1
|
Ying YT, Yang J, Ye HW, Chen MY, Liu X, Chen W, Xu JX, Tan X. Staphylococcus aureus reprograms CASP8 (caspase 8) signaling to evade cell death and Xenophagy. Autophagy 2025:1-14. [PMID: 40143428 DOI: 10.1080/15548627.2025.2483887] [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: 11/06/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/28/2025] Open
Abstract
Regulated cell death and xenophagy constitute fundamental cellular mechanisms against invading microorganisms. Staphylococcus aureus, a notorious pathogen, can invade and persist within host cells for extended periods. Here, we describe a novel mechanism by which S. aureus subverts these host defenses through the manipulation of the CASP8 (caspase 8) signaling pathway. Upon invasion, S. aureus triggers the assembly of a RIPK3 (receptor interacting serine/threonine kinase 3) complex to induce CASP8 autoprocessing. However, the bacterium inhibits CUL3 (cullin 3)-dependent K63-linked ubiquitination, leading to an atypical activation of CASP8. This non-canonical activation does not initiate the CASP8-CASP3 cascade but instead suppresses RIPK3-dependent necroptosis, a regulated cell death pathway typically activated when apoptosis fails. The resulting non-apoptotic, cleaved CASP8 redirects its enzymatic activity toward cleaving SQSTM1/p62, a selective macroautophagy/autophagy receptor, thus enabling S. aureus to evade antimicrobial xenophagy. The results of this study suggest that S. aureus reprograms the CASP8 signaling pathway from inducing cell death to preserving cell survival and inhibiting xenophagy, a critical strategy that supports its stealthy replication and persistence within host cells.Abbreviations: CASP3: caspase 3; CASP8: caspase 8; CFU: colony-forming units; CUL3: cullin 3; DUB: deubiquitinating enzyme; MAP1LC3B-II/LC3B-II: microtubule associated protein 1 light chain 3 beta-II; MOI: multiplicity of infection; RIPK1: receptor interacting protein kinase 1; RIPK3: receptor interacting protein kinase 3; S. aureus: Staphylococcus aureus.
Collapse
Affiliation(s)
- Yi-Tian Ying
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Yang
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Hui-Wen Ye
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Mei-Yi Chen
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Wei Chen
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Jin-Xin Xu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| | - Xun Tan
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Veterinary Medical Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
2
|
Zhang H, Jin Q, Li J, Wang J, Li M, Yin Q, Li Q, Qi Y, Feng L, Shen L, Qin Y, Cong Q. Astrocyte-derived complement C3 facilitated microglial phagocytosis of synapses in Staphylococcus aureus-associated neurocognitive deficits. PLoS Pathog 2025; 21:e1013126. [PMID: 40294039 PMCID: PMC12121917 DOI: 10.1371/journal.ppat.1013126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 05/29/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
The presence of pathogens is a significant challenge in causing brain infections and tissue damage. There is growing evidence that pathogen infections are commonly associated with cognitive dysfunction and mental health problems, but the underlying mechanisms are not yet fully understood. Here, we found microglia and astrocyte activation, neuronal damage, synapse loss, and cognitive impairment in a Staphylococcus aureus (S. aureus) induced mouse model. An unbiased transcription profile of isolated microglia derived from S. aureus-infected mice identified the involvement of microglial phagosome and regulation of neurogenesis. Our findings indicate that the complement C1q and C3 are upregulated, and astroglial release of C3 activates microglia to phagocytose synapses. Blocking the C3-C3aR axis can improve microglial phagocytosis, thus rescuing synapse loss and cognitive impairment in infected mice. These results indicate that S. aureus induces synapse elimination and cognitive impairment by activating microglia and astrocytes through C3-C3aR signaling. This suggests a mechanism of complement signaling bridged crosstalk between astrocyte and microglia in the S. aureus-associated post-infectious synapse loss and cognitive dysfunction, and provide potential therapeutic targets for managing pathogen-associated brain infections.
