51
|
Garcia Gonzalez J, Hernandez FJ. Nuclease activity: an exploitable biomarker in bacterial infections. Expert Rev Mol Diagn 2022; 22:265-294. [PMID: 35240900 DOI: 10.1080/14737159.2022.2049249] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION In the increasingly challenging field of clinical microbiology, diagnosis is a cornerstone whose accuracy and timing are crucial for the successful management, therapy, and outcome of infectious diseases. Currently employed biomarkers of infectious diseases define the scope and limitations of diagnostic techniques. As such, expanding the biomarker catalog is crucial to address unmet needs and bring about novel diagnostic functionalities and applications. AREAS COVERED This review describes the extracellular nucleases of 15 relevant bacterial pathogens and discusses the potential use of nuclease activity as a diagnostic biomarker. Articles were searched for in PubMed using terms: "nuclease", "bacteria", "nuclease activity" or "biomarker". For overview sections, original and review articles between 2000 and 2019 were searched for using terms: "infections", "diagnosis", "bacterial", "burden", "challenges". Informative articles were selected. EXPERT OPINION Using the catalytic activity of nucleases offers new possibilities compared to established biomarkers. Nucleic acid activatable reporters in combination with different transduction platforms and delivery methods can be used to detect disease-associated nuclease activity patterns in vitro and in vivo for prognostic and diagnostic applications. Even when these patterns are not obvious or of unknown etiology, screening platforms could be used to identify new disease reporters.
Collapse
Affiliation(s)
- Javier Garcia Gonzalez
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.,Wallenberg Centre for Molecular Medicine (WCMM), Linköping, Sweden.,Nucleic Acids Technologies Laboratory (NAT-lab), Linköping University, Linköping, Sweden
| | - Frank J Hernandez
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.,Wallenberg Centre for Molecular Medicine (WCMM), Linköping, Sweden.,Nucleic Acids Technologies Laboratory (NAT-lab), Linköping University, Linköping, Sweden
| |
Collapse
|
52
|
Abstract
Typhoid toxin is an A2B5 protein toxin and an important virulence factor for the human-adapted bacterial pathogen Salmonella enterica serovar Typhi, the causative agent of typhoid fever. Typhoid toxin contains two enzymatic subunits, PltA and CdtB, which dock onto a pentameric delivery platform composed of the protein PltB. It was recently reported that the same enzymatic subunits can assemble with a different delivery platform composed of the protein PltC, forming a distinct version of typhoid toxin. However, the differences in structure and receptor specificity between the PltC and PltB typhoid toxins remain unknown. Here, we determined atomic-level structures of the pentameric PltC subunit, the fully assembled PltC typhoid toxin, and the PltC pentamers in complex with glycan receptors. Biochemical and structural analyses indicate that PltB and PltC are unable to form heteromeric delivery complexes due to electrostatic repulsion at the subunit interface and thus form separate toxins only. We further observed that, despite low sequence similarity between PltB and PltC, they interact with PltA in a similar manner but that PltC exhibits stronger electrostatic interactions with PltA, enabling it to outcompete PltB in toxin assembly. The ligand-bound atomic structures of PltC show an additional glycan binding site not found in PltB and glycan array analysis indicates that PltB and PltC exhibit significant differences in glycan binding specificity. Collectively, this study offers atomic-level insights into how S. Typhi produces two distinct versions of typhoid toxin, thereby generating functional diversity in this key virulence factor. IMPORTANCE Typhoid fever is a devastating disease that kills more than 115,000 people every year and is caused by Salmonella Typhi. Typhoid toxin, exclusively produced by S. Typhi, was demonstrated to be responsible for the pathogenesis of typhoid fever. Typhoid toxin consists of a pentameric delivery B subunit to transport the catalytic A subunits into the host cell through binding of the glycan receptors. Recent study shows that S. Typhi encodes two homologous delivery B subunits that are able to associate with the same active subunits to produce alternative toxins with distinct functional characteristics. Here, we show that the two delivery subunits can form only homopentameric delivery platforms that compete to associate with typhoid toxin's active subunits and that the two resulting toxins have distinct glycan-binding properties that confer distinct functional traits. These findings highlight the unique assembly and functional diversification of typhoid toxins.
Collapse
|
53
|
Qian M, Cao S, Wang T, Xu X, Zhang Q. Apoptosis triggered by cytolethal distending toxin B subunit of Helicobacter hepaticus is aggravated by autophagy inhibition in mouse hepatocytes. Biochem Biophys Res Commun 2022; 598:40-46. [PMID: 35151202 DOI: 10.1016/j.bbrc.2022.01.121] [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: 12/03/2021] [Accepted: 01/29/2022] [Indexed: 11/02/2022]
Abstract
Hepatocytes injury caused by cytolethal distending toxin (CDT) are major events during helicobacter hepaticus (H.hepaticus) infection. Recent study showed that pre-survival autophagy was promoted against CdtB subunit induced DNA damage. In the present study, we demonstrated that inflammatory cytokines IL-6, IL-1β, TNF-α, IFN-α, IFN-γ expression and STAT phosphorylation were promoted by CdtB. Besides, CdtB decreased cell viability while promote apoptosis in mouse liver (AML12) cells. Especially, apoptotic protein caspase-9, caspase-3 and PARP were activated while the ratio of Bcl-2/Bax was decreased after CdtB treatment. Moreover, apoptosis induced by CdtB was inhibited due to Erk/p38 MAPK signaling pathway suppression performed with SB203580 or U0126. Meanwhile, we found that CdtB increased autophagic marker levels accompanied by Akt/mTOR/P70S6K signaling pathway in a dose dependent manner. To assess the correlation between autophagy and apoptosis induced by H.hepaticus, chloroquine (CQ, 50 μM) was employed to inhibit autophagy. The result showed that inhibition of autophagy with CQ treatment promoted apoptosis induced by CdtB. Altogether, all these results suggest that CdtB triggers apoptosis via MAPK/Erk/p38 signaling pathway in caspase dependent manner, which was prevented by autophagy in AML12 cells. Collectively, our findings provide new insights into the virulence potential of CdtB on the molecular pathogenesis throughout H.hepaticus infection.
Collapse
Affiliation(s)
- Miao Qian
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Shuyang Cao
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tao Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Xiangming Xu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Taizhou University, Taizhou, China.
| | - Quan Zhang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
| |
Collapse
|
54
|
Hbibi A, Bouziane A, Lyoussi B, Zouhdi M, Benazza D. Aggregatibacter actinomycetemcomitans: From Basic to Advanced Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1373:45-67. [DOI: 10.1007/978-3-030-96881-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
55
|
OUP accepted manuscript. Pathog Dis 2022; 80:6521441. [DOI: 10.1093/femspd/ftac003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/30/2021] [Accepted: 02/01/2022] [Indexed: 11/15/2022] Open
|
56
|
De Luca E, Álvarez-Narváez S, Maboni G, Baptista RP, Nemeth NM, Niedringhaus KD, Ladner JT, Lorch JM, Koroleva G, Lovett S, Palacios GF, Sanchez S. Comparative Genomics Analyses Support the Reclassification of Bisgaard Taxon 40 as Mergibacter gen. nov., With Mergibacter septicus sp. nov. as Type Species: Novel Insights Into the Phylogeny and Virulence Factors of a Pasteurellaceae Family Member Associated With Mortality Events in Seabirds. Front Microbiol 2021; 12:667356. [PMID: 34880834 PMCID: PMC8645869 DOI: 10.3389/fmicb.2021.667356] [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: 02/12/2021] [Accepted: 10/11/2021] [Indexed: 11/22/2022] Open
Abstract
The Pasteurellaceae family has been associated with fatal diseases in numerous avian species. Several new taxa within this family, including Bisgaard taxon 40, have been recently described in wild birds, but their genomic characteristics and pathogenicity are not well understood. We isolated Bisgaard taxon 40 from four species of seabirds, including one sampled during a mass, multi-species mortality event in Florida, United States. Here, we present a comprehensive phenotypic and genetic characterization of Bisgaard taxon 40 and comparative genomic analysis with reference strains from the Pasteurellaceae family, aiming at determining its phylogenetic position, antimicrobial susceptibility profile, and identifying putative virulence factors. In silico multilocus sequence-based and whole-genome-based phylogenetic analysis clustered all Bisgaard taxon 40 strains together on a distinct branch separated from the other members of the Pasteurellaceae family, indicating that Bisgaard taxon 40 could represent a new genus. These findings were further supported by protein similarity analyses using the concatenation of 31 conserved proteins and other taxonomic approaches such as the percentage of conserved protein test. Additionally, several putative virulence factors were identified, including those associated with adhesion (capsule, ompA, ompH) and colonization (exbD, fur, galU, galE, lpxA, lpxC, and kdsA) of the host and a cytolethal distending toxin (cdt), which may have played a role in disease development leading to the mortality event. Considerably low minimum inhibitory concentrations (MICs) were found for all the drugs tested, in concordance with the absence of antimicrobial resistance genes in these genomes. The novel findings of this study highlight genomic and phenotypic characteristics of this bacterium, providing insights into genome evolution and pathogenicity. We propose a reclassification of these organisms within the Pasteurellaceae family, designated as Mergibacter gen. nov., with Mergibacter septicus sp. nov. as the type species. The type strain is Mergibacter septicus A25201T (=DSM 112696).
Collapse
Affiliation(s)
- Eliana De Luca
- Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Sonsiray Álvarez-Narváez
- Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Grazieli Maboni
- Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Rodrigo P Baptista
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Nicole M Nemeth
- Southeastern Cooperative Wildlife Disease Study, Departments of Pathology and Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Kevin D Niedringhaus
- Southeastern Cooperative Wildlife Disease Study, Departments of Pathology and Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Veterinary Medical Teaching Hospital, University of California, Davis, Davis, CA, United States
| | - Jason T Ladner
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States.,U.S. Geological Survey, National Wildlife Health Center, Madison, WI, United States
| | - Jeffrey M Lorch
- U.S. Geological Survey, National Wildlife Health Center, Madison, WI, United States
| | - Galina Koroleva
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Sean Lovett
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Gustavo F Palacios
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Susan Sanchez
- Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
57
|
Callahan SM, Johnson JG. Transposon-Based Identification of Factors That Promote Campylobacter jejuni Nuclease Activity. Curr Protoc 2021; 1:e293. [PMID: 34875141 DOI: 10.1002/cpz1.293] [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: 11/08/2022]
Abstract
Nucleases are ubiquitous in pathogens and allow bacteria to acquire nucleotide nutrients, take up foreign DNA, induce tissue damage, degrade neutrophil extracellular traps, and modulate the host inflammatory response. Furthermore, nucleases can modulate numerous bacterial virulence factors, promoting bacterial growth and disease. To understand how bacteria can produce nucleases, an unbiased approach is needed to identify these systems. Campylobacter jejuni is the leading cause of bacterial-derived gastroenteritis and utilizes numerous systems to damage host DNA. Therefore, it is imperative to identify C. jejuni nucleases to understand the molecular mechanism of both infection and pathology. Detailed protocols for a transposon insertion sequencing-based DNase agar screen, a quantitative PCR nuclease screen, and PCR transposon insertion confirmation are included in this article. © 2021 Wiley Periodicals LLC. Basic Protocol 1: DNase agar colony screen of Campylobacter jejuni transposon insertion sequencing library isolates Basic Protocol 2: Quantitative PCR nuclease screen of transposon insertion sequencing library isolates Basic Protocol 3: PCR transposon insertion confirmation.
