open
Abstract
Background The bioactive peptides derived from snake venoms of the Viperidae family species have been promising as therapeutic candidates for neuroprotection due to their ability to prevent neuronal cell loss, injury, and death. Therefore, this study aimed to evaluate the cytoprotective effects of a synthetic proline-rich oligopeptide 7a (PRO-7a; Methods Both cells were pre-treated for four hours with different concentrations of PRO-7a, submitted to H2O2-induced damage for 20 h, and then the oxidative stress markers were analyzed. Also, two independent neuroprotective mechanisms were investigated: a) L-arginine metabolite generation via argininosuccinate synthetase (AsS) activity regulation to produce agmatine or polyamines with neuroprotective properties; b) M1 mAChR receptor subtype activation pathway to reduce oxidative stress and neuron injury. Results PRO-7a was not cytoprotective in C6 cells, but potentiated the H2O2-induced damage to cell integrity at a concentration lower than 0.38 μM. However, PRO-7a at 1.56 µM, on the other hand, modified H2O2-induced toxicity in PC12 cells by restoring cell integrity, mitochondrial metabolism, ROS generation, and arginase indirect activity. The α-Methyl-DL-aspartic acid (MDLA) and L-NΩ-Nitroarginine methyl ester (L-Name), specific inhibitors of AsS and nitric oxide synthase (NOS), which catalyzes the synthesis of polyamines and NO from L-arginine, did not suppress PRO-7a-mediated cytoprotection against oxidative stress. It suggested that its mechanism is independent of the production of L-arginine metabolites with neuroprotective properties by increased AsS activity. On the other hand, the neuroprotective effect of PRO-7a was blocked in the presence of dicyclomine hydrochloride (DCH), an M1 mAChR antagonist. Conclusions For the first time, this work provides evidence that PRO-7a-induced neuroprotection seems to be mediated through M1 mAChR activation in PC12 cells, which reduces oxidative stress independently of AsS activity and L-arginine bioavailability.
Collapse
Affiliation(s)
- Carlos Alberto-Silva
- Natural and Humanities Sciences Center (CCNH), Experimental
Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do
Campo, SP, Brazil
| | - Halyne Queiroz Pantaleão
- Natural and Humanities Sciences Center (CCNH), Experimental
Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do
Campo, SP, Brazil
| | - Brenda Rufino da Silva
- Natural and Humanities Sciences Center (CCNH), Experimental
Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do
Campo, SP, Brazil
| | - Julio Cezar Araujo da Silva
- Natural and Humanities Sciences Center (CCNH), Experimental
Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do
Campo, SP, Brazil
| | - Marcela Bermudez Echeverry
- Center for Mathematics, Computation and Cognition (CMCC), Federal
University of ABC, São Bernardo do Campo, SP, Brazil
| |
Collapse
|
21
|
Yusof NY, Quay DHX, Kamaruddin S, Jonet MA, Md Illias R, Mahadi NM, Firdaus-Raih M, Abu Bakar FD, Abdul Murad AM. Biochemical and in silico structural characterization of a cold-active arginase from the psychrophilic yeast, Glaciozyma antarctica PI12. Extremophiles 2024; 28:15. [PMID: 38300354 DOI: 10.1007/s00792-024-01333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 12/30/2023] [Indexed: 02/02/2024]
Abstract
Glaciozyma antarctica PI12 is a psychrophilic yeast isolated from Antarctica. In this work, we describe the heterologous production, biochemical properties and in silico structure analysis of an arginase from this yeast (GaArg). GaArg is a metalloenzyme that catalyses the hydrolysis of L-arginine to L-ornithine and urea. The cDNA of GaArg was reversed transcribed, cloned, expressed and purified as a recombinant protein in Escherichia coli. The purified protein was active against L-arginine as its substrate in a reaction at 20 °C, pH 9. At 10-35 °C and pH 7-9, the catalytic activity of the protein was still present around 50%. Mn2+, Ni2+, Co2+ and K+ were able to enhance the enzyme activity more than two-fold, while GaArg is most sensitive to SDS, EDTA and DTT. The predicted structure model of GaArg showed a very similar overall fold with other known arginases. GaArg possesses predominantly smaller and uncharged amino acids, fewer salt bridges, hydrogen bonds and hydrophobic interactions compared to the other counterparts. GaArg is the first reported arginase that is cold-active, facilitated by unique structural characteristics for its adaptation of catalytic functions at low-temperature environments. The structure and function of cold-active GaArg provide insights into the potentiality of new applications in various biotechnology and pharmaceutical industries.
Collapse
Affiliation(s)
- Nik Yusnoraini Yusof
- Institute for Research in Molecular Medicine (INFORMM), Health Campus, Universiti Sains Malaysia, Kubang Kerian, 16150, Kelantan, Malaysia.
- Department of Biological Sciences & Biotechnology, Faculty of Sciences and Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia.
| | - Doris Huai Xia Quay
- Department of Applied Physics, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, UKM, 43600, Bangi, Selangor, Malaysia.
| | - Shazilah Kamaruddin
- Department of Biological Sciences & Biotechnology, Faculty of Sciences and Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| | - Mohd Anuar Jonet
- Malaysia Genome and Vaccine Institute, Jalan Bangi Lama, 43000, Kajang, Selangor, Malaysia
| | - Rosli Md Illias
- Department of Bioprocess Engineering, Faculty of Chemical Engineering, Universiti Teknologi Malaysia, 81300, Skudai, Johor, Malaysia
| | - Nor Muhammad Mahadi
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| | - Mohd Firdaus-Raih
- Department of Applied Physics, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, UKM, 43600, Bangi, Selangor, Malaysia
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| | - Farah Diba Abu Bakar
- Department of Biological Sciences & Biotechnology, Faculty of Sciences and Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| | - Abdul Munir Abdul Murad
- Department of Biological Sciences & Biotechnology, Faculty of Sciences and Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| |
Collapse
|
22
|
Kausar MA, Narayan J, Agarwal P, Singh P, Ahmed RME, El-Hag ABM, Khalifa AM, Mohammed NARK, Singh R, Mahfooz S. Distribution and conservation of simple sequence repeats in plant pathogenic species of Zymoseptoria and development of genomic resources for its orphaned species. Antonie Van Leeuwenhoek 2024; 117:11. [PMID: 38170404 DOI: 10.1007/s10482-023-01915-z] [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: 10/06/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
To better understand the structure and evolution of the genomes of four plant pathogenic species of Zymoseptoria, we analyzed the occurrence, relative abundance (RA), and density (RD) of simple sequence repeats (SSRs) in their whole genome and transcriptome sequences. In this study, SSRs are defined as repeats of more than 12 bases in length. The genome and transcriptome sequences of Zymoseptoria ardabiliae show the highest RA (201.1 and 129.9) and RD (3229.4 and 1928.2) of SSRs, while those of Zymoseptoria pseudotritici show the lowest RA (167.2 and 118.5) and RD (2482.2 and 1687.0). The majority of SSRs in the genomic and transcriptome sequences of species were trinucleotide SSRs, while dinucleotide SSRs were the least common. The most common trinucleotide motifs in the transcriptomic sequences across all species were those that encoded the amino acid arginine. As per our motif conservation study, Zymoseptoria tritici (12.4%) possessed the most unique motifs, while Z. pseudotritici (3.9%) had the fewest. Overall, only 38.1% of the motifs were found to be conserved among the species. Gene enrichment studies reveal that three of the species, Z. ardabiliae, Zymoseptoria brevis, and Z. pseudotritici, have SSRs in their genes related to cellular metabolism, while the remaining Z. tritici harbors SSRs in genes related to DNA synthesis and gene expression. In an effort to improve the genetic resources for the orphan species of pathogenic Zymoseptoria, a total of 73,134 primers were created. The genomic resources developed in this study could help with analyses of genetic relatedness within the population and the development of species-specific markers.