Collapse
Affiliation(s)
- Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jijie Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiali Wang
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
| | - Mengqi Li
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
| | - Qiao Yin
- Department of Neurology, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Li
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
| | - Yuwan Qi
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
| | - Lingling Feng
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
| | - Liang Shen
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
| | - Yuan Qin
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
| | - Qifei Cong
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Neurology, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
3
|
Abo-Zeid FS, Abdurabbah EM, Fares NH, Al-Karmalawy AA. Ultrastructural effects of Staphylococcus aureus toxicity on albino mice kidney. Microb Pathog 2025; 198:107110. [PMID: 39527986 DOI: 10.1016/j.micpath.2024.107110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/25/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Staphylococcus aureus (S. aureus) is a prominent infectious etiological agent in humans, dairy animals, and camels. Camel milk has all the nutrients which are nutritious and advantageous to the growth of S. aureus that dominates most bacterial species. So, the present work was designed to investigate the effect of certain toxin gene of S. aureus bacteria isolated from raw camel milk on the fine structure of mice kidneys. Two toxin genes of S. aureus were identified and described through PCR using 23s rRNA primer for 23s rRNA gene. In addition, the effect of the isolated S. aureus bacteria, especially those carrying the gene "tst" was studied on the ultrastructure of mice kidney. Twenty male albino mice were allocated into 2 groups, control group and infected group (orally administered with a single dosage of S. aureus aqueous solutions from camel milk at a concentration of 5 × 108 colony forming unit/0.1 mL for three days). Infection of mice with S. aureus resulted in vacuolation, necrosis, and degeneration of tubular epithelial cells, intertubular congestion and inflammation, as well as dilatation and congestion of glomeruli. Therefore, this current study most likely indicates that camel milk infected with S. aureus has a significant and clear impact in inducing symptoms of kidney failure in communities that use these dairy products without health supervision, especially nomadic and pastoral communities.
Collapse
Affiliation(s)
- Faten S Abo-Zeid
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Ebtisam M Abdurabbah
- Department of Zoology, Faculty of Science, Omar Al-Mukhtar University, Al-Bayda, Libya
| | - Nagui H Fares
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad, 10023, Iraq; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt.
| |
Collapse
|
4
|
Liu F, Pan R, Ding H, Gu L, Yang Y, Li C, Xu Y, Hu R, Chen H, Zhang X, Nie Y. UBQLN4 is an ATM substrate that stabilizes the anti-apoptotic proteins BCL2A1 and BCL2L10 in mesothelioma. Mol Oncol 2021; 15:3738-3752. [PMID: 34245648 PMCID: PMC8637560 DOI: 10.1002/1878-0261.13058] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/18/2021] [Accepted: 07/09/2021] [Indexed: 11/15/2022] Open
Abstract
ATM serine/threonine kinase (ATM; previously known as ataxia-telangiectasia mutated) plays a critical role in maintaining genomic stability and regulates multiple downstream pathways, such as DNA repair, cell cycle arrest, and apoptosis. As a serine/threonine kinase, ATM has an array of downstream phosphorylation substrates, including checkpoint effector checkpoint kinase 2 (CHK2). ATM inhibits cell cycle progression by phosphorylating and activating CHK2, which plays an important role in the formation and development of tumors and participates in DNA repair responses after double-stranded DNA breaks. In this study, we used a recently developed mammalian functional genetic screening system to explore a series of ATM substrates and their role in DNA damage to enhance our understanding of the DNA damage response. Ubiquilin 4 (UBQLN4), which belongs to the ubiquilin family characterized by its ubiquitin-like (UBL) and ubiquitin-associated (UBA) domains, was identified as a new substrate for ATM. UBQLN4 is involved in various intracellular processes, such as autophagosome maturation, p21 regulation, and motor axon morphogenesis. However, the biological function of UBQLN4 remains to be elucidated. In this study, we not only identified UBQLN4 as a substrate for ATM, but also found that UBQLN4 interacts with and stabilizes the anti-apoptotic proteins Bcl-2-related protein A1 (BCL2A1) and Bcl-2-like protein 10 (BCL2L10) and prevents mesothelioma cell apoptosis in response to DNA damage. These findings expand our understanding of the role of UBQLN4 in mesothelioma and provide new insights into potential mesothelioma treatments targeting substrates for ATM.
Collapse
Affiliation(s)
- Fang Liu
- Medical CollegeGuizhou UniversityGuiyangChina
| | - RunSang Pan
- GuiYang Maternal and Child HospitalGuiyangChina
| | - HongYu Ding
- State Key Laboratory of Systems BiologyCAS Center for Excellence in Molecular Cell ScienceInnovation Center for Cell Signaling NetworkShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghaiChina
| | - LiLing Gu
- Medical CollegeGuizhou UniversityGuiyangChina
- Department of RehabilitationGuizhou Provincial People’s HospitalGuiyangChina
- NHC Key Laboratory of Pulmonary Immune‐related DiseasesGuizhou Provincial People’s HospitalGuiyangChina
| | - Yun Yang
- Medical CollegeGuizhou UniversityGuiyangChina
| | - ChuanYin Li
- State Key Laboratory of Systems BiologyCAS Center for Excellence in Molecular Cell ScienceInnovation Center for Cell Signaling NetworkShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghaiChina
| | - YongJie Xu
- NHC Key Laboratory of Pulmonary Immune‐related DiseasesGuizhou Provincial People’s HospitalGuiyangChina
| | - Ronggui Hu
- State Key Laboratory of Systems BiologyCAS Center for Excellence in Molecular Cell ScienceInnovation Center for Cell Signaling NetworkShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghaiChina
| | - Hui Chen
- NHC Key Laboratory of Pulmonary Immune‐related DiseasesGuizhou Provincial People’s HospitalGuiyangChina
| | - XiangYan Zhang
- NHC Key Laboratory of Pulmonary Immune‐related DiseasesGuizhou Provincial People’s HospitalGuiyangChina
| | - YingJie Nie
- NHC Key Laboratory of Pulmonary Immune‐related DiseasesGuizhou Provincial People’s HospitalGuiyangChina
| |
Collapse
|
5
|
Marchi S, Morroni G, Pinton P, Galluzzi L. Control of host mitochondria by bacterial pathogens. Trends Microbiol 2021; 30:452-465. [PMID: 34656395 DOI: 10.1016/j.tim.2021.09.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022]
Abstract
Mitochondria control various processes that are integral to cellular and organismal homeostasis, including Ca2+ fluxes, bioenergetic metabolism, and cell death. Perhaps not surprisingly, multiple pathogenic bacteria have evolved strategies to subvert mitochondrial functions in support of their survival and dissemination. Here, we discuss nonimmunological pathogenic mechanisms that converge on the ability of bacteria to control the mitochondrial compartment of host cells.