Collapse
Affiliation(s)
- Sean M Callahan
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee
| | - Jeremiah G Johnson
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee
| |
Collapse
|
58
|
Lai YR, Chang YF, Ma J, Chiu CH, Kuo ML, Lai CH. From DNA Damage to Cancer Progression: Potential Effects of Cytolethal Distending Toxin. Front Immunol 2021; 12:760451. [PMID: 34868002 PMCID: PMC8634426 DOI: 10.3389/fimmu.2021.760451] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
Cytolethal distending toxin (CDT), one of the most important genotoxins, is produced by several gram-negative bacteria and is involved in bacterial pathogenesis. Recent studies have shown that bacteria producing this peculiar genotoxin target host DNA, which potentially contributes to development of cancer. In this review, we highlighted the recent studies focusing on the idea that CDT leads to DNA damage, and the cells with inappropriately repaired DNA continue cycling, resulting in cancer development. Understanding the detailed mechanisms of genotoxins that cause DNA damage might be useful for targeting potential markers that drive cancer progression and help to discover new therapeutic strategies to prevent diseases caused by pathogens.
Collapse
Affiliation(s)
- Yi-Ru Lai
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Fang Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jason Ma
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Ming-Ling Kuo
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Ho Lai
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
59
|
Abstract
El cuerpo humano está expuesto continuamente a microorganismos tanto fijos como transitorios, así como sus metabolitos tóxicos, lo cual puede conducir a la aparición y progresión del cáncer en sitios distantes al hábitat particular de cada microbio. Diversos estudios científicos han hecho posible entender la relación estrecha que existe entre microbioma y cáncer, ya que los componentes del primero, al tener la capacidad de migrar a diferentes zonas del cuerpo, pueden contribuir al desarrollo de diversas enfermedades crónicas. Los estudios de metagenómica sugieren que la disbiosis, en la microbiota comensal, está asociada con trastornos inflamatorios y varios tipos de cáncer, los cuales pueden ocurrir por sus efectos sobre el metabolismo, la proliferación celular y la inmunidad. La microbiota puede producir el cáncer cuando existen condiciones predisponentes, como en la etapa inicial de la progresión tumoral (iniciación), inestabilidad genética, susceptibilidad a la respuesta inmune del huésped, a la progresión y la respuesta a la terapia. La relación más estrecha, entre el microbioma y el cáncer, es a través de la desregulación del sistema inmune. En este trabajo revisamos las actuales evidencias sobre la asociación entre la microbiota y algunos tipos de cáncer como el cáncer gástrico, colorrectal, próstata, ovario, oral, pulmón y mama.
Collapse
Affiliation(s)
- Francisco Arvelo
- Centro de Biociencias, Fundación Instituto de Estudios Avanzados-IDEA, Caracas, Venezuela
| | - Felipe Sojo
- Centro de Biociencias, Fundación Instituto de Estudios Avanzados-IDEA, Caracas, Venezuela
| | - Carlos Cotte
- Laboratorio de Cultivo de Tejidos y Biología de Tumores, Instituto de Biología Experimental, Universidad Central de Venezuela, Caracas, Venezuela
| |
Collapse
|
60
|
Lopes GV, Ramires T, Kleinubing NR, Scheik LK, Fiorentini ÂM, Padilha da Silva W. Virulence factors of foodborne pathogen Campylobacterjejuni. Microb Pathog 2021; 161:105265. [PMID: 34699927 DOI: 10.1016/j.micpath.2021.105265] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/27/2021] [Accepted: 10/21/2021] [Indexed: 12/26/2022]
Abstract
Campylobacter jejuni is a highly frequent cause of gastrointestinal foodborne disease in humans throughout the world. Disease outcomes vary from mild to severe diarrhea, and in rare cases the Guillain-Barré syndrome or reactive arthritis can develop as a post-infection complication. Transmission to humans usually occurs via the consumption of a range of foods, especially those associated with the consumption of raw or undercooked poultry meat, unpasteurized milk, and water-based environmental sources. When associated to food or water ingestion, the C. jejuni enters the human host intestine via the oral route and colonizes the distal ileum and colon. When it adheres and colonizes the intestinal cell surfaces, the C. jejuni is expected to express several putative virulence factors, which cause damage to the intestine either directly, by cell invasion and/or production of toxin(s), or indirectly, by triggering inflammatory responses. This review article highlights various C. jejuni characteristics - such as motility and chemotaxis - that contribute to the biological fitness of the pathogen, as well as factors involved in human host cell adhesion and invasion, and their potential role in the development of the disease. We have analyzed and critically discussed nearly 180 scientific articles covering the latest improvements in the field.
Collapse
Affiliation(s)
- Graciela Volz Lopes
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Tassiana Ramires
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Natalie Rauber Kleinubing
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Letícia Klein Scheik
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Ângela Maria Fiorentini
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil
| | - Wladimir Padilha da Silva
- Departamento de Ciência e Tecnologia Agroindustrial, Faculdade de Agronomia Eliseu Maciel, Universidade Federal de Pelotas (UFPel), Caixa Postal 354, 96160-000, Pelotas, RS, Brazil.
| |
Collapse
|
61
|
Hull R, Lolas G, Makrogkikas S, Jensen LD, Syrigos KN, Evangelou G, Padayachy L, Egbor C, Mehrotra R, Makhafola TJ, Oyomno M, Dlamini Z. Microbiomics in Collusion with the Nervous System in Carcinogenesis: Diagnosis, Pathogenesis and Treatment. Microorganisms 2021; 9:2129. [PMID: 34683450 PMCID: PMC8538279 DOI: 10.3390/microorganisms9102129] [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: 08/19/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022] Open
Abstract
The influence of the naturally occurring population of microbes on various human diseases has been a topic of much recent interest. Not surprisingly, continuously growing attention is devoted to the existence of a gut brain axis, where the microbiota present in the gut can affect the nervous system through the release of metabolites, stimulation of the immune system, changing the permeability of the blood-brain barrier or activating the vagus nerves. Many of the methods that stimulate the nervous system can also lead to the development of cancer by manipulating pathways associated with the hallmarks of cancer. Moreover, neurogenesis or the creation of new nervous tissue, is associated with the development and progression of cancer in a similar manner as the blood and lymphatic systems. Finally, microbes can secrete neurotransmitters, which can stimulate cancer growth and development. In this review we discuss the latest evidence that support the importance of microbiota and peripheral nerves in cancer development and dissemination.
Collapse
Affiliation(s)
- Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
| | - Georgios Lolas
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Department of Medicine, National & Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.S.); (G.E.)
| | | | - Lasse D. Jensen
- Division of Cardiovascular Medicine, Department of Health, Medicine and Caring Sciences, Faculty of Medicine, Linköping University, 581 83 Linköping, Sweden;
| | - Konstantinos N. Syrigos
- Department of Medicine, National & Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.S.); (G.E.)
| | - George Evangelou
- Department of Medicine, National & Kapodistrian University of Athens, 11527 Athens, Greece; (K.N.S.); (G.E.)
| | - Llewellyn Padayachy
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Department of Neurosurgery, University of Pretoria, Hatfield 0028, South Africa
| | - Cyril Egbor
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Department of Neurosurgery, University of Pretoria, Hatfield 0028, South Africa
| | - Ravi Mehrotra
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Centre for Health Innovation and Policy (CHIP) Foundation, Noida 201301, India
- Datar Cancer Genetics, Nashik 422010, India
| | - Tshepiso Jan Makhafola
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9300, South Africa
| | - Meryl Oyomno
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
- Department of Surgery, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0007, South Africa
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.H.); (G.L.); (L.P.); (C.E.); (R.M.); (T.J.M.); (M.O.)
| |
Collapse
|
62
|
Huang PY, Yang YC, Wang CI, Hsiao PW, Chiang HI, Chen TW. Increase in Akkermansiaceae in Gut Microbiota of Prostate Cancer-Bearing Mice. Int J Mol Sci 2021; 22:9626. [PMID: 34502535 PMCID: PMC8431795 DOI: 10.3390/ijms22179626] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/18/2021] [Accepted: 09/03/2021] [Indexed: 01/04/2023] Open
Abstract
Gut microbiota are reported to be associated with many diseases, including cancers. Several bacterial taxa have been shown to be associated with cancer development or response to treatment. However, longitudinal microbiota alterations during the development of cancers are relatively unexplored. To better understand how microbiota changes, we profiled the gut microbiota composition from prostate cancer-bearing mice and control mice at five different time points. Distinct gut microbiota differences were found between cancer-bearing mice and control mice. Akkermansiaceae was found to be significantly higher in the first three weeks in cancer-bearing mice, which implies its role in the early stage of cancer colonization. We also found that Bifidobacteriaceae and Enterococcaceae were more abundant in the second and last sampling week, respectively. The increments of Akkermansiaceae, Bifidobacteriaceae and Enterococcaceae were previously found to be associated with responses to immunotherapy, which suggests links between these bacteria families and cancers. Additionally, our function analysis showed that the bacterial taxa carrying steroid biosynthesis and butirosin and neomycin biosynthesis were increased, whereas those carrying naphthalene degradation decreased in cancer-bearing mice. Our work identified the bacteria taxa altered during prostate cancer progression and provided a resource of longitudinal microbiota profiles during cancer development in a mouse model.
Collapse
Affiliation(s)
- Pin-Yu Huang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan;
| | - Yu-Chih Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Nangang District, Taipei City 115, Taiwan; (Y.-C.Y.); (P.-W.H.)
| | - Chun-I Wang
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Linkou 333, Taiwan;
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Nangang District, Taipei City 115, Taiwan; (Y.-C.Y.); (P.-W.H.)
| | - Hsin-I Chiang
- Department of Animal Science, National Chung Hsing University, Taichung 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Ting-Wen Chen
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| |
Collapse
|
63
|
Varon C, Azzi-Martin L, Khalid S, Seeneevassen L, Ménard A, Spuul P. Helicobacters and cancer, not only gastric cancer? Semin Cancer Biol 2021; 86:1138-1154. [PMID: 34425210 DOI: 10.1016/j.semcancer.2021.08.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
The Helicobacter genus actually comprises 46 validly published species divided into two main clades: gastric and enterohepatic Helicobacters. These bacteria colonize alternative sites of the digestive system in animals and humans, and contribute to inflammation and cancers. In humans, Helicobacter infection is mainly related to H. pylori, a gastric pathogen infecting more than half of the world's population, leading to chronic inflammation of the gastric mucosa that can evolve into two types of gastric cancers: gastric adenocarcinomas and gastric MALT lymphoma. In addition, H. pylori but also non-H. pylori Helicobacter infection has been associated with many extra-gastric malignancies. This review focuses on H. pylori and its role in gastric cancers and extra-gastric diseases, as well as malignancies induced by non-H. pylori Helicobacters. Their different virulence factors and their involvement in carcinogenesis is discussed. This review highlights the importance of both gastric and enterohepatic Helicobacters in gastrointestinal and liver cancers.
Collapse
Affiliation(s)
- Christine Varon
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Lamia Azzi-Martin
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France; Univ. Bordeaux, UFR des Sciences Médicales, Bordeaux, France
| | - Sadia Khalid
- Tallinn University of Technology, Department of Chemistry and Biotechnology, Akadeemia RD 15, 12618, Tallinn, Estonia
| | - Lornella Seeneevassen
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Armelle Ménard
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Pirjo Spuul
- Tallinn University of Technology, Department of Chemistry and Biotechnology, Akadeemia RD 15, 12618, Tallinn, Estonia.
| |
Collapse
|
64
|
Cytolethal distending toxin: from genotoxin to a potential biomarker and anti-tumor target. World J Microbiol Biotechnol 2021; 37:150. [PMID: 34379213 DOI: 10.1007/s11274-021-03117-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/31/2021] [Indexed: 10/20/2022]
Abstract
Cytolethal Distending Toxin (CDT) belongs to the AB toxin family and is produced by a plethora of Gram-negative bacteria. Eight human-affecting enteropathogens harbor CDT that causes irritable bowel syndrome (IBS), dysentery, chancroid, and periodontitis worldwide. They have a novel molecular mode of action as they interfere in the eukaryotic cell-cycle progression leading to G2/M arrest and apoptosis. CDT, the first bacterial genotoxin described, is encoded in a single operon possessing three proteins, CdtA, CdtB, and CdtC. CdtA and CdtC are needed for the binding of the CDT toxin complex to the cholesterol-rich lipid domains of the host cell while the CdtB is the active moiety. Sequence and 3D structural-based analysis of CdtB showed similarities with nucleases and phosphatases, it was hypothesized that CdtB exercises a biochemical function identical to both these enzymes. CDT is secreted through the outer membrane vesicles from the producing bacteria. It is internalized in the target cells via clathrin-dependent endocytosis and translocated to the host cell nucleus through the Golgi complex and ER. This study discusses the virulence role of CDT, causing pathogenicity by acting as a tri-perditious complex in the CDT-producing species with an emphasis on its potential role as a biomarker and an anti-tumor agent.