Collapse
Affiliation(s)
- Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Hail, 2440, Hail, Saudi Arabia.
| | - Jitendra Narayan
- CSIR- Institute of Genomics and Integrative Biology, Mall Road, New Delhi, 110007, India
| | - Preeti Agarwal
- CSIR- Institute of Genomics and Integrative Biology, Mall Road, New Delhi, 110007, India
| | - Pallavi Singh
- Department of Biotechnology, Dr APJ Abdul Kalam Technical University, Lucknow, 226031, India
| | | | | | - Amany Mohammed Khalifa
- Department of Pathology, College of Medicine, University of Hail, 2440, Hail, Saudi Arabia
| | | | - Rajeev Singh
- Department of Environmental Science, Jamia Millia Islamia Central University, 110025, New Delhi, India
| | - Sahil Mahfooz
- The Academic Editors, Saryu Enclave, Awadh Vikas Yojna, Lucknow, 226002, India.
- Department of Industrial Microbiology, Deen Dayal Upadhyaya Gorakhpur University, Gorakhpur, 273009, India.
| |
Collapse
|
23
|
Kim K, Jinno C, Li X, Bravo D, Cox E, Ji P, Liu Y. Impact of an oligosaccharide-based polymer on the metabolic profiles and microbial ecology of weanling pigs experimentally infected with a pathogenic E. coli. J Anim Sci Biotechnol 2024; 15:1. [PMID: 38169416 PMCID: PMC10759389 DOI: 10.1186/s40104-023-00956-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/29/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Our previous study has reported that supplementation of oligosaccharide-based polymer enhances gut health and disease resistance of pigs infected with enterotoxigenic E. coli (ETEC) F18 in a manner similar to carbadox. The objective of this study was to investigate the impacts of oligosaccharide-based polymer or antibiotic on the host metabolic profiles and colon microbiota of weaned pigs experimentally infected with ETEC F18. RESULTS Multivariate analysis highlighted the differences in the metabolic profiles of serum and colon digesta which were predominantly found between pigs supplemented with oligosaccharide-based polymer and antibiotic. The relative abundance of metabolic markers of immune responses and nutrient metabolisms, such as amino acids and carbohydrates, were significantly differentiated between the oligosaccharide-based polymer and antibiotic groups (q < 0.2 and fold change > 2.0). In addition, pigs in antibiotic had a reduced (P < 0.05) relative abundance of Lachnospiraceae and Lactobacillaceae, whereas had greater (P < 0.05) Clostridiaceae and Streptococcaceae in the colon digesta on d 11 post-inoculation (PI) compared with d 5 PI. CONCLUSIONS The impact of oligosaccharide-based polymer on the metabolic and microbial profiles of pigs is not fully understood, and further exploration is needed. However, current research suggest that various mechanisms are involved in the enhanced disease resistance and performance in ETEC-challenged pigs by supplementing this polymer.
Collapse
Affiliation(s)
- Kwangwook Kim
- Department of Animal Science, University of California, Davis, CA, 95616, USA
- Present Affiliation: Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Cynthia Jinno
- Department of Animal Science, University of California, Davis, CA, 95616, USA
- Present Affiliation: Cedars-Sinai Medical Center, Los Angeles, CA, 90084, USA
| | - Xunde Li
- School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - David Bravo
- Pancosma|ADM, 1180, Rolle, Switzerland
- Present Affiliation: Nutreco Exploration, Nutreco, The Netherlands
| | - Eric Cox
- Department of Virology, Parasitology and Immunology, Ghent University, 9000, Ghent, Belgium
| | - Peng Ji
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
24
|
Kim DY, Park JY, Gee HY. Lactobacillus plantarum ameliorates NASH-related inflammation by upregulating L-arginine production. Exp Mol Med 2023; 55:2332-2345. [PMID: 37907736 PMCID: PMC10689779 DOI: 10.1038/s12276-023-01102-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/16/2023] [Accepted: 08/02/2023] [Indexed: 11/02/2023] Open
Abstract
Lactobacillus is a probiotic with therapeutic potential for several diseases, including liver disease. However, the therapeutic effect of L. plantarum against nonalcoholic steatohepatitis (NASH) and its underlying mechanisms remain unelucidated. Therefore, we delineated the L. plantarum-mediated NASH regulation in a mouse model to understand its therapeutic effect. We used a choline-deficient high-fat diet (CD-HFD)-induced murine model that recapitulated the critical features of human metabolic syndrome and investigated the effect of L. plantarum on NASH pathogenesis using transcriptomic, metagenomic, and immunohistochemistry analyses. Validation experiments were performed using liver organoids and a murine model fed a methionine-choline-deficient (MCD) diet. L. plantarum treatment in mice significantly decreased liver inflammation and improved metabolic phenotypes, such as insulin tolerance and the hepatic lipid content, compared with those in the vehicle group. RNA-sequencing analysis revealed that L. plantarum treatment significantly downregulated inflammation-related pathways. Shotgun metagenomic analysis revealed that L-arginine biosynthesis-related microbial genes were significantly upregulated in the L. plantarum group. We also confirmed the elevated arginine levels in the serum of the L. plantarum group. We further used liver organoids and mice fed an MCD diet to demonstrate that L-arginine alone was sufficient to alleviate liver inflammation. Our data revealed a novel and counterintuitive therapeutic effect of L. plantarum on alleviating NASH-related liver inflammation by increasing circulating L-arginine.
Collapse
Affiliation(s)
- Dong Yun Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of South Korea
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of South Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of South Korea
| | - Jun Yong Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of South Korea.
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of South Korea.
| | - Heon Yung Gee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of South Korea.
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of South Korea.
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Republic of South Korea.
| |
Collapse
|
25
|
Sarkar D, Sau AK. Illuminating the structure-function landscape of an evolutionary nonconserved motif in the arginases of Helicobacter gastric pathogens. IUBMB Life 2023; 75:782-793. [PMID: 37086465 DOI: 10.1002/iub.2728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/03/2023] [Indexed: 04/24/2023]
Abstract
The bimetallic enzyme arginase catalyses the conversion of L-arginine to L-ornithine and urea. In Helicobacter pylori (a known human gastric pathogen), this enzyme is an important virulence factor. In spite of the conservation of the catalytic and the metal-binding residues, the H. pylori homolog possesses a 13-residue motif (-153 ESEEKAWQKLCSL165 -) present in the middle of the protein sequence, whose role was recently elucidated. Despite several reviews available on arginases, no report has thoroughly illustrated the underlying basis for the importance of the above motif of the H. pylori enzyme in structure and function. In this review, we systematically describe a mechanistic basis for its importance in structure and function based on the known data. This motif of the H. pylori enzyme is present exclusively in the arginases of other Helicobacter gastric pathogens, where the critical residues are conserved, implying that the nonconserved stretch has been selected during the evolution of the enzyme in these gastric pathogens in a specific manner to perform its role in the structure and function. The combined information can be useful for understanding the function of arginases in other Helicobacter gastric pathogens. Additionally, this knowledge can be utilised to screen and design new small molecule inhibitors, specific to the arginases of these pathogens.
Collapse
Affiliation(s)
- Ditsa Sarkar
- Protein Engineering Laboratory, National Institute of Immunology, New Delhi, Delhi, India
| | - Apurba Kumar Sau
- Protein Engineering Laboratory, National Institute of Immunology, New Delhi, Delhi, India
| |
Collapse
|
26
|
Oljuskin T, Azodi N, Volpedo G, Bhattacharya P, Markle HL, Hamano S, Matlashewski G, Satoskar AR, Gannavaram S, Nakhasi HL. Leishmania major centrin knock-out parasites reprogram tryptophan metabolism to induce a pro-inflammatory response. iScience 2023; 26:107593. [PMID: 37744403 PMCID: PMC10517402 DOI: 10.1016/j.isci.2023.107593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 07/07/2023] [Accepted: 08/07/2023] [Indexed: 09/26/2023] Open
Abstract
Leishmaniasis is a parasitic disease that is prevalent in 90 countries, and yet no licensed human vaccine exists against it. Toward control of leishmaniasis, we have developed Leishmania major centrin gene deletion mutant strains (LmCen-/-) as a live attenuated vaccine, which induces a strong IFN-γ-mediated protection to the host. However, the immune mechanisms of such protection remain to be understood. Metabolomic reprogramming of the host cells following Leishmania infection has been shown to play a critical role in pathogenicity and shaping the immune response following infection. Here, we applied untargeted mass spectrometric analysis to study the metabolic changes induced by infection with LmCen-/- and compared those with virulent L. major parasite infection to identify the immune mechanism of protection. Our data show that immunization with LmCen-/- parasites, in contrast to virulent L. major infection promotes a pro-inflammatory response by utilizing tryptophan to produce melatonin and downregulate anti-inflammatory kynurenine-AhR and FICZ-AhR signaling.