Collapse
Affiliation(s)
- Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy.
| | - Gianluca Morroni
- Department of Biomedical Sciences & Public Health, Marche Polytechnic University, Ancona, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Université de Paris, Paris, France.
| |
Collapse
|
6
|
Staphylococcus aureus isolates from hospital clinics induce ROS-mediated DNA damage, apoptosis and gene expression alterations in male mice. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
7
|
Selvaraj C, Vierra M, Dinesh DC, Abhirami R, Singh SK. Structural insights of macromolecules involved in bacteria-induced apoptosis in the pathogenesis of human diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 126:1-38. [PMID: 34090612 DOI: 10.1016/bs.apcsb.2021.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Numbers of pathogenic bacteria can induce apoptosis in human host cells and modulate the cellular pathways responsible for inducing or inhibiting apoptosis. These pathogens are significantly recognized by host proteins and provoke the multitude of several signaling pathways and alter the cellular apoptotic stimuli. This process leads the bacterial entry into the mammalian cells and evokes a variety of responses like phagocytosis, release of mitochondrial cytochrome c, secretion of bacterial effectors, release of both apoptotic and inflammatory cytokines, and the triggering of apoptosis. Several mechanisms are involved in bacteria-induced apoptosis including, initiation of the endogenous death machinery, pore-forming proteins, and secretion of superantigens. Either small molecules or proteins may act as a binding partner responsible for forming the protein complexes and regulate enzymatic activity via protein-protein interactions. The bacteria induce apoptosis, attack the human cell and gain control over various types of cells and tissue. Since these processes are intricate in the defense mechanisms of host organisms against pathogenic bacteria and play an important function in host-pathogen interactions. In this chapter, we focus on the various bacterial-induced apoptosis mechanisms in host cells and discuss the important proteins and bacterial effectors that trigger the host cell apoptosis. The structural characterization of bacterial effector proteins and their interaction with human host cells are also considered.
Collapse
Affiliation(s)
- Chandrabose Selvaraj
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India.
| | - Marisol Vierra
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
| | | | - Rajaram Abhirami
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
8
|
Missiakas D, Winstel V. Selective Host Cell Death by Staphylococcus aureus: A Strategy for Bacterial Persistence. Front Immunol 2021; 11:621733. [PMID: 33552085 PMCID: PMC7859115 DOI: 10.3389/fimmu.2020.621733] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
Host cell death programs are fundamental processes that shape cellular homeostasis, embryonic development, and tissue regeneration. Death signaling and downstream host cell responses are not only critical to guide mammalian development, they often act as terminal responses to invading pathogens. Here, we briefly review and contrast how invading pathogens and specifically Staphylococcus aureus manipulate apoptotic, necroptotic, and pyroptotic cell death modes to establish infection. Rather than invading host cells, S. aureus subverts these cells to produce diffusible molecules that cause death of neighboring hematopoietic cells and thus shapes an immune environment conducive to persistence. The exploitation of cell death pathways by S. aureus is yet another virulence strategy that must be juxtaposed to mechanisms of immune evasion, autophagy escape, and tolerance to intracellular killing, and brings us closer to the true portrait of this pathogen for the design of effective therapeutics and intervention strategies.