Collapse
|
65
|
Christensen H, Kuhnert P, Foster G, Bisgaard M. Reclassification of [ Haemophilus] haemoglobinophilus as Canicola haemoglobinophilus gen. nov., comb. nov. including Bisgaard taxon 35. Int J Syst Evol Microbiol 2021; 71. [PMID: 34264807 DOI: 10.1099/ijsem.0.004881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
[Haemophilus] haemoglobinophilus and the unpublished Bisgaard taxon 35 are associated with respiratory and urogenital tract infections in dogs. A total of 21 strains including the type strain of [Haemophilus] haemoglobinophilus were included in the investigation. Strains of [Haemophilus] haemoglobinophilus and taxon 35 formed a monophyletic group demonstrating at least 97.8 and 96.5% similarities within the group based upon 16S rRNA and rpoB gene sequence comparisons, respectively. Glaesserella australis was the most closely related species to [Haemophilus] haemoglobinophilus and taxon 35 with 96.1 % 16S rRNA gene sequence similarity which is slightly higher than the 95 % separating most genera of the family Pasteurellaceae. However, the conserved protein sequence phylogeny documented a unique position of [Haemophilus] haemoglobinophilus with only 81 % identity to the most closely related species, genomospecies 1 of the genus Rodentibacter which is lower than the 85 % separating most genera of the family Pasteurellaceae. The conserved protein sequence identity to Haemophilus influenzae, the type species of the genus, was 77%, demonstrating that [Haemophilus] haemoglobinophilus is not properly classified as a member of the genus Haemophilus. On the basis of the phylogenetic comparisons, the taxa [Haemophilus] haemoglobinophilus and taxon 35 are proposed to be included with a novel genus Canicola with one species, Canicola haemoglobinophilus which is reclassified from [Haemophilus] haemoglobinophilus. Phenotypic characters obtained with isolates genetically approved to represent Canicola haemoglobinophilus were in accordance with those of the members of the family Pasteurellaceae, and the novel genus can be separated from most of the existing genera by a positive catalase reaction, lack of V-factor requirement for growth, lack of haemolysis of blood agar and negative Voges-Proskauer and urease tests. The novel genus cannot be separated by biochemical and physiological characteristics alone from the genera Aggregatibacter, Avibacterium, Frederiksenia and Spirabiliibacterium. However, MALDI-TOF mass spectroscopy and also RpoB amino acid signatures allowed a clear separation from these taxa, supporting the existence of a novel genus. The DNA G+C content is 37.0-37.8 mol% for the genus, based on the whole genomic sequences. The type strain of Canicola haemoglobinophilus is CCUG 3714T (=ATCC 19416T=NCTC 1659T) isolated in 1901 from the prepuce of a dog in Germany.
Collapse
Affiliation(s)
- Henrik Christensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 4 Stigbojlen, DK-1870 Frederiksberg C, Denmark
| | - Peter Kuhnert
- Institute of Veterinary Bacteriology, Vetsuisse Faculty, University of Bern, Laenggass-Strasse 122, CH-3001 Bern, Switzerland
| | - Geoffrey Foster
- SRUC Veterinary Service, An Lochran 10, Inverness Campus, Inverness, UK
| | - Magne Bisgaard
- Professor emeritus, Bisgaard Consulting, 40 Horsevænget, DK-4130 Viby Sjælland, Denmark
| |
Collapse
|
66
|
Host Chromatin Regulators Required for Aggregatibacter actinomycetemcomitans Cytolethal Distending Toxin Activity in Saccharomyces cerevisiae Model. Infect Immun 2021; 89:e0003621. [PMID: 33941581 DOI: 10.1128/iai.00036-21] [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: 11/20/2022] Open
Abstract
Cytolethal distending toxin (CDT) is a bacterial genotoxin that causes host cell cycle arrest and death. We previously employed a Saccharomyces cerevisiae model with inducible expression of the CDT catalytic subunit from Aggregatibacter actinomycetemcomitans, AaCdtB, and showed that a wide variety of host factors play a role in facilitating the activity of CdtB. Our observation that a yeast H2B mutant defective in chromatin condensation was partially resistant to CdtB implies that chromatin structure may affect CDT function. In this study, we identified host chromatin regulatory genes required for CdtB cytotoxicity. We found that the deletion of HTZ1 or certain subunits of SWR, INO80, and SIR complexes increased cellular resistance to CdtB. We hypothesized that CdtB may interact with Htz1 or the chromatin, but immunoprecipitation experiments failed to detect physical interaction between CdtB and Htz1 or the chromatin. However, we observed reduced nuclear localization of CdtB in several mutants, suggesting that impaired nuclear translocation may, at least partly, explain the mechanisms of CdtB resistance. In addition, mutations in chromatin regulatory genes induce changes in the global gene expression profile, and these may indirectly affect CdtB toxicity. Our results suggest that decreased expression of endoplasmic reticulum (ER)-Golgi transport-related genes that may be involved in CdtB transport and/or increased expression of DNA repair genes may contribute to CdtB resistance. These results suggest that the functions of chromatin regulators may contribute to the activity of CDT in host cells.
Collapse
|
67
|
The Host Cellular Immune Response to Infection by Campylobacter Spp. and Its Role in Disease. Infect Immun 2021; 89:e0011621. [PMID: 34031129 DOI: 10.1128/iai.00116-21] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Campylobacter spp. are the leading cause of bacterium-derived gastroenteritis worldwide, impacting 96 million individuals annually. Unlike other bacterial pathogens of the gastrointestinal tract, Campylobacter spp. lack many of the classical virulence factors that are often associated with the ability to induce disease in humans, including an array of canonical secretion systems and toxins. Consequently, the clinical manifestations of human campylobacteriosis and its resulting gastrointestinal pathology are believed to be primarily due to the host immune response toward the bacterium. Further, while gastrointestinal infection is usually self-limiting, numerous postinfectious disorders can occur, including the development of Guillain-Barré syndrome, reactive arthritis, and irritable bowel syndrome. Because gastrointestinal disease likely results from the host immune response, the development of these postinfectious disorders may be due to dysregulation or misdirection of the same inflammatory response. As a result, it is becoming increasingly important to the Campylobacter field, and human health, that the cellular immune responses toward Campylobacter be better understood, including which immunological events are critical to the development of disease and the postinfectious disorders mentioned above. In this review, we collectively cover the cellular immune responses across susceptible hosts to Campylobacter jejuni infection, along with the tissue pathology and postinfectious disorders which may develop.
Collapse
|
68
|
Lopez Chiloeches M, Bergonzini A, Frisan T. Bacterial Toxins Are a Never-Ending Source of Surprises: From Natural Born Killers to Negotiators. Toxins (Basel) 2021; 13:426. [PMID: 34204481 PMCID: PMC8235270 DOI: 10.3390/toxins13060426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
The idea that bacterial toxins are not only killers but also execute more sophisticated roles during bacteria-host interactions by acting as negotiators has been highlighted in the past decades. Depending on the toxin, its cellular target and mode of action, the final regulatory outcome can be different. In this review, we have focused on two families of bacterial toxins: genotoxins and pore-forming toxins, which have different modes of action but share the ability to modulate the host's immune responses, independently of their capacity to directly kill immune cells. We have addressed their immuno-suppressive effects with the perspective that these may help bacteria to avoid clearance by the host's immune response and, concomitantly, limit detrimental immunopathology. These are optimal conditions for the establishment of a persistent infection, eventually promoting asymptomatic carriers. This immunomodulatory effect can be achieved with different strategies such as suppression of pro-inflammatory cytokines, re-polarization of the immune response from a pro-inflammatory to a tolerogenic state, and bacterial fitness modulation to favour tissue colonization while preventing bacteraemia. An imbalance in each of those effects can lead to disease due to either uncontrolled bacterial proliferation/invasion, immunopathology, or both.
Collapse
Affiliation(s)
| | | | - Teresa Frisan
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden; (M.L.C.); (A.B.)
| |
Collapse
|
69
|
Mathiasen SL, Gall-Mas L, Pateras IS, Theodorou SDP, Namini MRJ, Hansen MB, Martin OCB, Vadivel CK, Ntostoglou K, Butter D, Givskov M, Geisler C, Akbar AN, Gorgoulis VG, Frisan T, Ødum N, Krejsgaard T. Bacterial genotoxins induce T cell senescence. Cell Rep 2021; 35:109220. [PMID: 34107253 DOI: 10.1016/j.celrep.2021.109220] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 02/16/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022] Open
Abstract
Several types of pathogenic bacteria produce genotoxins that induce DNA damage in host cells. Accumulating evidence suggests that a central function of these genotoxins is to dysregulate the host's immune response, but the underlying mechanisms remain unclear. To address this issue, we investigated the effects of the most widely expressed bacterial genotoxin, the cytolethal distending toxin (CDT), on T cells-the key mediators of adaptive immunity. We show that CDT induces premature senescence in activated CD4 T cells in vitro and provide evidence suggesting that infection with genotoxin-producing bacteria promotes T cell senescence in vivo. Moreover, we demonstrate that genotoxin-induced senescent CD4 T cells assume a senescence-associated secretory phenotype (SASP) which, at least partly, is orchestrated by the ATM-p38 signaling axis. These findings provide insight into the immunomodulatory properties of bacterial genotoxins and uncover a putative link between bacterial infections and T cell senescence.
Collapse
Affiliation(s)
- Sarah L Mathiasen
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Laura Gall-Mas
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ioannis S Pateras
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Sofia D P Theodorou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Martin R J Namini
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Morten B Hansen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Océane C B Martin
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Chella Krishna Vadivel
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Konstantinos Ntostoglou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Deborah Butter
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Michael Givskov
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Carsten Geisler
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Arne N Akbar
- Division of Medicine, University College London, London WC1E 6JF, UK
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, 11527 Athens, Greece; Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9PL, UK
| | - Teresa Frisan
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Molecular Biology and Umeå Center for Microbial Research (UCMR), Umeå University, 90187 Umeå, Sweden
| | - Niels Ødum
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Thorbjørn Krejsgaard
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
70
|
Ren L, Ye J, Zhao B, Sun J, Cao P, Yang Y. The Role of Intestinal Microbiota in Colorectal Cancer. Front Pharmacol 2021; 12:674807. [PMID: 33959032 PMCID: PMC8093878 DOI: 10.3389/fphar.2021.674807] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer is a multifactorial disease involving genetic, environmental, and lifestyle risk factors. Intestinal microbiota plays an important role in the occurrence and development of colorectal cancer. Studies have shown that the behavior of intestinal microbiota can lead to pathological changes in the host intestine, which can be divided into epigenetic changes and carcinogenic changes at the gene level, and ultimately promote the formation and development of colorectal cancer. Intestinal microbiota is mainly distributed in the intestinal epithelium, which is composed of a large number of microorganisms interacting with the host intestinal cells. It can affect the immune-inflammation and metabolism of the gastrointestinal tract, and may be used as a biomarker for disease diagnosis. Regulation of gut microbiota is a promising strategy for the prevention and treatment of colorectal cancer. This article reviews the role of intestinal microbiota in the development of colorectal cancer, including the related mechanisms of intestinal microbiota promoting colorectal cancer, the use of intestinal microbiota in the diagnosis of colorectal cancer, and the regulation of intestinal microbiota in the prevention or treatment of colorectal cancer.