Collapse
Affiliation(s)
- Timur Oljuskin
- Animal Parasitic Diseases Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
| | - Nazli Azodi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| | - Greta Volpedo
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| | - Hannah L. Markle
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| | - Shinjiro Hamano
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), The Joint Usage/Research Center on Tropical Disease, Nagasaki University, Nagasaki, Japan
- Nagasaki University Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki, Japan
| | - Greg Matlashewski
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Abhay R. Satoskar
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| | - Hira L. Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| |
Collapse
|
27
|
Liu A, Zhang L, Zhou A, Yang F, Yue Z, Wang J. Metabolomic and physiological changes of acid-tolerant Graesiella sp. MA1 during long-term acid stress. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:97209-97218. [PMID: 37589846 DOI: 10.1007/s11356-023-29295-x] [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: 03/02/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023]
Abstract
Algae plays a significant role for the primary production in the oligotrophic ecosystems such as the acid mine pit lakes. Graesiella sp. MA1 was a new acid-tolerant photosynthetic protist isolated from an acid mine pit lake. To understand the acid responses of Graesiella sp. MA1, its physiological changes and metabolomics were studied during long-term acid stress. Photosynthetic pigments, soluble proteins, and antioxidant systems of Graesiella sp. MA1 cells displayed two phases, the adaptation phase and the growth phase. During the adaptation phase, both photosynthetic pigments and soluble proteins were inhibited, while antioxidant activity of SOD, APX, and GSH were promoted to response to the organism's damage. Metabolomics results revealed lipids and organic acids were abundant components in Graesiella sp. MA1 cells. In response to acid stress, the levels of acid-dependent resistant amino acids, including glutamate, aspartate, arginine, proline, lysine, and histidine, accumulated continuously to maintain orderly intracellular metabolic processes. In addition, fatty acids were mainly unsaturated, which could improve the fluidity of the cell membranes under acid stress. Metabolomic and physiological changes showed that Graesiella sp. MA1 had tolerance during long-term acid stress and the potential to be used as a bioremediation strain for the acidic wastewater.
Collapse
Affiliation(s)
- Azuan Liu
- School of Resources and Environmental Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
- Anhui Engineering Research Center of Industrial Wastewater Treatment and Resource Recovery, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Lu Zhang
- School of Resources and Environmental Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
- Anhui Engineering Research Center of Industrial Wastewater Treatment and Resource Recovery, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Ao Zhou
- School of Resources and Environmental Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
- Anhui Engineering Research Center of Industrial Wastewater Treatment and Resource Recovery, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Fan Yang
- School of Resources and Environmental Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
- Anhui Engineering Research Center of Industrial Wastewater Treatment and Resource Recovery, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Zhengbo Yue
- School of Resources and Environmental Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
- Anhui Engineering Research Center of Industrial Wastewater Treatment and Resource Recovery, Hefei University of Technology, Hefei, 230009, Anhui, China
- Key Laboratory of Nanominerals and Pollution Control of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Jin Wang
- School of Resources and Environmental Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China.
- Anhui Engineering Research Center of Industrial Wastewater Treatment and Resource Recovery, Hefei University of Technology, Hefei, 230009, Anhui, China.
- Key Laboratory of Nanominerals and Pollution Control of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230009, Anhui, China.
| |
Collapse
|
28
|
Pantaleão HQ, Araujo da Silva JC, Rufino da Silva B, Echeverry MB, Alberto-Silva C. Peptide fraction from B. jararaca snake venom protects against oxidative stress-induced changes in neuronal PC12 cell but not in astrocyte-like C6 cell. Toxicon 2023; 231:107178. [PMID: 37302421 DOI: 10.1016/j.toxicon.2023.107178] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/24/2023] [Accepted: 06/04/2023] [Indexed: 06/13/2023]
Abstract
Venom-derived proteins and peptides have prevented neuronal cell loss, damage, and death in the study of neurodegenerative disorders. The cytoprotective effects of the peptide fraction (PF) from Bothrops jararaca snake venom were evaluated against oxidative stress changes in neuronal PC12 cells and astrocyte-like C6 cells. PC12 and C6 cells were pre-treated for 4 h with different concentrations of PF, and then H2O2 was added (0.5 mM in PC12 cells; 0.4 mM in C6 cells) and incubated for 20 h more. In PC12 cells, PF at 0.78 μg mL-1 increased viability (113.6 ± 6.3%) and metabolism (96.3 ± 10.3%) cell against H2O2-induced neurotoxicity (75.6 ± 5.8%; 66.5 ± 3.3%, respectively), reducing oxidative stress markers such as ROS generation, NO production, and arginase indirect activity through urea synthesis. Despite that, PF showed no cytoprotective effects in C6 cells, but potentiated the H2O2-induced damage at a concentration lower than 0.07 μg mL-1. Furthermore, the role of metabolites derived from L-arginine metabolism was verified in PF-mediated neuroprotection in PC12 cells, using specific inhibitors of two of the key enzymes in the L-arginine metabolic pathway: the α-Methyl-DL-aspartic acid (MDLA) to argininosuccinate synthetase (AsS), responsible for the recycling of L-citrulline to L-arginine; and, L-NΩ-Nitroarginine methyl ester (L-Name) to nitric oxide synthase (NOS), which catalyzes the synthesis of NO from L-arginine. The inhibition of AsS and NOS suppressed PF-mediated cytoprotection against oxidative stress, indicating that its mechanism is dependent on the production pathway of L-arginine metabolites such as NO and, more importantly, polyamines from ornithine metabolism, which are involved in the neuroprotection mechanism described in the literature. Overall, this work provides novel opportunities for evaluating whether the neuroprotective properties of PF shown in particular neuronal cells are sustained and for exploring potential drug development pathways for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Halyne Queiroz Pantaleão
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory Federal University of ABC (UFABC), São Bernardo Do Campo, 09606-070, SP, Brazil
| | - Julio Cezar Araujo da Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory Federal University of ABC (UFABC), São Bernardo Do Campo, 09606-070, SP, Brazil
| | - Brenda Rufino da Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory Federal University of ABC (UFABC), São Bernardo Do Campo, 09606-070, SP, Brazil
| | - Marcela Bermudez Echeverry
- Center for Mathematics, Computation and Cognition (CMCC), UFABC, São Bernardo Do Campo, 09606-070, SP, Brazil
| | - Carlos Alberto-Silva
- Natural and Humanities Sciences Center (CCNH), Experimental Morphophysiology Laboratory Federal University of ABC (UFABC), São Bernardo Do Campo, 09606-070, SP, Brazil.