Collapse
Affiliation(s)
- Dominique Missiakas
- Howard Taylor Ricketts Laboratory, Department of Microbiology, University of Chicago, Lemont, IL, United States
| | - Volker Winstel
- Research Group Pathogenesis of Bacterial Infections, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
9
|
Abstract
Despite being regarded as an extracellular bacterium, the pathogen Staphylococcus aureus can invade and survive within human cells. The intracellular niche is considered a hideout from the host immune system and antibiotic treatment and allows bacterial proliferation. The opportunistic human pathogen Staphylococcus aureus causes serious infectious diseases that range from superficial skin and soft tissue infections to necrotizing pneumonia and sepsis. While classically regarded as an extracellular pathogen, S. aureus is able to invade and survive within human cells. Host cell exit is associated with cell death, tissue destruction, and the spread of infection. The exact molecular mechanism employed by S. aureus to escape the host cell is still unclear. In this study, we performed a genome-wide small hairpin RNA (shRNA) screen and identified the calcium signaling pathway as being involved in intracellular infection. S. aureus induced a massive cytosolic Ca2+ increase in epithelial host cells after invasion and intracellular replication of the pathogen. This was paralleled by a decrease in endoplasmic reticulum Ca2+ concentration. Additionally, calcium ions from the extracellular space contributed to the cytosolic Ca2+ increase. As a consequence, we observed that the cytoplasmic Ca2+ rise led to an increase in mitochondrial Ca2+ concentration, the activation of calpains and caspases, and eventually to cell lysis of S. aureus-infected cells. Our study therefore suggests that intracellular S. aureus disturbs the host cell Ca2+ homeostasis and induces cytoplasmic Ca2+ overload, which results in both apoptotic and necrotic cell death in parallel or succession.
Collapse
|
10
|
Mulcahy ME, O'Brien EC, O'Keeffe KM, Vozza EG, Leddy N, McLoughlin RM. Manipulation of Autophagy and Apoptosis Facilitates Intracellular Survival of Staphylococcus aureus in Human Neutrophils. Front Immunol 2020; 11:565545. [PMID: 33262756 PMCID: PMC7686353 DOI: 10.3389/fimmu.2020.565545] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/15/2020] [Indexed: 01/13/2023] Open
Abstract
Polymorphonuclear neutrophils (PMN) are critical for first line innate immune defence against Staphylococcus aureus. Mature circulating PMN maintain a short half-life ending in constitutive apoptotic cell death. This makes them unlikely candidates as a bacterial intracellular niche. However, there is significant evidence to suggest that S. aureus can survive intracellularly within PMN and this contributes to persistence and dissemination during infection. The precise mechanism by which S. aureus parasitizes these cells remains to be established. Herein we propose a novel mechanism by which S. aureus subverts both autophagy and apoptosis in PMN in order to maintain an intracellular survival niche during infection. Intracellular survival of S. aureus within primary human PMN was associated with an accumulation of the autophagic flux markers LC3-II and p62, while inhibition of the autophagy pathway led to a significant reduction in intracellular survival of bacteria. This intracellular survival of S. aureus was coupled with a delay in neutrophil apoptosis as well as increased expression of several anti-apoptotic factors. Importantly, blocking autophagy in infected PMN partially restored levels of apoptosis to that of uninfected PMN, suggesting a connection between the autophagic and apoptotic pathways during intracellular survival. These results provide a novel mechanism for S. aureus intracellular survival and suggest that S. aureus may be subverting crosstalk between the autophagic and apoptosis pathways in order to maintain an intracellular niche within human PMN.
Collapse
Affiliation(s)
- Michelle E Mulcahy
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Eóin C O'Brien
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kate M O'Keeffe
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Emilio G Vozza
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Neal Leddy
- bioTEM, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
11
|
Watkins KE, Unnikrishnan M. Evasion of host defenses by intracellular Staphylococcus aureus. ADVANCES IN APPLIED MICROBIOLOGY 2020; 112:105-141. [PMID: 32762866 DOI: 10.1016/bs.aambs.2020.05.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Staphylococcus aureus is one of the leading causes of hospital and community-acquired infections worldwide. The increasing occurrence of antibiotic resistant strains and the high rates of recurrent staphylococcal infections have placed several treatment challenges on healthcare systems. In recent years, it has become evident that S. aureus is a facultative intracellular pathogen, able to invade and survive in a range of cell types. The ability to survive intracellularly provides this pathogen with yet another way to evade antibiotics and immune responses during infection. Intracellular S. aureus have been strongly linked to several recurrent infections, including severe bone infections and septicemias. S. aureus is armed with an array of virulence factors as well as an intricate network of regulators that enable it to survive, replicate and escape from a number of immune and nonimmune host cells. It is able to successfully manipulate host cell pathways and use it as a niche to multiply, disseminate, as well as persist during an infection. This bacterium is also known to adapt to the intracellular environment by forming small colony variants, which are metabolically inactive. In this review we will discuss the clinical evidence, the molecular pathways involved in S. aureus intracellular persistence, and new treatment strategies for targeting intracellular S. aureus.