Collapse
Affiliation(s)
- Lingli Ren
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Juan Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bing Zhao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinbing Sun
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, Changshu, China
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,Yangtze River Pharmaceutical Group, Taizhou, China
| |
Collapse
|
71
|
Yuan L, Wang W, Zhang W, Zhang Y, Wei C, Li J, Zhou D. Gut Microbiota in Untreated Diffuse Large B Cell Lymphoma Patients. Front Microbiol 2021; 12:646361. [PMID: 33927704 PMCID: PMC8076791 DOI: 10.3389/fmicb.2021.646361] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal microecology plays an important role in the development and progression of hematological malignancies. However, characteristics of gut microbiota in diffuse large B cell lymphoma (DLBCL) have not been reported. The microbiota composition of fecal samples from 25 untreated DLBCL patients and 26 healthy volunteers was examined by 16S rRNA gene sequencing. On α-diversity analysis, there was no significant difference in species diversity and abundance between the two groups. However, a significant difference was observed on β-diversity analysis. The intestinal microbiota in patients with DLBCL showed a continuous evolutionary relationship, which progressed from phylum, proteobacteria, to genus, Escherichia-Shigella. Their abundance was significantly higher than that of the control group. At the genus level, Allisonella, lachnospira, and Roseburia were more abundant in patients with DLBCL than in the control group. Functional prediction by PICRUSt indicated that thiamine metabolism and phenylalanine, tyrosine, and tryptophan biosynthesis were significantly lower in the DLBCL group than in the control group. In conclusion, our results clearly demonstrate that the gut microbiota was changed significantly in DLBCL. The study highlights fundamental differences in the microbial diversity and composition of patients with DLBCL and paves the way for future prospective studies and microbiome-directed interventional trials to improve patient outcomes.
Collapse
Affiliation(s)
- Li Yuan
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Wang
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Zhang
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Zhang
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chong Wei
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Daobin Zhou
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
72
|
González‐Sánchez P, DeNicola GM. The microbiome(s) and cancer: know thy neighbor(s). J Pathol 2021; 254:332-343. [DOI: 10.1002/path.5661] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Paloma González‐Sánchez
- Department of Cancer Physiology H. Lee Moffitt Cancer Center and Research Institute Tampa FL USA
| | - Gina M DeNicola
- Department of Cancer Physiology H. Lee Moffitt Cancer Center and Research Institute Tampa FL USA
| |
Collapse
|
73
|
Yeh JY, Lin HJ, Kuo CJ, Feng CL, Chou CH, Lin CD, Wu HY, Li CY, Chiu CH, Lai CH. Campylobacter jejuni Cytolethal Distending Toxin C Exploits Lipid Rafts to Mitigate Helicobacter pylori-Induced Pathogenesis. Front Cell Dev Biol 2021; 8:617419. [PMID: 33708766 PMCID: PMC7940356 DOI: 10.3389/fcell.2020.617419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/28/2020] [Indexed: 12/15/2022] Open
Abstract
Helicobacter pylori infection is associated with several gastrointestinal diseases, including gastritis, peptic ulcer, and gastrointestinal adenocarcinoma. Two major cytotoxins, vacuolating cytotoxin A (VacA) and cytotoxin-associated gene A (CagA), interact closely with lipid rafts, contributing to H. pylori-associated disease progression. The Campylobacter jejuni cytolethal distending toxin consists of three subunits: CdtA, CdtB, and CdtC. Among them, CdtA and CdtC bind to membrane lipid rafts, which is crucial for CdtB entry into cells. In this study, we employed recombinant CdtC (rCdtC) to antagonize the functions of H. pylori cytotoxin in cells. Our results showed that rCdtC alleviates cell vacuolation induced by H. pylori VacA. Furthermore, rCdtC reduces H. pylori CagA translocation, which decreases nuclear factor kappa-B activation and interleukin-8 production, resulting in the mitigation of gastric epithelial cell inflammation. These results reveal that CdtC hijacks cholesterol to compete for H. pylori cytotoxin actions via lipid rafts, ameliorating H. pylori-induced pathogenesis.
Collapse
Affiliation(s)
- Jia-Yin Yeh
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hwai-Jeng Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang-Ho Hospital, New Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Jung Kuo
- Chang Gung Microbiota Therapy Center, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Lung Feng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hsinchu Hospital, Hsinchu, Taiwan.,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chia-Huei Chou
- Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Infectious Disease, Department of Otolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Der Lin
- Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Infectious Disease, Department of Otolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Hui-Yu Wu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chen-Yi Li
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
74
|
Chen YA, Lai YR, Wu HY, Lo YJ, Chang YF, Hung CL, Lin CJ, Lo UG, Lin H, Hsieh JT, Chiu CH, Lin YH, Lai CH. Bacterial Genotoxin-Coated Nanoparticles for Radiotherapy Sensitization in Prostate Cancer. Biomedicines 2021; 9:biomedicines9020151. [PMID: 33557143 PMCID: PMC7913852 DOI: 10.3390/biomedicines9020151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer (PCa) is one of the most commonly diagnosed cancers in men and usually becomes refractory because of recurrence and metastasis. CD44, a transmembrane glycoprotein, serves as a receptor for hyaluronic acid (HA). It has been found to be abundantly expressed in cancer stem cells (CSCs) that often exhibit a radioresistant phenotype. Cytolethal distending toxin (CDT), produced by Campylobacter jejuni, is a tripartite genotoxin composed of CdtA, CdtB, and CdtC subunits. Among the three, CdtB acts as a type I deoxyribonuclease (DNase I), which creates DNA double-strand breaks (DSBs). Nanoparticles loaded with antitumor drugs and specific ligands that recognize cancerous cell receptors are promising methods to overcome the therapeutic challenges. In this study, HA-decorated nanoparticle-encapsulated CdtB (HA-CdtB-NPs) were prepared and their targeted therapeutic activity in radioresistant PCa cells was evaluated. Our results showed that HA-CdtB-NPs sensitized radioresistant PCa cells by enhancing DSB and causing G2/M cell-cycle arrest, without affecting the normal prostate epithelial cells. HA-CdtB-NPs possess maximum target specificity and delivery efficiency of CdtB into the nucleus and enhance the effect of radiation in radioresistant PCa cells. These findings demonstrate that HA-CdtB-NPs exert target specificity accompanied with radiomimetic activity and can be developed as an effective strategy against radioresistant PCa.
Collapse
Affiliation(s)
- Yu-An Chen
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-A.C.); (Y.-R.L.); (H.-Y.W.); (Y.-J.L.); (Y.-F.C.)
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan;
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (C.-J.L.); (U.-G.L.); (J.-T.H.)
| | - Yi-Ru Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-A.C.); (Y.-R.L.); (H.-Y.W.); (Y.-J.L.); (Y.-F.C.)
| | - Hui-Yu Wu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-A.C.); (Y.-R.L.); (H.-Y.W.); (Y.-J.L.); (Y.-F.C.)
| | - Yen-Ju Lo
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-A.C.); (Y.-R.L.); (H.-Y.W.); (Y.-J.L.); (Y.-F.C.)
| | - Yu-Fang Chang
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-A.C.); (Y.-R.L.); (H.-Y.W.); (Y.-J.L.); (Y.-F.C.)
| | - Chiu-Lien Hung
- Targeted Drug and Delivery Technology Division, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 30011, Taiwan;
| | - Chun-Jung Lin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (C.-J.L.); (U.-G.L.); (J.-T.H.)
| | - U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (C.-J.L.); (U.-G.L.); (J.-T.H.)
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan;
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (C.-J.L.); (U.-G.L.); (J.-T.H.)
- Department of Medical Research, School of Medicine, China Medical University and Hospital, Taichung 40447, Taiwan
| | - Cheng-Hsun Chiu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-A.C.); (Y.-R.L.); (H.-Y.W.); (Y.-J.L.); (Y.-F.C.)
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou 33305, Taiwan
- Correspondence: (C.-H.C.); (Y.-H.L.); (C.-H.L.)
| | - Yu-Hsin Lin
- Department of Medical Research, School of Medicine, China Medical University and Hospital, Taichung 40447, Taiwan
- Center for Advanced Pharmaceutics and Drug Delivery Research, Department and Institute of Pharmacology, Institute of Biopharmaceutical Sciences, Faculty of Pharmacy, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Correspondence: (C.-H.C.); (Y.-H.L.); (C.-H.L.)
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-A.C.); (Y.-R.L.); (H.-Y.W.); (Y.-J.L.); (Y.-F.C.)
- Department of Medical Research, School of Medicine, China Medical University and Hospital, Taichung 40447, Taiwan
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou 33305, Taiwan
- Department of Nursing, Asia University, Taichung 41354, Taiwan
- Correspondence: (C.-H.C.); (Y.-H.L.); (C.-H.L.)
| |
Collapse
|
75
|
Paulson AR, O’Callaghan M, Zhang XX, Rainey PB, Hurst MRH. In vivo transcriptome analysis provides insights into host-dependent expression of virulence factors by Yersinia entomophaga MH96, during infection of Galleria mellonella. G3 (BETHESDA, MD.) 2021; 11:jkaa024. [PMID: 33561230 PMCID: PMC7849909 DOI: 10.1093/g3journal/jkaa024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/19/2020] [Indexed: 12/31/2022]
Abstract
The function of microbes can be inferred from knowledge of genes specifically expressed in natural environments. Here, we report the in vivo transcriptome of the entomopathogenic bacterium Yersinia entomophaga MH96, captured during initial, septicemic, and pre-cadaveric stages of intrahemocoelic infection in Galleria mellonella. A total of 1285 genes were significantly upregulated by MH96 during infection; 829 genes responded to in vivo conditions during at least one stage of infection, 289 responded during two stages of infection, and 167 transcripts responded throughout all three stages of infection compared to in vitro conditions at equivalent cell densities. Genes upregulated during the earliest infection stage included components of the insecticidal toxin complex Yen-TC (chi1, chi2, and yenC1), genes for rearrangement hotspot element containing protein yenC3, cytolethal distending toxin cdtAB, and vegetative insecticidal toxin vip2. Genes more highly expressed throughout the infection cycle included the putative heat-stable enterotoxin yenT and three adhesins (usher-chaperone fimbria, filamentous hemagglutinin, and an AidA-like secreted adhesin). Clustering and functional enrichment of gene expression data also revealed expression of genes encoding type III and VI secretion system-associated effectors. Together these data provide insight into the pathobiology of MH96 and serve as an important resource supporting efforts to identify novel insecticidal agents.
Collapse
Affiliation(s)
- Amber R Paulson
- Forage Science, AgResearch Ltd., Lincoln 8140, New Zealand
- New Zealand Institute for Advanced Study, Massey University, Auckland 0745, New Zealand
- Department of Biology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | | | - Xue-Xian Zhang
- School of Natural and Computational Sciences, Massey University, Auckland 0745, New Zealand
| | - Paul B Rainey
- New Zealand Institute for Advanced Study, Massey University, Auckland 0745, New Zealand
- Laboratoire de Génétique de l’Evolution CBI, ESPCI Paris, Université PSL, CNRS, Paris 75005, France
- Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön 24306, Germany
| | - Mark R H Hurst
- Forage Science, AgResearch Ltd., Lincoln 8140, New Zealand
| |
Collapse
|
76
|
Benedetti F, Curreli S, Gallo RC, Zella D. Tampering of Viruses and Bacteria with Host DNA Repair: Implications for Cellular Transformation. Cancers (Basel) 2021; 13:E241. [PMID: 33440726 PMCID: PMC7826954 DOI: 10.3390/cancers13020241] [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: 12/23/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
A reduced ability to properly repair DNA is linked to a variety of human diseases, which in almost all cases is associated with an increased probability of the development of cellular transformation and cancer. DNA damage, that ultimately can lead to mutations and genomic instability, is due to many factors, such as oxidative stress, metabolic disorders, viral and microbial pathogens, excess cellular proliferation and chemical factors. In this review, we examine the evidence connecting DNA damage and the mechanisms that viruses and bacteria have evolved to hamper the pathways dedicated to maintaining the integrity of genetic information, thus affecting the ability of their hosts to repair the damage(s). Uncovering new links between these important aspects of cancer biology might lead to the development of new targeted therapies in DNA-repair deficient cancers and improving the efficacy of existing therapies. Here we provide a comprehensive summary detailing the major mechanisms that viruses and bacteria associated with cancer employ to interfere with mechanisms of DNA repair. Comparing these mechanisms could ultimately help provide a common framework to better understand how certain microorganisms are involved in cellular transformation.