| |
Collapse
|
29
|
Brigo N, Neumaier E, Pfeifhofer-Obermair C, Grubwieser P, Engl S, Berger S, Seifert M, Reinstadler V, Oberacher H, Weiss G. Timing of Interleukin-4 Stimulation of Macrophages Determines Their Anti-Microbial Activity during Infection with Salmonella enterica Serovar Typhimurium. Cells 2023; 12:1164. [PMID: 37190073 PMCID: PMC10137269 DOI: 10.3390/cells12081164] [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: 12/23/2022] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Priming of macrophages with interferon-gamma (IFNγ) or interleukin-4 (IL-4) leads to polarisation into pro-inflammatory or anti-inflammatory subtypes, which produce key enzymes such as inducible nitric oxide synthase (iNOS) and arginase 1 (ARG1), respectively, and in this way determine host responses to infection. Importantly, L-arginine is the substrate for both enzymes. ARG1 upregulation is associated with increased pathogen load in different infection models. However, while differentiation of macrophages with IL-4 impairs host resistance to the intracellular bacterium Salmonella enterica serovar Typhimurium (S.tm), little is known on the effects of IL-4 on unpolarised macrophages during infection. Therefore, bone-marrow-derived macrophages (BMDM) from C57BL/6N, Tie2Cre+/-ARG1fl/fl (KO), Tie2Cre-/-ARG1fl/fl (WT) mice were infected with S.tm in the undifferentiated state and then stimulated with IL-4 or IFNγ. In addition, BMDM of C57BL/6N mice were first polarised upon stimulation with IL-4 or IFNγ and then infected with S.tm. Interestingly, in contrast to polarisation of BMDM with IL-4 prior to infection, treatment of non-polarised S.tm-infected BMDM with IL-4 resulted in improved infection control whereas stimulation with IFNγ led to an increase in intracellular bacterial numbers compared to unstimulated controls. This effect of IL-4 was paralleled by decreased ARG1 levels and increased iNOS expression. Furthermore, the L-arginine pathway metabolites ornithine and polyamines were enriched in unpolarised cells infected with S.tm and stimulated with IL-4. Depletion of L-arginine reversed the protective effect of IL-4 toward infection control. Our data show that stimulation of S.tm-infected macrophages with IL-4 reduced bacterial multiplication via metabolic re-programming of L-arginine-dependent pathways.
Collapse
Affiliation(s)
- Natascha Brigo
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Emely Neumaier
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Philipp Grubwieser
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Sabine Engl
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Sylvia Berger
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Vera Reinstadler
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, Muellerstrasse 44, 6020 Innsbruck, Austria
| | - Herbert Oberacher
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, Muellerstrasse 44, 6020 Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| |
Collapse
|
30
|
Ijaz S, Haq IU, Malik R, Nadeem G, Ali HM, Kaur S. In silico characterization of differentially expressed short-read nucleotide sequences identified in dieback stress-induced transcriptomic analysis reveals their role as antimicrobial peptides. FRONTIERS IN PLANT SCIENCE 2023; 14:1168221. [PMID: 37021314 PMCID: PMC10069654 DOI: 10.3389/fpls.2023.1168221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/02/2023] [Indexed: 06/19/2023]
Abstract
We investigated the in silico characterization of short-length nucleotide sequences that were differentially expressed in dieback stress-induced transcriptomic analysis. They displayed homology with C-terminal flanking peptides and defensins-like proteins, revealing their antimicrobial activity. Their predicted fingerprints displayed protein signatures related to antimicrobial peptides. These short-length RGAs have been shown to possess structural motifs such as APLT P-type ATPase, casein kinase II (CK2), protein kinase 3, protein kinase C (PKC), and N-glycosylation site that are the attributes of disease resistance genes. The prediction of arginine and lysine residues in active binding sites in ligand docking analysis prophesied them as antimicrobial peptides due to their strong relation with antimicrobial activity. The in silico structural-functional characterization has predicted their role in resistance against microbial pathogens. Moreover, the predicted antimicrobial peptide regions showed their homology with the signature domain of PR-5-like protein and AMP family Thaumatin.
Collapse
Affiliation(s)
- Siddra Ijaz
- Centre of Agricultural Biochemistry and Biotechnology (CABB), University of Agriculture, Faisalabad, Pakistan
| | - Imran Ul Haq
- Department of Plant Pathology, University of Agriculture, Faisalabad, Pakistan
| | - Riffat Malik
- Centre of Agricultural Biochemistry and Biotechnology (CABB), University of Agriculture, Faisalabad, Pakistan
| | - Ghalia Nadeem
- Centre of Agricultural Biochemistry and Biotechnology (CABB), University of Agriculture, Faisalabad, Pakistan
| | - Hayssam M. Ali
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sukhwinder Kaur
- Department of Plant Pathology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
31
|
Cevey ÁC, Pieralisi AV, Donato M, Rada J, Gelpi RJ, Mirkin GA, Goren NB, Penas FN. Macrophages Mediate Healing Properties of Fenofibrate in Experimental Chagasic Cardiomyopathy. ACS Infect Dis 2023; 9:213-220. [PMID: 36661566 DOI: 10.1021/acsinfecdis.2c00535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Chronic cardiomyopathy is one of the most relevant outcomes of Chagas disease associated with parasite persistence and exacerbated inflammatory response. Fenofibrate, a third generation fibric acid derivative and peroxisome proliferator-activated receptor-α ligand, is involved in the regulation of inflammatory response. However, the participation of macrophages in this scenario has not been elucidated. Here we show, for the first time, that macrophages play a fundamental role in the fenofibrate-mediated modulation of heart pro-inflammatory response and fibrosis caused by the infection with Trypanosoma cruzi. Furthermore, macrophages are required for fenofibrate to improve the loss of ventricular function and this restoration correlates with an anti-inflammatory microenvironment. Understanding the contributions of macrophages to the healing properties of fenofibrate reinforces its potential use as a therapeutic drug, with the aim of helping to solve a public health problem, such as chronic Chagas disease.
Collapse
Affiliation(s)
- Ágata Carolina Cevey
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Azul Victoria Pieralisi
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Martín Donato
- Facultad de Medicina, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Jimena Rada
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Ricardo Jorge Gelpi
- Facultad de Medicina, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Gerardo Ariel Mirkin
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM). Facultad de Medicina, CONICET - Universidad de Buenos Aires. Buenos Aires C1121ABG, Argentina
| | - Nora Beatriz Goren
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Federico Nicolás Penas
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| |
Collapse
|
32
|
Carfrae LA, Brown ED. Nutrient stress is a target for new antibiotics. Trends Microbiol 2023; 31:571-585. [PMID: 36709096 DOI: 10.1016/j.tim.2023.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/28/2023]
Abstract
Novel approaches are required to address the looming threat of pan-resistant Gram-negative pathogens and forestall the rise of untreatable infections. Unconventional targets that are uniquely important during infection and tractable to high-throughput drug discovery methods hold high potential for innovation in antibiotic discovery programs. In this context, inhibitors of bacterial nutrient stress are particularly exciting candidates for future antibiotic development. Amino acid, nucleotide, and vitamin biosynthesis pathways are critical for bacterial growth in nutrient-limiting conditions in the laboratory and the host. Although historically dismissed as dispensable for pathogens, a wealth of transposon mutagenesis and single-mutant studies have emerged which demonstrate that several such pathways are critical for infection. Indeed, high-throughput screens of diverse synthetic compounds and natural products have uncovered inhibitors of nutrient biosynthesis. Herein, we review bacterial nutrient biosynthesis and its role during host infection. Further, we explore screening platforms developed to search for inhibitors of these targets and highlight successes among these. Finally, we feature important and sometimes surprising connections between bacterial nutrient biosynthesis, antibiotic activity, and antibiotic resistance.
Collapse
Affiliation(s)
- Lindsey A Carfrae
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, L8S 4L8, Canada; Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Eric D Brown
- Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, L8S 4L8, Canada; Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4L8, Canada; Present address: Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario, L8S 4L8, Canada.
| |
Collapse
|
33
|
Nüse B, Holland T, Rauh M, Gerlach RG, Mattner J. L-arginine metabolism as pivotal interface of mutual host-microbe interactions in the gut. Gut Microbes 2023; 15:2222961. [PMID: 37358082 PMCID: PMC10294761 DOI: 10.1080/19490976.2023.2222961] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 06/05/2023] [Indexed: 06/27/2023] Open
Abstract
L-arginine (L-arg) is a versatile amino acid and a central intestinal metabolite in mammalian and microbial organisms. Thus, L-arg participates as precursor of multiple metabolic pathways in the regulation of cell division and growth. It also serves as a source of carbon, nitrogen, and energy or as a substrate for protein synthesis. Consequently, L-arg can simultaneously modify mammalian immune functions, intraluminal metabolism, intestinal microbiota, and microbial pathogenesis. While dietary intake, protein turnover or de novo synthesis usually supply L-arg in sufficient amounts, the expression of several key enzymes of L-arg metabolism can change rapidly and dramatically following inflammation, sepsis, or injury. Consequently, the availability of L-arg can be restricted due to increased catabolism, transforming L-arg into an essential amino acid. Here, we review the enzymatic pathways of L-arg metabolism in microbial and mammalian cells and their role in immune function, intraluminal metabolism, colonization resistance, and microbial pathogenesis in the gut.