Collapse
|
12
|
Vaughn JM, Abdi RD, Gillespie BE, Kerro Dego O. Genetic diversity and virulence characteristics of Staphylococcus aureus isolates from cases of bovine mastitis. Microb Pathog 2020; 144:104171. [PMID: 32224210 DOI: 10.1016/j.micpath.2020.104171] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022]
Abstract
Staphylococcus aureus is one of the major bacterial mastitis pathogens with significant effects on animal and human health. Some studies showed that S. aureus strains that infect different host species are genetically distinct, although most strains can infect a wide range of host species. However, there are no clearly defined clonal patterns of S. aureus strains that are known to infect a specific host. The objectives of this study were to evaluate the clonal diversity and virulence characteristics of S. aureus isolates from cases of bovine mastitis. Bacteriological tests were conducted on milk samples from cases of bovine mastitis from 11 dairy farms including some milk samples from unknown farms in Eastern Tennessee. Overall, a total of 111 S. aureus were isolated and identified, and further evaluated for their genetic diversity by pulsed-field gel electrophoresis (PFGE) and virulence characteristics by PCR. Genotypic virulence factors including staphylococcal enterotoxins, and toxic shock syndrome toxin 1 (tsst-1) were tested by PCR. In addition, the association among several known virulence factors of these isolates based on our current and previous studies in our lab were evaluated. Previously generated data that were included in the analysis of association among virulence factors were the presence of biofilm production associated genes in the ica operon such as icaA, icaD and icaAB, and phenotypic virulence characteristics such as hemolysis on blood agar, slime production and resistance or susceptibility to ten commonly used antimicrobials in dairy farms. The PFGE results showed the presence of 16 PFGE types (designated A - P) throughout farms, of which three pulsotypes, I, M and O were the most frequently isolated PFGE types from most farms. The PFGE type M was the most prevalent of all 16 PFGE types, with 64 isolates being present among nine farms. The PCR results of enterotoxin genes showed that out of the total 111 tested 84 (75.7%) were negative whereas 13 (11.7%), 2 (1.8%), 3 (2.7%), 1 (0.9%) and 8 (7.2%) were positive for seb, seb and sec, sec, see, and tsst-1, respectively. All 111 isolates were negative for sea and sej. Results of the evaluation of I, M and O strains adhesion to and invasion into mammary epithelial cells showed that the total count of each strain of bacteria adhered to and invaded into mammary epithelial cell line (MAC-T cells) was not significantly different (P > 0.05). This may be an indication that there is no significant difference in their ability to establish early host-pathogen interaction and colonization of the host. There were no statistically significant associations among PFGE types and other known virulence factors of these strains. However, PFGE types O and M tend to cluster with β-hemolysin, absence of enterotoxins and susceptibility to antimicrobials. In conclusion, there was not any association between pulsotype and genotypic and phenotypic virulence factors. S. aureus isolates from cases of bovine mastitis had diverse genotypes that possessed variable virulence factors.
Collapse
Affiliation(s)
- Jacqueline M Vaughn
- The University of Tennessee, Department of Animal Science, Knoxville, TN, 37996, USA
| | - Reta Duguma Abdi
- The University of Tennessee, Department of Animal Science, Knoxville, TN, 37996, USA; Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Greenvale, NY, 11548, USA
| | | | - Oudessa Kerro Dego
- The University of Tennessee, Department of Animal Science, Knoxville, TN, 37996, USA.
| |
Collapse
|
13
|
Johansson C, Rautelin H, Kaden R. Staphylococcus argenteus and Staphylococcus schweitzeri are cytotoxic to human cells in vitro due to high expression of alpha-hemolysin Hla. Virulence 2020; 10:502-510. [PMID: 31131704 PMCID: PMC6550535 DOI: 10.1080/21505594.2019.1620062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus argenteus and Staphylococcus schweitzeri are newly identified species of the S. aureus-related complex. S. argenteus, as occurring globally and showing significant prevalence and comparable infection and morbidity rates compared to S. aureus, is becoming clinically important. Whole genome sequencing has revealed the presence of several virulence genes but the molecular mechanisms of S. argenteus infection and virulence are largely unknown. Here, we studied the effect of a previously characterized clinical S. argenteus isolate on human cells in vitro. The clinical isolate, together with the S. argenteus type strain MSHR1132T and the S. schweitzeri type strain FSA084T, had a cytotoxic effect on the cells, which showed necrotic cell death after a few hours of treatment. The protein causing the cytotoxic effect was purified and identified by mass spectrometry as alpha-hemolysin, Hla, which is awell-known pore-forming toxin in S.aureus. The cytotoxic effect could be blocked with an antibody against Hla. S.argenteus showed 12–15 fold higher expression levels of hla at the RNA level and 4–6 fold higher expression levels at the protein level compared to S.aureus. The higher expression levels of hla were supported by higher RNA levels of the regulatory factors sarA and saeR. Also, the RNAIII component of the accessory gene regulator (agr) quorum sensing system was 8,000–10,000 fold higher in the S.argenteus isolates compared to S.aureus. This is the first study on the effect of S.argenteus on ahuman cell line and strengthens the idea of significant virulence of S.argenteus.