Collapse
Affiliation(s)
- Francesca Benedetti
- Institute of Human Virology and Global Virus Network Center, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Sabrina Curreli
- Institute of Human Virology and Global Virus Network Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.C.); (R.C.G.)
| | - Robert C. Gallo
- Institute of Human Virology and Global Virus Network Center, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.C.); (R.C.G.)
| | - Davide Zella
- Institute of Human Virology and Global Virus Network Center, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| |
Collapse
|
77
|
Samanta S. Potential Impacts of Prebiotics and Probiotics in Cancer Prevention. Anticancer Agents Med Chem 2020; 22:605-628. [PMID: 33305713 DOI: 10.2174/1871520621999201210220442] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/15/2020] [Accepted: 10/26/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cancer is a serious problem throughout the world. The pathophysiology of cancer is multifactorial and is also related to gut microbiota. Intestinal microbes are the useful resident of the healthy human. They play various aspects of human health including nutritional biotransformation, flushing of the pathogens, toxin neutralization, immune response, and onco-suppression. Disruption in the interactions among the gut microbiota, intestinal epithelium, and the host immune system are associated with gastrointestinal disorders, neurodegenerative diseases, metabolic syndrome, and cancer. Probiotic bacteria (Lactobacillus spp., Bifidobacterium spp.) have been regarded as beneficial to health and shown to play a significant role in immunomodulation and displayed preventive role against obesity, diabetes, liver disease, inflammatory bowel disease, tumor progression, and cancer. OBJECTIVE The involvement of gut microorganisms in cancer development and prevention has been recognized as a balancing factor. The events of dysbiosis emphasize metabolic disorder and carcinogenesis. The gut flora potentiates immunomodulation and minimizes the limitations of usual chemotherapy. The significant role of prebiotics and probiotics on the improvement of immunomodulation and antitumor properties has been considered. METHODS I had reviewed the literature on the multidimensional activities of prebiotics and probiotics from the NCBI website database PubMed, Springer Nature, Science Direct (Elsevier), Google Scholar database to search relevant articles. Specifically, I had focused on the role of prebiotics and probiotics in immunomodulation and cancer prevention. RESULTS Prebiotics are the nondigestible fermentable sugars that selectively influence the growth of probiotic organisms that exert immunomodulation over the cancerous growth. The oncostatic properties of bacteria are mediated through the recruitment of cytotoxic T cells, natural killer cells, and oxidative stress-induced apoptosis in the tumor microenvironment. Moreover, approaches have also been taken to use probiotics as an adjuvant in cancer therapy. CONCLUSION The present review has indicated that dysbiosis is the crucial factor in many pathological situations including cancer. Applications of prebiotics and probiotics exhibit the immune-surveillance as oncostatic effects. These events increase the possibilities of new therapeutic strategies for cancer prevention.
Collapse
Affiliation(s)
- Saptadip Samanta
- Department of Physiology, Midnapore College, Midnapore, Paschim Medinipur, 721101, West Bengal,. India
| |
Collapse
|
78
|
Saha C, Horst-Kreft D, Kross I, van der Spek PJ, Louwen R, van Baarlen P. Campylobacter jejuni Cas9 Modulates the Transcriptome in Caco-2 Intestinal Epithelial Cells. Genes (Basel) 2020; 11:genes11101193. [PMID: 33066557 PMCID: PMC7650535 DOI: 10.3390/genes11101193] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/01/2020] [Accepted: 10/11/2020] [Indexed: 12/12/2022] Open
Abstract
The zoonotic human pathogen Campylobacter jejuni is known for its ability to induce DNA-damage and cell death pathology in humans. The molecular mechanism behind this phenomenon involves nuclear translocation by Cas9, a nuclease in C. jejuni (CjeCas9) that is the molecular marker of the Type II CRISPR-Cas system. However, it is unknown via which cellular pathways CjeCas9 drives human intestinal epithelial cells into cell death. Here, we show that CjeCas9 released by C. jejuni during the infection of Caco-2 human intestinal epithelial cells directly modulates Caco-2 transcriptomes during the first four hours of infection. Specifically, our results reveal that CjeCas9 activates DNA damage (p53, ATM (Ataxia Telangiectasia Mutated Protein)), pro-inflammatory (NF-κB (Nuclear factor-κB)) signaling and cell death pathways, driving Caco-2 cells infected by wild-type C. jejuni, but not when infected by a cas9 deletion mutant, towards programmed cell death. This work corroborates our previous finding that CjeCas9 is cytotoxic and highlights on a RNA level the basal cellular pathways that are modulated.
Collapse
Affiliation(s)
- Chinmoy Saha
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (D.H.-K.); (I.K.); (R.L.)
- Correspondence: ; Tel.: +31-638620563
| | - Deborah Horst-Kreft
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (D.H.-K.); (I.K.); (R.L.)
| | - Inez Kross
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (D.H.-K.); (I.K.); (R.L.)
| | - Peter J. van der Spek
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - Rogier Louwen
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (D.H.-K.); (I.K.); (R.L.)
| | - Peter van Baarlen
- Host–Microbe Interactomics, Wageningen University and Research, 6708 WD Wageningen, The Netherlands;
| |
Collapse
|
79
|
Pons BJ, Loiseau N, Hashim S, Tadrist S, Mirey G, Vignard J. Functional Study of Haemophilus ducreyi Cytolethal Distending Toxin Subunit B. Toxins (Basel) 2020; 12:toxins12090530. [PMID: 32825080 PMCID: PMC7551728 DOI: 10.3390/toxins12090530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
The Cytolethal Distending Toxin (CDT) is produced by many Gram-negative pathogenic bacteria responsible for major foodborne diseases worldwide. CDT induces DNA damage and cell cycle arrest in host-cells, eventually leading to senescence or apoptosis. According to structural and sequence comparison, the catalytic subunit CdtB is suggested to possess both nuclease and phosphatase activities, carried by a single catalytic site. However, the impact of each activity on cell-host toxicity is yet to be characterized. Here, we analyze the consequences of cell exposure to different CDT mutated on key CdtB residues, focusing on cell viability, cell cycle defects, and DNA damage induction. A first class of mutant, devoid of any activity, targets putative catalytic (H160A), metal binding (D273R), and DNA binding residues (R117A-R144A-N201A). The second class of mutants (A163R, F156-T158, and the newly identified G114T), which gathers mutations on residues potentially involved in lipid substrate binding, has only partially lost its toxic effects. However, their defects are alleviated when CdtB is artificially introduced inside cells, except for the F156-T158 double mutant that is defective in nuclear addressing. Therefore, our data reveal that CDT toxicity is mainly correlated to CdtB nuclease activity, whereas phosphatase activity may probably be involved in CdtB intracellular trafficking.
Collapse
Affiliation(s)
| | | | | | | | - Gladys Mirey
- Correspondence: (G.M.); (J.V.); Tel.: +33-582-066-338 (G.M.)
| | - Julien Vignard
- Correspondence: (G.M.); (J.V.); Tel.: +33-582-066-338 (G.M.)
| |
Collapse
|
80
|
Chen H, Zhang M, Hochstrasser M. The Biochemistry of Cytoplasmic Incompatibility Caused by Endosymbiotic Bacteria. Genes (Basel) 2020; 11:genes11080852. [PMID: 32722516 PMCID: PMC7465683 DOI: 10.3390/genes11080852] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/29/2022] Open
Abstract
Many species of arthropods carry maternally inherited bacterial endosymbionts that can influence host sexual reproduction to benefit the bacterium. The most well-known of such reproductive parasites is Wolbachia pipientis. Wolbachia are obligate intracellular α-proteobacteria found in nearly half of all arthropod species. This success has been attributed in part to their ability to manipulate host reproduction to favor infected females. Cytoplasmic incompatibility (CI), a phenomenon wherein Wolbachia infection renders males sterile when they mate with uninfected females, but not infected females (the rescue mating), appears to be the most common. CI provides a reproductive advantage to infected females in the presence of a threshold level of infected males. The molecular mechanisms of CI and other reproductive manipulations, such as male killing, parthenogenesis, and feminization, have remained mysterious for many decades. It had been proposed by Werren more than two decades ago that CI is caused by a Wolbachia-mediated sperm modification and that rescue is achieved by a Wolbachia-encoded rescue factor in the infected egg. In the past few years, new research has highlighted a set of syntenic Wolbachia gene pairs encoding CI-inducing factors (Cifs) as the key players for the induction of CI and its rescue. Within each Cif pair, the protein encoded by the upstream gene is denoted A and the downstream gene B. To date, two types of Cifs have been characterized based on the enzymatic activity identified in the B protein of each protein pair; one type encodes a deubiquitylase (thus named CI-inducing deubiquitylase or cid), and a second type encodes a nuclease (named CI-inducing nuclease or cin). The CidA and CinA proteins bind tightly and specifically to their respective CidB and CinB partners. In transgenic Drosophila melanogaster, the expression of either the Cid or Cin protein pair in the male germline induces CI and the expression of the cognate A protein in females is sufficient for rescue. With the identity of the Wolbachia CI induction and rescue factors now known, research in the field has turned to directed studies on the molecular mechanisms of CI, which we review here.
Collapse
Affiliation(s)
- Hongli Chen
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06511, USA; (H.C.); (M.Z.)
| | - Mengwen Zhang
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06511, USA; (H.C.); (M.Z.)
- Department of Chemistry, Yale University, New Haven, CT 06511, USA
| | - Mark Hochstrasser
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06511, USA; (H.C.); (M.Z.)
- Department of Molecular, Cellular, & Developmental Biology, Yale University, New Haven, CT 06511, USA
- Correspondence:
| |
Collapse
|
81
|
Saha C, Mohanraju P, Stubbs A, Dugar G, Hoogstrate Y, Kremers GJ, van Cappellen WA, Horst-Kreft D, Laffeber C, Lebbink JH, Bruens S, Gaskin D, Beerens D, Klunder M, Joosten R, Demmers JAA, van Gent D, Mouton JW, van der Spek PJ, van der Oost J, van Baarlen P, Louwen R. Guide-free Cas9 from pathogenic Campylobacter jejuni bacteria causes severe damage to DNA. SCIENCE ADVANCES 2020; 6:eaaz4849. [PMID: 32596446 PMCID: PMC7299616 DOI: 10.1126/sciadv.aaz4849] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 05/06/2020] [Indexed: 05/11/2023]
Abstract
CRISPR-Cas9 systems are enriched in human pathogenic bacteria and have been linked to cytotoxicity by an unknown mechanism. Here, we show that upon infection of human cells, Campylobacter jejuni secretes its Cas9 (CjeCas9) nuclease into their cytoplasm. Next, a native nuclear localization signal enables CjeCas9 nuclear entry, where it catalyzes metal-dependent nonspecific DNA cleavage leading to cell death. Compared to CjeCas9, native Cas9 of Streptococcus pyogenes (SpyCas9) is more suitable for guide-dependent editing. However, in human cells, native SpyCas9 may still cause some DNA damage, most likely because of its ssDNA cleavage activity. This side effect can be completely prevented by saturation of SpyCas9 with an appropriate guide RNA, which is only partially effective for CjeCas9. We conclude that CjeCas9 plays an active role in attacking human cells rather than in viral defense. Moreover, these unique catalytic features may therefore make CjeCas9 less suitable for genome editing applications.