Collapse
Affiliation(s)
- Björn Nüse
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Tim Holland
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Roman G. Gerlach
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, FAUErlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
34
|
Wei Y, Wang Z, Liu Y, Liao B, Zong Y, Shi Y, Liao M, Wang J, Zhou X, Cheng L, Ren B. Extracellular vesicles of Candida albicans regulate its own growth through the l-arginine/nitric oxide pathway. Appl Microbiol Biotechnol 2022; 107:355-367. [PMCID: PMC9703431 DOI: 10.1007/s00253-022-12300-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Yu Wei
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Yaqi Liu
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Yawen Zong
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Min Liao
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Biao Ren
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| |
Collapse
|
35
|
Park HT, Lee SM, Ko S, Kim S, Park HE, Shin MK, Kim D, Yoo HS. Delineating transcriptional crosstalk between Mycobacterium avium subsp. paratuberculosis and human THP-1 cells at the early stage of infection via dual RNA-seq analysis. Vet Res 2022; 53:71. [PMID: 36100945 PMCID: PMC9469519 DOI: 10.1186/s13567-022-01089-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/13/2022] [Indexed: 12/02/2022] Open
Abstract
Mycobacterium avium subsp. paratuberculosis (MAP) is the causative agent of Johne’s disease, a chronic debilitating disease in ruminants. To control this disease, it is crucial to understand immune evasion and the mechanism of persistence by analyzing the early phase interplays of the intracellular pathogens and their hosts. In the present study, host–pathogen interactions at the transcriptomic level were investigated in an in vitro macrophage infection model. When differentiated human THP-1 cells were infected with MAP, the expression of various genes associated with stress responses and metabolism was altered in both host and MAP at 3 h post-infection. MAP upregulates stress-responsive global gene regulators, such as two-component systems and sigma factors, in response to oxidative and cell wall stress. Downstream genes involved in type VII secretion systems, cell wall synthesis (polyketide biosynthesis proteins), and iron uptake were changed in response to the intracellular environment of macrophages. On the host side, upregulation of inflammatory cytokine genes was observed along with pattern recognition receptor genes. Notably, alterations in gene sets involved in arginine metabolism were observed in both the host and MAP, along with significant downregulation of NOS2 expression. These observations suggest that the utilization of metabolites such as arginine by intracellular MAP might affect host NO production. Our dual RNA-seq data can provide novel insights by capturing the global transcriptome with higher resolution, especially in MAP, thus enabling a more systematic understanding of host–pathogen interactions.
Collapse
Affiliation(s)
- Hong-Tae Park
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Sang-Mok Lee
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Korea
| | - Seyoung Ko
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Korea
| | - Suji Kim
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Hyun-Eui Park
- Department of Microbiology, College of Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju, 52828, Korea
| | - Min-Kyoung Shin
- Department of Microbiology, College of Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju, 52828, Korea
| | - Donghyuk Kim
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Korea.
| | - Han Sang Yoo
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
36
|
Metabolomics insights into the interaction between Pseudomonas plecoglossicida and Epinephelus coioides. Sci Rep 2022; 12:13309. [PMID: 35922642 PMCID: PMC9349296 DOI: 10.1038/s41598-022-17387-6] [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: 04/04/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
As a highly infectious epidemic in aquaculture, Pseudomonas plecoglossicida infection results in high mortality of teleosts and serious economic losses. Host–pathogen interactions shape the outcome of an infection, yet we still understand little about the molecular mechanism of these pathogen-mediated processes. Here, a P. plecoglossicida strain (NZBD9) and Epinephelus coioides were investigated as a model system to characterize pathogen-induced host metabolic remodeling over the course of infection. We present a non-targeted metabolomics profiling of E. coioides spleens from uninfected E. coioides and those infected with wild-type and clpV-RNA interference (RNAi) strains. The most significant changes of E. coioides upon infection were associated with amino acids, lysophospatidylcholines, and unsaturated fatty acids, involving disturbances in host nutritional utilization and immune responses. Dihydrosphingosine and fatty acid 16:2 were screened as potential biomarkers for assessing P. plecoglossicida infection. The silencing of the P. plecoglossicida clpV gene significantly recovered the lipid metabolism of infected E. coioides. This comprehensive metabolomics study provides novel insights into how P. plecoglossicida shape host metabolism to support their survival and replication and highlights the potential of the virulence gene clpV in the treatment of P. plecoglossicida infection in aquaculture.
Collapse
|
37
|
McGowan ENS, Wong O, Jones E, Nguyen J, Wee J, Demaria MC, Deliyanti D, Johnson CJ, Hickey MJ, McConville MJ, Wilkinson-Berka JL, Wright MD, Binger KJ. Tetraspanin CD82 restrains phagocyte migration but supports macrophage activation. iScience 2022; 25:104520. [PMID: 35754722 PMCID: PMC9213772 DOI: 10.1016/j.isci.2022.104520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/31/2022] [Accepted: 05/27/2022] [Indexed: 12/03/2022] Open
Abstract
Phagocytes migrate into tissues to combat infection and maintain tissue homeostasis. As dysregulated phagocyte migration and function can lead to inflammation or susceptibility to infection, identifying molecules that control these processes is critical. Here, we show that the tetraspanin CD82 restrains the migration of neutrophils and macrophages into tissues. Cd82−/− phagocytes exhibited excessive migration during in vivo models of peritoneal inflammation, superfusion of CXCL1, retinopathy of prematurity, and infection with the protozoan parasite L. mexicana. However, with the latter, while Cd82−/− macrophages infiltrated infection sites at higher proportions, cutaneous L. mexicana lesions were larger and persisted, indicating a failure to control infection. Analyses of in vitro bone-marrow-derived macrophages showed CD82 deficiency altered cellular morphology, and impaired gene expression and metabolism in response to anti-inflammatory activation. Altogether, this work reveals an important role for CD82 in restraining phagocyte infiltration and mediating their differentiation in response to stimulatory cues. Tetraspanin CD82 restrains phagocyte migration in murine models of inflammation Excessive migration of Cd82−/− myeloid cells exacerbates retinal inflammation Cd82−/− macrophages have a reduced ability to clear Leishmania mexicana parasites CD82 is required for the normal morphology and activation of M2 macrophages
Collapse
Affiliation(s)
- Erin N S McGowan
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Osanna Wong
- Department of Immunology and Pathology, Alfred Research Alliance, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Eleanor Jones
- Department of Immunology and Pathology, Alfred Research Alliance, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.,Flow Cytometry and Imaging Facility, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
| | - Julie Nguyen
- Department of Immunology and Pathology, Alfred Research Alliance, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC 3168, Australia
| | - Janet Wee
- Department of Immunology and Pathology, Alfred Research Alliance, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC 3168, Australia
| | - Maria C Demaria
- Department of Immunology and Pathology, Alfred Research Alliance, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Devy Deliyanti
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Chad J Johnson
- Bioimaging Platform, La Trobe University, Bundoora, VIC 3086, Australia
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC 3168, Australia
| | - Malcolm J McConville
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Jennifer L Wilkinson-Berka
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Mark D Wright
- Department of Immunology and Pathology, Alfred Research Alliance, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Katrina J Binger
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia.,Department of Immunology and Pathology, Alfred Research Alliance, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.,Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Bundoora, VIC 3086, Australia
| |
Collapse
|
38
|
Gu C, Zhou Z, Yu Z, He M, He L, Luo Z, Xiao W, Yang Q, Zhao F, Li W, Shen L, Han J, Cao S, Zuo Z, Deng J, Yan Q, Ren Z, Zhao M, Yu S. The Microbiota and It’s Correlation With Metabolites in the Gut of Mice With Nonalcoholic Fatty Liver Disease. Front Cell Infect Microbiol 2022; 12:870785. [PMID: 35694542 PMCID: PMC9186341 DOI: 10.3389/fcimb.2022.870785] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, nonalcoholic fatty liver disease (NAFLD) has become the most common liver disease in the world. As an important model animal, the characteristics of gut microbiota alteration in mice with NAFLD have been studied but the changes in metabolite abundance in NAFLD mice and how the gut microbiota affects these intestinal metabolites remain unclear. In this experiment, a mouse model for NAFLD was established by a high-fat diet. The use of 16S rDNA technology showed that while there were no significant changes in the alpha diversity in the cecum of NAFLD mice, the beta diversity changed significantly. The abundance of Blautia, Unidentified-Lachnospiraceae, Romboutsia, Faecalibaculum, and Ileibacterium increased significantly in NAFLD mice, while Allobaculum and Enterorhabdus decreased significantly. Amino acids, lipids, bile acids and nucleotide metabolites were among the 167 significantly different metabolites selected. The metabolic pathways of amino acids, SFAs, and bile acids were significantly enhanced, while the metabolic pathways of PUFAs, vitamins, and nucleotides were significantly inhibited. Through correlation and MIMOSA2 analysis, it is suggested that gut microbiota does not affect the changes of lipids and bile acids but can reduce thiamine, pyridoxine, and promote L-phenylalanine and tyramine production. The findings of this study will help us to better understand the relationship between gut microbiota and metabolites in NAFLD.