Collapse
Affiliation(s)
- Cecilia Johansson
- a Clinical Microbiology, Department of Medical Sciences , Uppsala University , Uppsala , Sweden
| | - Hilpi Rautelin
- a Clinical Microbiology, Department of Medical Sciences , Uppsala University , Uppsala , Sweden
| | - René Kaden
- a Clinical Microbiology, Department of Medical Sciences , Uppsala University , Uppsala , Sweden
| |
Collapse
|
14
|
Shah J, Rouaud F, Guerrera D, Vasileva E, Popov LM, Kelley WL, Rubinstein E, Carette JE, Amieva MR, Citi S. A Dock-and-Lock Mechanism Clusters ADAM10 at Cell-Cell Junctions to Promote α-Toxin Cytotoxicity. Cell Rep 2019; 25:2132-2147.e7. [PMID: 30463011 DOI: 10.1016/j.celrep.2018.10.088] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/01/2018] [Accepted: 10/24/2018] [Indexed: 01/08/2023] Open
Abstract
We previously identified PLEKHA7 and other junctional proteins as host factors mediating death by S. aureus α-toxin, but the mechanism through which junctions promote toxicity was unclear. Using cell biological and biochemical methods, we now show that ADAM10 is docked to junctions by its transmembrane partner Tspan33, whose cytoplasmic C terminus binds to the WW domain of PLEKHA7 in the presence of PDZD11. ADAM10 is locked at junctions through binding of its cytoplasmic C terminus to afadin. Junctionally clustered ADAM10 supports the efficient formation of stable toxin pores. Instead, disruption of the PLEKHA7-PDZD11 complex inhibits ADAM10 and toxin junctional clustering. This promotes toxin pore removal from the cell surface through an actin- and macropinocytosis-dependent process, resulting in cell recovery from initial injury and survival. These results uncover a dock-and-lock molecular mechanism to target ADAM10 to junctions and provide a paradigm for how junctions regulate transmembrane receptors through their clustering.
Collapse
Affiliation(s)
- Jimit Shah
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Florian Rouaud
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Diego Guerrera
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Ekaterina Vasileva
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland
| | - Lauren M Popov
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William L Kelley
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1211-4 Geneva, Switzerland
| | - Eric Rubinstein
- INSERM, Université Paris-Sud, UMRS_935, 94807 Villejuif Cedex, France
| | - Jan E Carette
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Manuel R Amieva
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sandra Citi
- Department of Cell Biology, Faculty of Sciences, University of Geneva, 1211-4 Geneva, Switzerland; Institute for Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211-4 Geneva, Switzerland.
| |
Collapse
|
15
|
Yue Y, Zhou T, Xu X, Sun Q, Wang C, Zhu J, Zheng F. Influence of transcription regulator SAUSA300_1968 on the virulence protein secretion and immune evasion by Staphylococcus aureus. Microb Pathog 2019; 136:103690. [DOI: 10.1016/j.micpath.2019.103690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022]
|
16
|
Singh V, Phukan UJ. Interaction of host and Staphylococcus aureus protease-system regulates virulence and pathogenicity. Med Microbiol Immunol 2019; 208:585-607. [PMID: 30483863 DOI: 10.1007/s00430-018-0573-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
Staphylococcus aureus causes various health care- and community-associated infections as well as certain chronic TH2 driven inflammatory diseases. It is a potent pathogen with serious virulence and associated high morbidity. Severe pathogenicity is accredited to the S. aureus secreted virulence factors such as proteases and host protease modulators. These virulence factors promote adhesion and invasion of bacteria through damage of tight junction barrier and keratinocytes. They inhibit activation and transmigration of various immune cells such as neutrophils (and neutrophil proteases) to evade opsono-phagocytosis and intracellular bacterial killing. Additionally, they protect the bacteria from extracellular killing by disrupting integrity of extracellular matrix. Platelet activation and agglutination is also impaired by these factors. They also block the classical as well as alternative pathways of complement activation and assist in spread of infection through blood and tissue. As these factors are exquisite factors of S. aureus mediated disease development, we have focused on review of diversification of various protease-system associated virulence factors, their structural building, diverse role in disease development and available therapeutic counter measures. This review summarises the role of protease-associated virulence factors during invasion and progression of disease.