Collapse
Affiliation(s)
- Chinmoy Saha
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - Andrew Stubbs
- Clinical Bioinformatics, Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Gaurav Dugar
- Institute of Molecular Infection Biology (IMIB)/Research Center for Infectious Diseases (ZINF), University of Würzburg, Würzburg, Germany
| | - Youri Hoogstrate
- Clinical Bioinformatics, Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Gert-Jan Kremers
- Optical Imaging Center, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - Deborah Horst-Kreft
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Charlie Laffeber
- Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Joyce H.G. Lebbink
- Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Radiation Oncology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Serena Bruens
- Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Duncan Gaskin
- Institute of Food Research, Gut Health and Food Safety Programme, Norwich Research Park, Norwich, UK
| | - Dior Beerens
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Maarten Klunder
- Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Rob Joosten
- Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Jeroen A. A. Demmers
- Proteomics Center, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Dik van Gent
- Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Johan W. Mouton
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Peter J. van der Spek
- Clinical Bioinformatics, Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - John van der Oost
- Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Peter van Baarlen
- Host-Microbe Interactomics Group, University of Wageningen, Wageningen, Netherlands
| | - Rogier Louwen
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
82
|
Rad AH, Aghebati-Maleki L, Kafil HS, Abbasi A. Molecular mechanisms of postbiotics in colorectal cancer prevention and treatment. Crit Rev Food Sci Nutr 2020; 61:1787-1803. [PMID: 32410512 DOI: 10.1080/10408398.2020.1765310] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The occurrence of colorectal cancer (CRC) has been rising expeditiously and anticipated that 2.4 million new occasions of CRC will be detected yearly around the world until the year 2035. Due to some side-effects and complications of conventional CRC therapies, bioactive components such as microbial-derived biomolecules (postbiotics) have been attaining great significance by researchers for adjuvant therapy in CRC patients. The term 'postbiotics' encompasses an extensive range of complex micro- and macro-molecules (<50, 50-100, and 100< kDa) such as inactivated microbial cells, cell fractions or metabolites, which confer various physiological health benefits to the host when administered in adequate amounts. Postbiotics modulate the composition of the gut microbiota and the functionality of the immune system, as well as promote the CRC treatment effectiveness and reduces its side-effects in CRC patients due to possessing anti-oxidant, anti-proliferative, anti-inflammatory, and anti-cancer activities. Presently scientific literature confirms that postbiotics with their unique characteristics in terms of clinical (safe origin), technological (stability), and economic (low production costs) aspects can be used as promising tools for both prevent and adjuvant treat strategies in CRC patients without any serious undesirable side-effects. This review provides an overview of the concept and safety issues regarding postbiotics, with emphasis on their biological role in the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Aziz Homayouni Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Abbasi
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student's research committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
83
|
Soleimanpour S, Hasanian SM, Avan A, Yaghoubi A, Khazaei M. Bacteriotherapy in gastrointestinal cancer. Life Sci 2020; 254:117754. [PMID: 32389833 DOI: 10.1016/j.lfs.2020.117754] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 01/13/2023]
Abstract
The most prevalent gastrointestinal (GI) cancers include colorectal cancer, stomach cancer, and liver cancer, known as the most common causes of cancer-related death in both men and women populations in the world. Traditional therapeutic approaches, including surgery, radiotherapy, and chemotherapy have failed in the effective treatment of cancer. Therefore, there is an urgent need for finding new effective anticancer agents. The available evidence and also the promising results of using bacteria as the anticancer agents on numerous cancer cell lines have attracted the attention of scientists for the therapeutic role of bacteria in the field of cancer therapy. Moreover, several studies on the bacteriotherapy agents have used genetic engineering to overcome the challenges and enhance the efficacy with the least drawbacks. Numerous bacterial species that can specifically target and internalize into the tumor cells are used live, attenuated, or genetically as compared to selectively consider the hypoxic condition of tumor, which results in the tumor suppression. The present study is a comprehensive review of the current literature on the use of bacteria and their substances such as bacteriocins and toxins in the treatment of different types of gastrointestinal cancers.
Collapse
Affiliation(s)
- Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hasanian
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atieh Yaghoubi
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
84
|
Silbergleit M, Vasquez AA, Miller CJ, Sun J, Kato I. Oral and intestinal bacterial exotoxins: Potential linked to carcinogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 171:131-193. [PMID: 32475520 DOI: 10.1016/bs.pmbts.2020.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Growing evidence suggests that imbalances in resident microbes (dysbiosis) can promote chronic inflammation, immune-subversion, and production of carcinogenic metabolites, thus leading to neoplasia. Yet, evidence to support a direct link of individual bacteria species to human sporadic cancer is still limited. This chapter focuses on several emerging bacterial toxins that have recently been characterized for their potential oncogenic properties toward human orodigestive cancer and the presence of which in human tissue samples has been documented. These include cytolethal distending toxins produced by various members of gamma and epsilon Proteobacteria, Dentilisin from mammalian oral Treponema, Pasteurella multocida toxin, two Fusobacterial toxins, FadA and Fap2, Bacteroides fragilis toxin, colibactin, cytotoxic necrotizing factors and α-hemolysin from Escherichia coli, and Salmonella enterica AvrA. It was clear that these bacterial toxins have biological activities to induce several hallmarks of cancer. Some toxins directly interact with DNA or chromosomes leading to their breakdowns, causing mutations and genome instability, and others modulate cell proliferation, replication and death and facilitate immune evasion and tumor invasion, prying specific oncogene and tumor suppressor pathways, such as p53 and β-catenin/Wnt. In addition, most bacterial toxins control tumor-promoting inflammation in complex and diverse mechanisms. Despite growing laboratory evidence to support oncogenic potential of selected bacterial toxins, we need more direct evidence from human studies and mechanistic data from physiologically relevant experimental animal models, which can reflect chronic infection in vivo, as well as take bacterial-bacterial interactions among microbiome into consideration.
Collapse
Affiliation(s)
| | - Adrian A Vasquez
- Department of Civil and Environmental Engineering, Wayne State University, Healthy Urban Waters, Detroit, MI, United States
| | - Carol J Miller
- Department of Civil and Environmental Engineering, Wayne State University, Healthy Urban Waters, Detroit, MI, United States
| | - Jun Sun
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Ikuko Kato
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States; Department of Pathology, Wayne State University School of Medicine, Detroit, MI, United States.
| |
Collapse
|
85
|
Virulence Traits of Inpatient Campylobacter jejuni Isolates, and a Transcriptomic Approach to Identify Potential Genes Maintaining Intracellular Survival. Microorganisms 2020; 8:microorganisms8040531. [PMID: 32272707 PMCID: PMC7232156 DOI: 10.3390/microorganisms8040531] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
There are still major gaps in our understanding of the bacterial factors that influence the outcomes of human Campylobacter jejuni infection. The aim of this study was to compare the virulence-associated features of 192 human C. jejuni strains isolated from hospitalized patients with diarrhoea (150/192, 78.1%), bloody diarrhoea (23/192, 11.9%), gastroenteritis (3/192, 1.6%), ulcerative colitis (3/192, 1.5%), and stomach ache (2/192, 1.0%). Traits were analysed with genotypic and phenotypic methods, including PCR and extracellular matrix protein (ECMP) binding, adhesion, and invasion capacities. Results were studied alongside patient symptoms, but no distinct links with them could be determined. Since the capacity of C. jejuni to invade host epithelial cells is one of its most enigmatic attributes, a high throughput transcriptomic analysis was performed in the third hour of internalization with a C. jejuni strain originally isolated from bloody diarrhoea. Characteristic groups of genes were significantly upregulated, outlining a survival strategy of internalized C. jejuni comprising genes related (1) to oxidative stress; (2) to a protective sheath formed by the capsule, LOS, N-, and O- glycosylation systems; (3) to dynamic metabolic activity supported by different translocases and the membrane-integrated component of the flagellar apparatus; and (4) to hitherto unknown genes.
Collapse
|
86
|
Barrett M, Hand CK, Shanahan F, Murphy T, O'Toole PW. Mutagenesis by Microbe: the Role of the Microbiota in Shaping the Cancer Genome. Trends Cancer 2020; 6:277-287. [PMID: 32209443 DOI: 10.1016/j.trecan.2020.01.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Cancers arise through the process of somatic evolution fueled by the inception of somatic mutations. We lack a complete understanding of the sources of these somatic mutations. Humans host a vast repertoire of microbes collectively known as the microbiota. The microbiota plays a role in altering the tumor microenvironment and proliferation. In addition, microbes have been shown to elicit DNA damage which provides the driver for somatic mutations. An understanding of microbiota-driven mutational mechanisms would contribute to a more complete understanding of the origins of the cancer genome. Here, we review the modes by which microbes stimulate DNA damage and the effect of these phenomena upon the cancer genomic architecture, specifically in the form of mutational spectra and mutational signatures.
Collapse
Affiliation(s)
- Maurice Barrett
- APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland; School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland
| | - Collette K Hand
- Department of Pathology, University College Cork, Cork, Ireland
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland; Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
| | - Thomas Murphy
- Department of Surgery, Mercy University Hospital, Cork, Ireland
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland; School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland.
| |
Collapse
|
87
|
Verster KI, Wisecaver JH, Karageorgi M, Duncan RP, Gloss AD, Armstrong EE, Price DK, Menon AR, Ali ZM, Whiteman NK. Horizontal Transfer of Bacterial Cytolethal Distending Toxin B Genes to Insects. Mol Biol Evol 2020; 36:2105-2110. [PMID: 31236589 DOI: 10.1093/molbev/msz146] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Horizontal gene transfer events have played a major role in the evolution of microbial species, but their importance in animals is less clear. Here, we report horizontal gene transfer of cytolethal distending toxin B (cdtB), prokaryotic genes encoding eukaryote-targeting DNase I toxins, into the genomes of vinegar flies (Diptera: Drosophilidae) and aphids (Hemiptera: Aphididae). We found insect-encoded cdtB genes are most closely related to orthologs from bacteriophage that infect Candidatus Hamiltonella defensa, a bacterial mutualistic symbiont of aphids that confers resistance to parasitoid wasps. In drosophilids, cdtB orthologs are highly expressed during the parasitoid-prone larval stage and encode a protein with ancestral DNase activity. We show that cdtB has been domesticated by diverse insects and hypothesize that it functions in defense against their natural enemies.
Collapse
Affiliation(s)
- Kirsten I Verster
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA
| | | | - Marianthi Karageorgi
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA
| | - Rebecca P Duncan
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA
| | - Andrew D Gloss
- Department of Ecology and Evolution, University of Chicago, Chicago, IL
| | | | - Donald K Price
- School of Life Sciences, University of Nevada, Las Vegas, NV
| | - Aruna R Menon
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA
| | - Zainab M Ali
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA
| | - Noah K Whiteman
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
88
|
The Cell-Cycle Regulatory Protein p21 CIP1/WAF1 Is Required for Cytolethal Distending Toxin (Cdt)-Induced Apoptosis. Pathogens 2020; 9:pathogens9010038. [PMID: 31906446 PMCID: PMC7168616 DOI: 10.3390/pathogens9010038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/22/2019] [Accepted: 12/28/2019] [Indexed: 12/27/2022] Open
Abstract
The Aggregatibacter actinomycetemcomitans cytolethal distending toxin (Cdt) induces lymphocytes to undergo cell-cycle arrest and apoptosis; toxicity is dependent upon the active Cdt subunit, CdtB. We now demonstrate that p21CIP1/WAF1 is critical to Cdt-induced apoptosis. Cdt induces increases in the levels of p21CIP1/WAF1 in lymphoid cell lines, Jurkat and MyLa, and in primary human lymphocytes. These increases were dependent upon CdtB’s ability to function as a phosphatidylinositol (PI) 3,4,5-triphosphate (PIP3) phosphatase. It is noteworthy that Cdt-induced increases in the levels of p21CIP1/WAF1 were accompanied by a significant decline in the levels of phosphorylated p21CIP1/WAF1. The significance of Cdt-induced p21CIP1/WAF1 increase was assessed by preventing these changes with a two-pronged approach; pre-incubation with the novel p21CIP1/WAF1 inhibitor, UC2288, and development of a p21CIP1/WAF1-deficient cell line (Jurkatp21−) using clustered regularly interspaced short palindromic repeats (CRISPR)/cas9 gene editing. UC2288 blocked toxin-induced increases in p21CIP1/WAF1, and JurkatWT cells treated with this inhibitor exhibited reduced susceptibility to Cdt-induced apoptosis. Likewise, Jurkatp21− cells failed to undergo toxin-induced apoptosis. The linkage between Cdt, p21CIP1/WAF1, and apoptosis was further established by demonstrating that Cdt-induced increases in levels of the pro-apoptotic proteins Bid, Bax, and Bak were dependent upon p21CIP1/WAF1 as these changes were not observed in Jurkatp21− cells. Finally, we determined that the p21CIP1/WAF1 increases were dependent upon toxin-induced increases in the level and activity of the chaperone heat shock protein (HSP) 90. We propose that p21CIP1/WAF1 plays a key pro-apoptotic role in mediating Cdt-induced toxicity.