Collapse
Affiliation(s)
- Congwei Gu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
| | - Zihan Zhou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zehui Yu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
| | - Manli He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
| | - Lvqin He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
| | - Zhengzhong Luo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Wudian Xiao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
| | - Qian Yang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
| | - Fangfang Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Weiyao Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Liuhong Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jianhong Han
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
| | - Suizhong Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qigui Yan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingde Zhao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- *Correspondence: Mingde Zhao, ; Shumin Yu,
| | - Shumin Yu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Mingde Zhao, ; Shumin Yu,
| |
Collapse
|
39
|
Liu N, Shi F, Yang L, Liao W, Cao Y. Oncogenic viral infection and amino acid metabolism in cancer progression: Molecular insights and clinical implications. Biochim Biophys Acta Rev Cancer 2022; 1877:188724. [DOI: 10.1016/j.bbcan.2022.188724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 02/08/2023]
|
40
|
Nutrient Sensing and Biofilm Modulation: The Example of L-arginine in Pseudomonas. Int J Mol Sci 2022; 23:ijms23084386. [PMID: 35457206 PMCID: PMC9028604 DOI: 10.3390/ijms23084386] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 12/01/2022] Open
Abstract
Bacterial biofilm represents a multicellular community embedded within an extracellular matrix attached to a surface. This lifestyle confers to bacterial cells protection against hostile environments, such as antibiotic treatment and host immune response in case of infections. The Pseudomonas genus is characterised by species producing strong biofilms difficult to be eradicated and by an extraordinary metabolic versatility which may support energy and carbon/nitrogen assimilation under multiple environmental conditions. Nutrient availability can be perceived by a Pseudomonas biofilm which, in turn, readapts its metabolism to finally tune its own formation and dispersion. A growing number of papers is now focusing on the mechanism of nutrient perception as a possible strategy to weaken the biofilm barrier by environmental cues. One of the most important nutrients is amino acid L-arginine, a crucial metabolite sustaining bacterial growth both as a carbon and a nitrogen source. Under low-oxygen conditions, L-arginine may also serve for ATP production, thus allowing bacteria to survive in anaerobic environments. L-arginine has been associated with biofilms, virulence, and antibiotic resistance. L-arginine is also a key precursor of regulatory molecules such as polyamines, whose involvement in biofilm homeostasis is reported. Given the biomedical and biotechnological relevance of biofilm control, the state of the art on the effects mediated by the L-arginine nutrient on biofilm modulation is presented, with a special focus on the Pseudomonas biofilm. Possible biotechnological and biomedical applications are also discussed.
Collapse
|
41
|
The arginine deaminase system plays distinct roles in Borrelia burgdorferi and Borrelia hermsii. PLoS Pathog 2022; 18:e1010370. [PMID: 35286343 PMCID: PMC8947608 DOI: 10.1371/journal.ppat.1010370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/24/2022] [Accepted: 02/14/2022] [Indexed: 11/23/2022] Open
Abstract
Borrelia species are amino acid auxotrophs that utilize di- and tri- peptides obtained through their oligopeptide transport system to supply amino acids for replicative growth during their enzootic cycles. However, Borrelia species from both the Lyme disease (LD) and relapsing fever (RF) groups harbor an amino acid transport and catabolism system, the Arginine Deiminase System (ADI), that could potentially augment intracellular L-arginine required for growth. RF spirochetes contain a “complete”, four gene ADI (arcA, B, D, and C) while LD spirochetes harbor arcA, B, and sometimes D but lack arcC (encoding carbamate kinase). In this study, we evaluated the role of the ADI system in bacterial survival and virulence and discovered important differences in RF and LD ADIs. Both in vitro and in a murine model of infection, B. hermsii cells significantly reduced extracellular L-arginine levels and that reduction was dependent on arginine deiminase expression. Conversely, B. burgdorferi did not reduce the concentration of L-arginine during in vitro growth experiments nor during infection of the mammalian host, suggesting a fundamental difference in the ability to directly utilize L-arginine compared to B. hermsii. Further experiments using a panel of mutants generated in both B. burgdorferi and B. hermsii, identified important differences in growth characteristics and ADI transcription and protein expression. We also found that the ADI system plays a key role in blood and spleen colonization in RF spirochetes. In this study we have identified divergent metabolic strategies in two closely related human pathogens, that ultimately impacts the host-pathogen interface during infection. Reports of tick-borne diseases have been steadily increasing in the US and the number of Lyme disease cases caused by B. burgdorferi have tripled since the late 1990’s. Although less common, cases of tick-borne relapsing fever, caused by B. hermsii and B. turicatae in the US, have increased as well. While transmitted by different ticks and maintained in unique enzootic cycles, the closely related spirochetes B. burgdorferi and B. hermsii share numerous genetic features including a truncated and streamlined capacity for metabolic activity. In this study we combine genetic and biochemical assays to define the role of the ADI in the infective cycles of B. burgdorferi and B. hermsii. When we compared B. burgdorferi and B. hermsii, we identified important differences in their respective ADI’s including operon arrangement, sensitivity to L-arginine and L-ornithine levels, as well as gene and protein expression. In addition, we show that arginine deiminase is required to reduce host L-arginine levels during murine infection with B. hermsii. This study provides new insights into the metabolic activities of two medically relevant spirochetes and highlights the dynamic nature of host-pathogen interactions.