Collapse
Affiliation(s)
- Vigyasa Singh
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, India
| | - Ujjal Jyoti Phukan
- School of Life Science, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
17
|
Forbes NS, Coffin RS, Deng L, Evgin L, Fiering S, Giacalone M, Gravekamp C, Gulley JL, Gunn H, Hoffman RM, Kaur B, Liu K, Lyerly HK, Marciscano AE, Moradian E, Ruppel S, Saltzman DA, Tattersall PJ, Thorne S, Vile RG, Zhang HH, Zhou S, McFadden G. White paper on microbial anti-cancer therapy and prevention. J Immunother Cancer 2018; 6:78. [PMID: 30081947 PMCID: PMC6091193 DOI: 10.1186/s40425-018-0381-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022] Open
Abstract
In this White Paper, we discuss the current state of microbial cancer therapy. This paper resulted from a meeting ('Microbial Based Cancer Therapy') at the US National Cancer Institute in the summer of 2017. Here, we define 'Microbial Therapy' to include both oncolytic viral therapy and bacterial anticancer therapy. Both of these fields exploit tumor-specific infectious microbes to treat cancer, have similar mechanisms of action, and are facing similar challenges to commercialization. We designed this paper to nucleate this growing field of microbial therapeutics and increase interactions between researchers in it and related fields. The authors of this paper include many primary researchers in this field. In this paper, we discuss the potential, status and opportunities for microbial therapy as well as strategies attempted to date and important questions that need to be addressed. The main areas that we think will have the greatest impact are immune stimulation, control of efficacy, control of delivery, and safety. There is much excitement about the potential of this field to treat currently intractable cancer. Much of the potential exists because these therapies utilize unique mechanisms of action, difficult to achieve with other biological or small molecule drugs. By better understanding and controlling these mechanisms, we will create new therapies that will become integral components of cancer care.
Collapse
Affiliation(s)
- Neil S Forbes
- grid.266683.f0000 0001 2184 9220Department of Chemical EngineeringUniversity of Massachusetts 159 Goessmann Hall 01003 Amherst MA USA
| | | | - Liang Deng
- 0000 0001 2171 9952grid.51462.34Department of Medicine, Memorial Sloan Kettering Cancer Center 10065 New York NY USA
| | - Laura Evgin
- 0000 0004 0459 167Xgrid.66875.3aMayo Clinic Rochester USA
| | - Steve Fiering
- 0000 0001 2179 2404grid.254880.3Geisel School of Medicine at Dartmouth Hanover USA
| | | | - Claudia Gravekamp
- 0000000121791997grid.251993.5Albert Einstein College of Medicine Bronx USA
| | - James L Gulley
- 0000 0004 1936 8075grid.48336.3aNational Cancer Institute, National Institutes of Health Bethesda USA
| | | | - Robert M Hoffman
- 0000 0001 2107 4242grid.266100.3UC, San Diego San Diego USA
- 0000 0004 0461 1271grid.417448.aAntiCancer Inc. San Diego USA
| | - Balveen Kaur
- 0000000121548364grid.55460.32University of Texas Austin USA
| | - Ke Liu
- 0000 0001 2243 3366grid.417587.8Center for Biologics Evaluation and ResearchUS Food and Drug Administration Silver Spring USA
| | | | - Ariel E Marciscano
- 0000 0004 1936 8075grid.48336.3aNational Cancer Institute, National Institutes of Health Bethesda USA
| | | | - Sheryl Ruppel
- 0000 0004 4665 8158grid.419407.fLeidos Biomedical Research, Inc. Frederick USA
| | - Daniel A Saltzman
- 0000000419368657grid.17635.36University of Minnesota Minneapolis USA
| | | | - Steve Thorne
- 0000 0004 1936 9000grid.21925.3dUniversity of Pittsburgh Pittsburgh USA
| | - Richard G Vile
- 0000 0004 0459 167Xgrid.66875.3aMayo Clinic Rochester USA
| | | | - Shibin Zhou
- 0000 0001 2171 9311grid.21107.35Johns Hopkins University Baltimore USA
| | - Grant McFadden
- 0000 0001 2151 2636grid.215654.1Center for Immunotherapy, Vaccines and Virotherapy , Biodesign InstituteArizona State University 727 E Tyler Street, Room A330E 85281 Tempe AZ USA
| |
Collapse
|
18
|
Govender J, Loos B, Marais E, Engelbrecht AM. Melatonin improves cardiac and mitochondrial function during doxorubicin-induced cardiotoxicity: A possible role for peroxisome proliferator-activated receptor gamma coactivator 1-alpha and sirtuin activity? Toxicol Appl Pharmacol 2018; 358:86-101. [PMID: 29966675 DOI: 10.1016/j.taap.2018.06.031] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/27/2018] [Accepted: 06/28/2018] [Indexed: 01/02/2023]
Abstract