Collapse
|
89
|
Sun Y, Zhang G, Hou X, Xiao S, Yang X, Xie Y, Huang X, Wang F, Mo X, Ding X, Xia L, Hu S. SrfABC Toxin from Xenorhabdus stockiae Induces Cytotoxicity and Apoptosis in HeLa Cells. Toxins (Basel) 2019; 11:toxins11120685. [PMID: 31766712 PMCID: PMC6950479 DOI: 10.3390/toxins11120685] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 11/16/2022] Open
Abstract
Our previous study showed that the srfABC operon, which was originally identified in Salmonella enterica as an SsrB-regulated operon clustered with the flagellar class 2 operon, exhibited significant cytotoxicity against insect midgut CF-203 cells and injectable insecticidal activity against Helicoverpa armigera larvae. The srfABC operon was widely distributed among bacteria, which raises the question of their biological roles in different species. In this study, we investigated the cytotoxic effect of SrfABC toxin on mammalian cell lines. When simultaneously expressed in the Escherichia coli cytoplasm, SrfABC exhibited cytotoxicity against all tested mammalian cancer cell lines (B16, 4T-1, Hep-3B, and HeLa) in a dose-dependent manner. Intracellular expression of SrfA-FLAG, SrfB-FLAG, or SrfC-FLAG also resulted in inhibition of proliferation and apoptosis on HeLa cells. When incubated with HeLa cells separately, SrfA, SrfB, and SrfC proteins alone could enter HeLa cells, then induce apoptosis and cytotoxicity. SrfC protein shifts its localization from cytoplasm to nucleus with the aid of SrfA and/or SrfB protein. Although SrfA, SrfB, and SrfC proteins alone exhibited a cytotoxic effect against HeLa cells, all three components were essential for the full cytotoxicity. Native PAGE and co-immunoprecipitation assay demonstrated that SrfA, SrfB, and SrfC proteins could interact with each other and form a heteromeric complex.
Collapse
|
90
|
Belibasakis GN, Maula T, Bao K, Lindholm M, Bostanci N, Oscarsson J, Ihalin R, Johansson A. Virulence and Pathogenicity Properties of Aggregatibacter actinomycetemcomitans. Pathogens 2019; 8:E222. [PMID: 31698835 PMCID: PMC6963787 DOI: 10.3390/pathogens8040222] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
Aggregatibacter actinomycetemcomitans is a periodontal pathogen colonizing the oral cavity of a large proportion of the human population. It is equipped with several potent virulence factors that can cause cell death and induce or evade inflammation. Because of the large genetic diversity within the species, both harmless and highly virulent genotypes of the bacterium have emerged. The oral condition and age, as well as the geographic origin of the individual, influence the risk to be colonized by a virulent genotype of the bacterium. In the present review, the virulence and pathogenicity properties of A. actinomycetemcomitans will be addressed.
Collapse
Affiliation(s)
- Georgios N. Belibasakis
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, S-141 04 Huddinge, Sweden; (G.N.B.); (K.B.); (N.B.)
| | - Terhi Maula
- Department of Biochemistry, University of Turku, FI-20014 Turku, Finland; (T.M.); (R.I.)
| | - Kai Bao
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, S-141 04 Huddinge, Sweden; (G.N.B.); (K.B.); (N.B.)
| | - Mark Lindholm
- Department of Odontology, Umeå University, S-901 87 Umeå, Sweden; (M.L.); (J.O.)
| | - Nagihan Bostanci
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, S-141 04 Huddinge, Sweden; (G.N.B.); (K.B.); (N.B.)
| | - Jan Oscarsson
- Department of Odontology, Umeå University, S-901 87 Umeå, Sweden; (M.L.); (J.O.)
| | - Riikka Ihalin
- Department of Biochemistry, University of Turku, FI-20014 Turku, Finland; (T.M.); (R.I.)
| | - Anders Johansson
- Department of Odontology, Umeå University, S-901 87 Umeå, Sweden; (M.L.); (J.O.)
| |
Collapse
|
91
|
Cytolethal Distending Toxin Subunit B: A Review of Structure-Function Relationship. Toxins (Basel) 2019; 11:toxins11100595. [PMID: 31614800 PMCID: PMC6832162 DOI: 10.3390/toxins11100595] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 01/27/2023] Open
Abstract
The Cytolethal Distending Toxin (CDT) is a bacterial virulence factor produced by several Gram-negative pathogenic bacteria. These bacteria, found in distinct niches, cause diverse infectious diseases and produce CDTs differing in sequence and structure. CDTs have been involved in the pathogenicity of the associated bacteria by promoting persistent infection. At the host-cell level, CDTs cause cell distension, cell cycle block and DNA damage, eventually leading to cell death. All these effects are attributable to the catalytic CdtB subunit, but its exact mode of action is only beginning to be unraveled. Sequence and 3D structure analyses revealed similarities with better characterized proteins, such as nucleases or phosphatases, and it has been hypothesized that CdtB exerts a biochemical activity close to those enzymes. Here, we review the relationships that have been established between CdtB structure and function, particularly by mutation experiments on predicted key residues in different experimental systems. We discuss the relevance of these approaches and underline the importance of further study in the molecular mechanisms of CDT toxicity, particularly in the context of different pathological conditions.
Collapse
|
92
|
Kim J, Park H, Kim J, Kim JH, Jung JI, Cho S, Ryu S, Jeon B. Comparative Analysis of Aerotolerance, Antibiotic Resistance, and Virulence Gene Prevalence in Campylobacter jejuni Isolates from Retail Raw Chicken and Duck Meat in South Korea. Microorganisms 2019; 7:microorganisms7100433. [PMID: 31658662 PMCID: PMC6843641 DOI: 10.3390/microorganisms7100433] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/02/2019] [Accepted: 10/08/2019] [Indexed: 12/22/2022] Open
Abstract
Human infections with Campylobacter are primarily associated with the consumption of contaminated poultry meat. In this study, we isolated Campylobacterjejuni from retail raw chicken and duck meat in Korea and compared their aerotolerance, antibiotic resistance, and virulence gene prevalence. Whereas C. jejuni isolates from chicken dominantly belonged to multilocus sequence typing (MLST) clonal complex (CC)-21, CC-45 is the common MLST sequence type in duck meat isolates. C. jejuni strains from both chicken and duck meat were highly tolerant to aerobic stress. The prevalence of virulence genes was higher in C. jejuni strains from chicken than those from duck meat. However, antibiotic resistance was higher in duck meat isolates than chicken isolates. Based on the prevalence of virulence genes and antibiotic resistance, fluoroquinolone-resistant C. jejuni strains harboring all tested virulence genes except virB11 were predominant on retail poultry. Fluoroquinolone-resistant C. jejuni strains carrying most virulence genes were more frequently isolated in summer than in winter. The comparative profiling analysis in this study successfully demonstrated that antibiotic-resistant and pathogenic strains of C. jejuni are highly prevalent on retail poultry and that retail duck meat is an important vehicle potentially transmitting C. jejuni to humans in Korea.
Collapse
Affiliation(s)
- Jinshil Kim
- Department of Food and Animal Biotechnology, Research Institute for Agriculture and Life Sciences, and Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Korea.
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
| | - Hyeeun Park
- Department of Food and Animal Biotechnology, Research Institute for Agriculture and Life Sciences, and Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Korea.
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
| | - Junhyung Kim
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea.
| | - Jong Hyun Kim
- The Korea Centers for Disease Control and Prevention, Osong 28159, Korea.
| | - Jae In Jung
- The Korea Centers for Disease Control and Prevention, Osong 28159, Korea.
| | - Seongbeom Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea.
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, Research Institute for Agriculture and Life Sciences, and Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Korea.
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
| | - Byeonghwa Jeon
- Department of Food and Animal Biotechnology, Research Institute for Agriculture and Life Sciences, and Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Korea.
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
- Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
93
|
Sha S, Ni L, Stefil M, Dixon M, Mouraviev V. The human gastrointestinal microbiota and prostate cancer development and treatment. Investig Clin Urol 2019; 61:S43-S50. [PMID: 32055753 PMCID: PMC7004837 DOI: 10.4111/icu.2020.61.s1.s43] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/16/2019] [Indexed: 01/14/2023] Open
Abstract
The human gastrointestinal microbiome contains commensal bacteria and other microbiota that have been gaining increasing attention in the context of cancer development and response to treatment. Microbiota play a role in the maintenance of host barrier surfaces that contribute to both local inflammation and other systemic metabolic functions. In the context of prostate cancer, the gastrointestinal microbiome may play a role through metabolism of estrogen, an increase of which has been linked to the induction of prostatic neoplasia. Specific microbiota such as Bacteroides, Streptococcus, Bacteroides massiliensis, Faecalibacterium prausnitzii, Eubacterium rectalie, and Mycoplasma genitalium have been associated with differing risks of prostate cancer development or extensiveness of prostate cancer disease. In this Review, we discuss gastrointestinal microbiota's effects on prostate cancer development, the ability of the microbiome to regulate chemotherapy for prostate cancer treatment, and the importance of using Next Generation Sequencing to further discern the microbiome's systemic influence on prostate cancer.
Collapse
Affiliation(s)
- Sybil Sha
- Dartmouth Medical School, Hanover, NH, USA
| | - Liqiang Ni
- University of Central Florida, Orlando, FL, USA
| | | | | | | |
Collapse
|
94
|
Sharma P, Garg N, Sharma A, Capalash N, Singh R. Nucleases of bacterial pathogens as virulence factors, therapeutic targets and diagnostic markers. Int J Med Microbiol 2019; 309:151354. [PMID: 31495663 DOI: 10.1016/j.ijmm.2019.151354] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/12/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022] Open
Abstract
New frontiers of therapy are being explored against the upcoming bacterial diseases rendered untreatable due to multiple, extreme and pan- antibiotic resistance. Nucleases are ubiquitous in bacterial pathogens performing various functions like acquiring nucleotide nutrients, allowing or preventing uptake of foreign DNA, controlling biofilm formation/dispersal/architecture, invading host by tissue damage, evading immune defence by degrading DNA matrix of neutrophil extracellular traps (NETs) and immunomodulating the host immune response. Secretory nucleases also provide means of survival to other bacteria like iron-reducing Shewanella and such functions help them adapt and survive proficiently. Other than their pro-pathogen roles in survival, nucleases can be used directly as therapeutics. One of the powerful armours of pathogens is the formation of biofilms, thus helping them resist and persist in the harshest of environments. As eDNA forms the structural and binding component of biofilm, nucleases can be used against the adhering component, thus increasing the permeability of antimicrobial agents. Nucleases have recently become a model system of intense study for their biological functions and medical applications in diagnosis, immunoprophylaxis and therapy. Rational implications of these enzymes can impact human medicine positively in future by opening new ways for therapeutics which have otherwise reached saturation due to multi drug resistance.