Collapse
|
42
|
Huang Z, Zhang Y, Zheng X, Liu Z, Yao D, Zhao Y, Chen X, Aweya JJ. Functional characterization of arginine metabolic pathway enzymes in the antibacterial immune response of penaeid shrimp. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 127:104293. [PMID: 34648768 DOI: 10.1016/j.dci.2021.104293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/09/2021] [Accepted: 10/10/2021] [Indexed: 06/13/2023]
Abstract
Arginine metabolism pathway enzymes and products are important modulators of several physiological processes in animals, including immune response. Although some components of the arginine metabolic pathway have been reported in penaeid shrimps, no systematic study has explored all the key pathway enzymes involved in shrimp antimicrobial response. Here, we explored the role of the three key arginine metabolism enzymes (nitric-oxide synthase (NOS), arginase (ARG), agmatinase (AGM)) in Penaeus vannamei antimicrobial immunity. First, P. vannamei homologs of ARG and AGM (PvARG and PvAGM) were cloned and found to be evolutionally conserved with invertebrate counterparts. Transcript levels of PvARG, PvAGM, and PvNOS were ubiquitously expressed in healthy shrimp tissues and induced in hemocytes and hepatopancreas upon challenge with Gram-negative (Vibrio parahaemolyticus) and Gram-positive (Streptoccocus iniae) bacteria, suggesting their involvement in shrimp antimicrobial immune response. Besides, RNA interference knockdown and enzyme activity assay revealed an antagonistic relationship between PvARG/PvAGM and PvNOS, while this relationship was broken upon pathogen stimulation. Interestingly, knockdown of PvNOS increased Vibrio abundance in shrimp hemolymph, whereas knockdown of PvAGR reduced Vibrio abundance. Taken together, our present data shows that homologs of the key arginine metabolism pathway enzymes in penaeid shrimp (PvARG, PvAGM, and PvNOS) work synergistically and/or antagonistically to modulate antibacterial immune response.
Collapse
Affiliation(s)
- Zishu Huang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China
| | - Yueling Zhang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, 511458, China
| | - Xiaoyu Zheng
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China
| | - Zhuoyan Liu
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China
| | - Defu Yao
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China
| | - Yongzhen Zhao
- Guangxi Academy of Fishery Sciences, Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Nanning, 530021, China
| | - Xiaohan Chen
- Guangxi Academy of Fishery Sciences, Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Nanning, 530021, China
| | - Jude Juventus Aweya
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou, 515063, China.
| |
Collapse
|
43
|
Sarkar D, Vijayan R, Gourinath S, Sau AK. A unique aromatic cluster near the active site of H. pylori CPA is essential for catalytic function. Biophys J 2022; 121:248-262. [PMID: 34932956 PMCID: PMC8790187 DOI: 10.1016/j.bpj.2021.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/16/2021] [Accepted: 12/16/2021] [Indexed: 01/21/2023] Open
Abstract
Polyamines are essential for cell growth and proliferation. In plants and many bacteria, including Helicobacter pylori, the parent polyamine putrescine is only produced through the metabolism of N-carbamoylputrescine by N-carbamoylputrescine amidase (CPA). Thus, CPA is a crucial intermediate enzyme. Moreover, the absence of CPA in humans makes its presence in H. pylori a potential target for the development of new therapeutics against this pathogen. Despite this enzyme's presence in plants and bacteria, its function is not completely explored. Using structure-guided biochemical and biophysical studies on H. pylori CPA, we discovered an aromatic cluster containing four conserved tryptophans near the catalytic site and elucidated its role. Mutational studies revealed that they are individually vital to enzyme function. Unlike wild-type, which forms a hexamer, the Trp to Ala mutants only formed dimers. Interestingly, two other conserved residues, Gln155 and Asp278, interact with the tryptophan cluster and perform similar roles. Our results indicate that aromatic-aromatic and H-bonding contacts between the residues (Trp156-Trp273, Trp196-Gln155, and Trp153-Asp278) play a crucial role in stimulating activity through hexamer formation. Additionally, Trp156 is essential to generating a catalytically efficient hexamer. These results suggest dual roles for the tryptophans; in hexamer formation and in generating its functionally active form, thereby providing a mechanistic understanding into the role of the cluster. We also elucidated the catalytic roles of Glu43, Lys115, and Cys152, which are present at the active site. Our findings highlight, for the first time, the importance of a tryptophan cluster in H. pylori CPA that can be exploited to design therapeutic inhibitors.
Collapse
Affiliation(s)
- Ditsa Sarkar
- National Institute of Immunology, New Delhi, India
| | | | | | - Apurba Kumar Sau
- National Institute of Immunology, New Delhi, India,Correspondence:
| |
Collapse
|
44
|
Vytrykhovskyi AI, Fedorchenko MV. REPERFUSION INJURY IN ACUTE PERIOD OF MYOCARDIAL INFARCTION - WAYS OF PREVENTION AND CORRECTION. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2022; 75:2514-2518. [PMID: 36472290 DOI: 10.36740/wlek202210137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
OBJECTIVE The aim: To identify pathophysiological peculiarities of myocardial reperfusion injury and ways of its reduction based on the literature data analysis. PATIENTS AND METHODS Materials and methots: This literature review was made by searching the PubMed database using key words . Additional data were sought in the Google search engine by entering key words: " risk factors, ischemic heart disease, arrhythmia, sudden cardiac death, heart rhythm, heart failure." in the Polish, English, Russian and Ukrainian language versions. CONCLUSION Conclusions: Considering conducted data analysis, provided data indicate the prospects of phosphocreatine usage in treatment scheme of heart rhythm disorders and heart failure on the background of myocardial ischemia and elimination of reperfusion injury and myocardial remodeling consequences.
Collapse
Affiliation(s)
- Andriy I Vytrykhovskyi
- IVANO-FRANKIVSK NATIONAL MEDICAL UNIVERSITY, IVANO-FRANKIVSK, UKRAINE; CNO «IVANO-FRANKIVSK REGIONAL CARDIOLOGICAL CENTER» IFSS, IVANO-FRANKIVSK, UKRAINE
| | - Muhaylo V Fedorchenko
- IVANO-FRANKIVSK NATIONAL MEDICAL UNIVERSITY, IVANO-FRANKIVSK, UKRAINE; CNO «IVANO-FRANKIVSK REGIONAL CARDIOLOGICAL CENTER» IFSS, IVANO-FRANKIVSK, UKRAINE
| |
Collapse
|
45
|
Hai-De W, Shuai L, Bing-Bing W, Jie L, Jian-Zhong X, Wei-Guo Z. Metabolic engineering of Escherichia coli for efficient production of l-arginine. ADVANCES IN APPLIED MICROBIOLOGY 2022; 122:127-150. [PMID: 37085192 DOI: 10.1016/bs.aambs.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
As a semi-essential amino acid, l-arginine (l-Arg) plays an important role in food, health care, and medical treatment. At present, the main method of producing l-Arg is the use of microbial fermentation. Therefore, the selection and breeding of high-efficiency microbial strains is the top priority. To continuously improve the l-Arg production performance of the strains, a series of metabolic engineering strategies have been tried to transform the strains. The production of l-Arg by metabolically engineered Corynebacterium glutamicum (C. glutamicum) reached a relatively high level. Escherichia coli (E. coli), as a strain with great potential for l-Arg production, also has a large number of research strategies aimed at screening effective E. coli for producing l-Arg. E. coli also has a number of advantages over C. glutamicum in producing l-Arg. Therefore, it is of great significance to screen out excellent and stable E. coli to produce l-Arg. Here, based on recent research results, we review the metabolic pathways of l-Arg production in E. coli, the research progress of l-Arg production in E. coli, and various regulatory strategies implemented in E. coli.
Collapse
|
46
|
Ashour NH, El-Tanbouly DM, El Sayed NS, Khattab MM. Roflumilast ameliorates cognitive deficits in a mouse model of amyloidogenesis and tauopathy: Involvement of nitric oxide status, Aβ extrusion transporter ABCB1, and reversal by PKA inhibitor H89. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110366. [PMID: 34051306 DOI: 10.1016/j.pnpbp.2021.110366] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/01/2021] [Accepted: 05/23/2021] [Indexed: 01/13/2023]
Abstract
The biological cascade of second messenger-cyclic adenosine monophosphate (cAMP) -as a molecular mechanism implicated in memory and learning regulation has captured the attention of neuroscientists worldwide. cAMP triggers its foremost effector, protein kinase A (PKA), resulting in the activation of innumerable downstream targets. Roflumilast (ROF), a phosphodiesterase 4 inhibitor, has demonstrated a greater efficiency in enhancing cAMP signaling in various neurological disorders. This study was conducted to identify various downstream targets of PKA as mechanistic tools through which ROF could hinder the progressive cognitive impairment following central streptozotocin (STZ) administration in mice. Animals were injected with STZ (3 mg/kg/i.c.v) once. Five hours later, mice received ROF (0.4 mg/kg) with or without the PKA inhibitor, H89, for 21 days. ROF highly preserved the structure of hippocampal neurons. It improved the ability of mice to develop short-term memories and retrieve spatial memories in Y-maze and Morris water maze tests, respectively. ROF enhanced the gene expression of ABCB1 transporters and pregnane X receptors (PXR), and hampered Aβ accumulation in hippocampus. Simultaneously, it interfered with the processes of tau phosphorylation and nitration. This effect was associated with an upsurge in hippocampal arginase activity as well as a decline in glycogen synthase kinase-3β activity, nitric oxide synthase (NOS) activity, and inducible NOS expression. Contrariwise, ROF's beneficial effects were utterly abolished by co-administration of H89. In conclusion, boosting PKA, by ROF, modulated PXR/ABCB1 expression and arginase/NOS activities to restrict the main post-translational modifications of tau, Aβ deposition and, accordingly, cognitive deterioration of sporadic Alzheimer's disease.