Collapse
Affiliation(s)
- Prince Sharma
- Department of Microbiology, Panjab University, Chandigarh, India.
| | - Nisha Garg
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Anshul Sharma
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Neena Capalash
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - Ravinder Singh
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
95
|
Burnham PM, Hendrixson DR. Campylobacter jejuni: collective components promoting a successful enteric lifestyle. Nat Rev Microbiol 2019; 16:551-565. [PMID: 29892020 DOI: 10.1038/s41579-018-0037-9] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Campylobacter jejuni is the leading cause of bacterial diarrhoeal disease in many areas of the world. The high incidence of sporadic cases of disease in humans is largely due to its prevalence as a zoonotic agent in animals, both in agriculture and in the wild. Compared with many other enteric bacterial pathogens, C. jejuni has strict growth and nutritional requirements and lacks many virulence and colonization determinants that are typically used by bacterial pathogens to infect hosts. Instead, C. jejuni has a different collection of factors and pathways not typically associated together in enteric pathogens to establish commensalism in many animal hosts and to promote diarrhoeal disease in the human population. In this Review, we discuss the cellular architecture and structure of C. jejuni, intraspecies genotypic variation, the multiple roles of the flagellum, specific nutritional and environmental growth requirements and how these factors contribute to in vivo growth in human and avian hosts, persistent colonization and pathogenesis of diarrhoeal disease.
Collapse
Affiliation(s)
- Peter M Burnham
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David R Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
96
|
Alternate subunit assembly diversifies the function of a bacterial toxin. Nat Commun 2019; 10:3684. [PMID: 31417089 PMCID: PMC6695444 DOI: 10.1038/s41467-019-11592-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 07/04/2019] [Indexed: 11/27/2022] Open
Abstract
Bacterial toxins with an AB5 architecture consist of an active (A) subunit inserted into a ring-like platform comprised of five delivery (B) subunits. Salmonella Typhi, the cause of typhoid fever, produces an unusual A2B5 toxin known as typhoid toxin. Here, we report that upon infection of human cells, S. Typhi produces two forms of typhoid toxin that have distinct delivery components but share common active subunits. The two typhoid toxins exhibit different trafficking properties, elicit different effects when administered to laboratory animals, and are expressed using different regulatory mechanisms and in response to distinct metabolic cues. Collectively, these results indicate that the evolution of two typhoid toxin variants has conferred functional versatility to this virulence factor. More broadly, this study reveals a new paradigm in toxin biology and suggests that the evolutionary expansion of AB5 toxins was likely fueled by the plasticity inherent to their structural design coupled to the functional versatility afforded by the combination of homologous toxin components. Salmonella Typhi produces the typhoid toxin. Here, Fowler et al. show that S. Typhi produces two forms of typhoid toxin that are differentially regulated and display different trafficking properties and different effects when administered to laboratory animals.
Collapse
|
97
|
Denmongkholchai S, Katare P, Choochuay S, Thanyasrisung P, Tsuruda K, Sugai M, Mongkolsuk S, Matangkasombut O. Genome-Wide Identification of Host Genes Required for Toxicity of Bacterial Cytolethal Distending Toxin in a Yeast Model. Front Microbiol 2019; 10:890. [PMID: 31080443 PMCID: PMC6497811 DOI: 10.3389/fmicb.2019.00890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/08/2019] [Indexed: 12/16/2022] Open
Abstract
Background Aggregatibacter actinomycetemcomitans, a periodontal pathogen, secretes a cytolethal distending toxin (AaCDT) that causes host cell cycle arrest and cell death. Although CDT could be an important virulence factor, it is unclear how it enters the nucleus to exert its cytotoxicity. Objective To investigate the mechanisms of AaCDT by genome-wide screening for host mutations that confer resistance to the catalytic subunit, AaCdtB, in a Saccharomyces cerevisiae model. Methods We transformed the yeast haploid deletion library, a collection of yeast mutants with single gene deletions of virtually all non-essential ORFs in the genome, with plasmids carrying galactose-inducible AaCdtB. Yeast mutants that showed resistance to AaCdtB were selected and rescreened by a spotting assay. AaCdtB expression was confirmed by western blot analysis; any mutants that showed no or weak expression of AaCdtB were omitted from the analysis. The lists of genes whose mutations confer resistance to AaCdtB were analyzed for Gene Ontology (GO) term enrichments. Localization of AaCdtB-EGFP was examined using fluorescent microscopy. Nuclear localization relative to EGFP control was calculated and compared to wild-type. Results Out of approximately 5,000 deletion mutants, we isolated 243 mutants that are resistant to AaCdtB. GO analyses indicated that genes associated with organic anion transport are significantly enriched (16 genes). Furthermore, several genes associated with the nucleus and endoplasmic reticulum (ER) were identified. Localization studies of AaCdtB, in mutants with the deletion of genes associated with the GO term organic anion transport, showed lower nuclear localization than wild-type. The results suggest that these genes may be required for AaCdtB translocation into the nucleus and its cytotoxicity. Conclusion The genome-wide screen in the yeast deletion library allowed us to identify a large number of host genes required for AaCdtB cytotoxicity. Further investigation could lead to more insights into the mechanisms of CdtB intoxication.
Collapse
Affiliation(s)
- Siriyod Denmongkholchai
- Interdepartmental Program in Medical Microbiology, Graduate School, Chulalongkorn University, Bangkok, Thailand.,Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prashant Katare
- Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Graduate Program in Oral Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Sarocha Choochuay
- Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Panida Thanyasrisung
- Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Keiko Tsuruda
- Department of Oral Epidemiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Motoyuki Sugai
- Department of Antimicrobial Resistance, Graduate School of Biomedical and Health Sciences, Project Research Center for Nosocomial Infectious Diseases (RCNID), Hiroshima University, Hiroshima, Japan.,Antimicrobial Resistance Research Center, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | - Skorn Mongkolsuk
- Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Oranart Matangkasombut
- Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, Thailand
| |
Collapse
|
98
|
Chang SJ, Jin SC, Jiao X, Galán JE. Unique features in the intracellular transport of typhoid toxin revealed by a genome-wide screen. PLoS Pathog 2019; 15:e1007704. [PMID: 30951565 PMCID: PMC6469816 DOI: 10.1371/journal.ppat.1007704] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/17/2019] [Accepted: 03/13/2019] [Indexed: 12/29/2022] Open
Abstract
Typhoid toxin is a virulence factor for Salmonella Typhi and Paratyphi, the cause of typhoid fever in humans. This toxin has a unique architecture in that its pentameric B subunit, made of PltB, is linked to two enzymatic A subunits, the ADP ribosyl transferase PltA and the deoxyribonuclease CdtB. Typhoid toxin is uniquely adapted to humans, recognizing surface glycoprotein sialoglycans terminated in acetyl neuraminic acid, which are preferentially expressed by human cells. The transport pathway to its cellular targets followed by typhoid toxin after receptor binding is currently unknown. Through a genome-wide CRISPR/Cas9-mediated screen we have characterized the mechanisms by which typhoid toxin is transported within human cells. We found that typhoid toxin hijacks specific elements of the retrograde transport and endoplasmic reticulum-associated degradation machineries to reach its subcellular destination within target cells. Our study reveals unique and common features in the transport mechanisms of bacterial toxins that could serve as the bases for the development of novel anti-toxin therapeutic strategies.
Collapse
Affiliation(s)
- Shu-Jung Chang
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sheng Chih Jin
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Xuyao Jiao
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jorge E Galán
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
99
|
Massari F, Mollica V, Di Nunno V, Gatto L, Santoni M, Scarpelli M, Cimadamore A, Lopez-Beltran A, Cheng L, Battelli N, Montironi R, Brandi G. The Human Microbiota and Prostate Cancer: Friend or Foe? Cancers (Basel) 2019; 11:E459. [PMID: 30935126 PMCID: PMC6521295 DOI: 10.3390/cancers11040459] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 02/06/2023] Open
Abstract
The human microbiome is gaining increasing attention in the medical community, as knowledge on its role not only in health but also in disease development and response to therapies is expanding. Furthermore, the connection between the microbiota and cancer, especially the link between the gut microbiota and gastrointestinal tumors, is becoming clearer. The interaction between the microbiota and the response to chemotherapies and, more recently, to immunotherapy has been widely studied, and a connection between a peculiar type of microbiota and a better response to these therapies and a different incidence in toxicities has been hypothesized. As knowledge on the gut microbiota increases, interest in the residing microbial population in other systems of our body is also increasing. Consequently, the urinary microbiota is under evaluation for its possible implications in genitourinary diseases, including cancer. Prostate cancer is the most common cancer in the male population; thus, research regarding its etiology and possible factors correlated to disease progression or the response to specific therapies is thriving. This review has the purpose to recollect the current knowledge on the relationship between the human microbiota and prostate cancer.
Collapse
Affiliation(s)
- Francesco Massari
- Division of Oncology, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Veronica Mollica
- Division of Oncology, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Vincenzo Di Nunno
- Division of Oncology, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | - Lidia Gatto
- Division of Oncology, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy.
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, 60126 Ancona, Italy.
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, 60126 Ancona, Italy.
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, 60126 Ancona, Italy.
| | - Giovanni Brandi
- Oncology Unit, Department of Experimental, Diagnostic and Specialty Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy.
| |
Collapse
|
100
|
Pons BJ, Bezine E, Hanique M, Guillet V, Mourey L, Chicher J, Frisan T, Vignard J, Mirey G. Cell transfection of purified cytolethal distending toxin B subunits allows comparing their nuclease activity while plasmid degradation assay does not. PLoS One 2019; 14:e0214313. [PMID: 30921382 PMCID: PMC6438463 DOI: 10.1371/journal.pone.0214313] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/11/2019] [Indexed: 12/30/2022] Open
Abstract
The Cytolethal Distending Toxin (CDT) is produced by many pathogenic bacteria. CDT is known to induce genomic DNA damage to host eukaryotic cells through its catalytic subunit, CdtB. CdtB is structurally homologous to DNase I and has a nuclease activity, dependent on several key residues. Yet some differences between various CdtB subunit activities, and discrepancies between biochemical and cellular data, have been observed. To better characterise the role of CdtB in the induction of DNA damage, we affinity-purified wild-type and mutants of CdtB, issued from E. coli and H. ducreyi, under native and denaturing conditions. We then compared their nuclease activity by a classic in vitro assay using plasmid DNA, and two different eukaryotic assays–the first assay where host cells were transfected with a plasmid encoding CdtB, the second assay where host cells were directly transfected with purified CdtB. We show here that in vitro nuclease activities are difficult to quantify, whereas CdtB activities in host cells can be easily interpreted and confirmed the loss of function of the catalytic mutant. Our results highlight the importance of performing multiple assays while studying the effects of bacterial genotoxins, and indicate that the classic in vitro assay should be complemented with cellular assays.
Collapse
Affiliation(s)
- Benoît J. Pons
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
- Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Elisabeth Bezine
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
- Institut National Polytechnique de Toulouse, Toulouse, France
| | - Mélissa Hanique
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
| | - Valérie Guillet
- Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
- Centre National de la Recherche Scientifique (CNRS), Institut de Pharmacologie et de Biologie Structurale (IPBS), Toulouse, France
| | - Lionel Mourey
- Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
- Centre National de la Recherche Scientifique (CNRS), Institut de Pharmacologie et de Biologie Structurale (IPBS), Toulouse, France
| | - Johana Chicher
- Plateforme protéomique Strasbourg Esplanade, Institut de Biologie Moléculaire et Cellulaire (IBMC), FRC1589 Strasbourg, France
| | - Teresa Frisan
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Julien Vignard
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
- * E-mail: (GM); (JV)
| | - Gladys Mirey
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
- Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
- * E-mail: (GM); (JV)
| |
Collapse
|