Collapse
Affiliation(s)
- Nada H Ashour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Dalia M El-Tanbouly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
47
|
Prieto A, Bernabeu M, Sánchez-Herrero JF, Pérez-Bosque A, Miró L, Bäuerl C, Collado C, Hüttener M, Juárez A. Modulation of AggR levels reveals features of virulence regulation in enteroaggregative E. coli. Commun Biol 2021; 4:1295. [PMID: 34785760 PMCID: PMC8595720 DOI: 10.1038/s42003-021-02820-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) strains are one of the diarrheagenic pathotypes. EAEC strains harbor a virulence plasmid (pAA2) that encodes, among other virulence determinants, the aggR gene. The expression of the AggR protein leads to the expression of several virulence determinants in both plasmids and chromosomes. In this work, we describe a novel mechanism that influences AggR expression. Because of the absence of a Rho-independent terminator in the 3'UTR, aggR transcripts extend far beyond the aggR ORF. These transcripts are prone to PNPase-mediated degradation. Structural alterations in the 3'UTR result in increased aggR transcript stability, leading to increased AggR levels. We therefore investigated the effect of increased AggR levels on EAEC virulence. Upon finding the previously described AggR-dependent virulence factors, we detected novel AggR-regulated genes that may play relevant roles in EAEC virulence. Mutants exhibiting high AggR levels because of structural alterations in the aggR 3'UTR show increased mobility and increased pAA2 conjugation frequency. Furthermore, among the genes exhibiting increased fold change values, we could identify those of metabolic pathways that promote increased degradation of arginine, fatty acids and gamma-aminobutyric acid (GABA), respectively. In this paper, we discuss how the AggR-dependent increase in specific metabolic pathways activity may contribute to EAEC virulence.
Collapse
Affiliation(s)
- Alejandro Prieto
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain
| | - Manuel Bernabeu
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain
| | | | - Anna Pérez-Bosque
- Department of Biochemistry and Physiology, Universitat de Barcelona, Barcelona, Spain
- Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, Barcelona, Spain
| | - Lluïsa Miró
- Department of Biochemistry and Physiology, Universitat de Barcelona, Barcelona, Spain
- Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, Barcelona, Spain
| | - Christine Bäuerl
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Carmen Collado
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Mário Hüttener
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain.
| | - Antonio Juárez
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain.
- Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain.
| |
Collapse
|
48
|
Marselli L, Bosi E, De Luca C, Del Guerra S, Tesi M, Suleiman M, Marchetti P. Arginase 2 and Polyamines in Human Pancreatic Beta Cells: Possible Role in the Pathogenesis of Type 2 Diabetes. Int J Mol Sci 2021; 22:ijms222212099. [PMID: 34829980 PMCID: PMC8625980 DOI: 10.3390/ijms222212099] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/16/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Arginase 2 (ARG2) is a manganese metalloenzyme involved in several tissue specific processes, from physiology to pathophysiology. It is variably expressed in extra-hepatic tissues and is located in the mitochondria. In human pancreatic beta cells, ARG2 is downregulated in type 2 diabetes. The enzyme regulates the synthesis of polyamines, that are involved in pancreas development and regulation of beta cell function. Here, we discuss several features of ARG2 and polyamines, which can be relevant to the pathophysiology of type 2 diabetes.
Collapse
|
49
|
Gan Z, Zhang M, Xie D, Wu X, Hong C, Fu J, Fan L, Wang S, Han S. Glycinergic Signaling in Macrophages and Its Application in Macrophage-Associated Diseases. Front Immunol 2021; 12:762564. [PMID: 34675940 PMCID: PMC8523992 DOI: 10.3389/fimmu.2021.762564] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidences support that amino acids direct the fate decision of immune cells. Glycine is a simple structural amino acid acting as an inhibitory neurotransmitter. Besides, glycine receptors as well as glycine transporters are found in macrophages, indicating that glycine alters the functions of macrophages besides as an inhibitory neurotransmitter. Mechanistically, glycine shapes macrophage polarization via cellular signaling pathways (e.g., NF-κB, NRF2, and Akt) and microRNAs. Moreover, glycine has beneficial effects in preventing and/or treating macrophage-associated diseases such as colitis, NAFLD and ischemia-reperfusion injury. Collectively, this review highlights the conceivable role of glycinergic signaling for macrophage polarization and indicates the potential application of glycine supplementation as an adjuvant therapy in macrophage-associated diseases.
Collapse
Affiliation(s)
- Zhending Gan
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Meiyu Zhang
- College of Animal Science and Technology, Guangdong Polytechnic of Science and Trade, Guangzhou, China
| | - Donghui Xie
- Nanchang Academy of Agricultural Sciences, Nanchang, China
| | - Xiaoyan Wu
- College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Science, Lanzhou, China
| | - Changming Hong
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jian Fu
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Lijuan Fan
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shengyi Wang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Science, Lanzhou, China
| | - Sufang Han
- College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
50
|
Seif MA, Al-Mohammed HI. ASSESSMENT OF THE OXIDATIVE AND NITROSATIVE STRESS IN THE SERUM OF SAUDI PATIENTS WITH CUTANEOUS LEISHMANIASIS BEFORE AND AFTER TREATMENT. J Parasitol 2021; 107:810-816. [PMID: 34648629 DOI: 10.1645/20-109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Macrophages, within which Leishmania species replicate, generate large amounts of reactive oxygen species (ROS) and reactive nitrogen species (RNS) to kill these parasites. The present study assessed the oxidative and nitrosative stress, and specific immune enzymes in the serum of patients with cutaneous leishmaniasis (Cl) before and after treatment and in the control individuals. Serum activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), L-arginase, myeloperoxidase (MPO), and adenosine deaminase (ADA) and the levels of reduced glutathione, malondialdehyde (MDA), and nitric oxide (NO) were studied. The activities of L-arginase, MPO, and ADA and the levels of MDA and NO were significantly elevated (P < 0.001), while the activities of SOD, CAT, and GSH-Px, and the levels of reduced glutathione (GSH) were significantly (P < 0.001) reduced in untreated patients as compared with values of patients after treatment and of control individuals. The treatment, which included intramuscular injection of sodium stibogluconate and meglumine antimoniate, ameliorated these factors in comparison to the untreated group. These results suggest that oxidative and nitrosative stress may play an important role in the pathogenesis of untreated cutaneous leishmaniasis. Furthermore, the reduction in oxidative and nitrosative stress in the treated Cl patients may be due to the drug decreasing energy production by the parasite, which eventually leads to its death.
Collapse
Affiliation(s)
- Mossad A Seif
- Division of Biochemistry, Biomedical Sciences Department, College of Medicine, King Faisal University, P.O. Box 10950, Hufof, Al-Ahsa, 31982, Kingdom of Saudi Arabia
| | - Hamdan I Al-Mohammed
- Division of Parasitology, Biomedical Sciences Department, College of Medicine, King Faisal University, P.O. Box 10950, Hufof, Al-Ahsa, 31982, Kingdom of Saudi Arabia
| |
Collapse
|