Mitochondrial dysfunction is a central element in the development of doxorubicin (DXR)-induced cardiotoxicity. In this context, melatonin is known to influence mitochondrial homeostasis and function. This study aimed to investigate the effects of melatonin on cardiac function, tumor growth, mitochondrial fission and fusion, PGC1-α and sirtuin activity in an acute model of DXR-induced cardiotoxicity. During the in vitro study, H9c2 rat cardiomyoblasts were pre-treated with melatonin (10 μM, 24 h) followed by DXR exposure (3 μM, 24 h). Following treatment, cellular ATP levels and mitochondrial morphology were assessed. In the in vivo study, female Sprague Dawley rats (16 weeks old), were inoculated with a LA7 rat mammary tumor cell line and tumors were measure daily. Animals were injected with DXR (3 × 4 mg/kg) and/or received melatonin (6 mg/kg) for 14 days in their drinking water. Rat hearts were used to conduct isolated heart perfusions to assess cardiac function and thereafter, heart tissue was used for immunoblot analysis. DXR treatment increased cell death and mitochondrial fission which were reduced with melatonin treatment. Cardiac output increased in rats treated with DXR + melatonin compared to DXR-treated rats. Tumor volumes was significantly reduced in DXR + melatonin-treated rats on Day 8 in comparison to DXR-treated rats. Furthermore, DXR + melatonin treatment increased cellular ATP levels, PGC1-α and SIRT1 expression which was attenuated by DXR treatment. These results indicate that melatonin treatment confers a dual cardio-protective and oncostatic effect by improving mitochondrial function and cardiac function whilst simultaneously retarding tumor growth during DXR-induced cardiotoxicity.
Collapse
Affiliation(s)
- Jenelle Govender
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa.
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Erna Marais
- Department of Medical Physiology, Faculty of Medicine, Stellenbosch University, Tygerberg Campus, 7505, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
19
|
Moyano AJ, Racca AC, Soria G, Saka HA, Andreoli V, Smania AM, Sola C, Bocco JL. c-Jun Proto-Oncoprotein Plays a Protective Role in Lung Epithelial Cells Exposed to Staphylococcal α-Toxin. Front Cell Infect Microbiol 2018; 8:170. [PMID: 29888211 PMCID: PMC5981160 DOI: 10.3389/fcimb.2018.00170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/03/2018] [Indexed: 12/17/2022] Open
Abstract
c-Jun is a member of the early mammalian transcriptional regulators belonging to the AP-1 family, which participates in a wide range of cellular processes such as proliferation, apoptosis, tumorigenesis, and differentiation. Despite its established role in cell survival upon stress, its participation in the stress response induced by bacterial infections has been poorly investigated. To study the potential role of c-Jun in this context we choose the widely studied α-toxin produced by Staphylococcus aureus, a pore-forming toxin that is a critical virulence factor in the pathogenesis of these bacteria. We analyzed the effect of α-toxin treatment in the activation, expression, and protein levels of c-Jun in A549 lung epithelial cells. Furthermore, we explored the role of c-Jun in the cellular fate after exposure to α-toxin. Our results show that staphylococcal α-toxin per se is able to activate c-Jun by inducing phosphorylation of its Serine 73 residue. Silencing of the JNK (c-Jun N-terminal Kinase) signaling pathway abrogated most of this activation. On the contrary, silencing of the ERK (Extracellular Signal-Regulated Kinase) pathway exacerbated this response. Intriguingly, while the exposure to α-toxin induced a marked increase in the levels of c-Jun transcripts, c-Jun protein levels noticeably decreased in the same time-frame as a consequence of active proteolytic degradation through the proteasome-dependent pathway. In addition, we established that c-Jun promoted cell survival when cells were challenged with α-toxin. Similarly, c-Jun phosphorylation was also induced in cells upon intoxication with the cytolysin produced by Vibrio cholerae in a JNK-dependent manner, suggesting that c-Jun-JNK axis would be a conserved responsive cellular pathway to pore-forming toxins. This study contributes to understanding the role of the multifaceted c-Jun proto-oncoprotein in cell response to bacterial pore-forming toxins, positioning it as a relevant component of the complex early machinery mounted to deal with staphylococcal infections.
Collapse
Affiliation(s)
- Alejandro J Moyano
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ana C Racca
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gastón Soria
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Héctor A Saka
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Verónica Andreoli
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea M Smania
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Química Biológica de Córdoba, CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Claudia Sola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - José L Bocco
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
20
|
Soto C, Bergado G, Blanco R, Griñán T, Rodríguez H, Ros U, Pazos F, Lanio ME, Hernández AM, Álvarez C. Sticholysin II-mediated cytotoxicity involves the activation of regulated intracellular responses that anticipates cell death. Biochimie 2018; 148:18-35. [DOI: 10.1016/j.biochi.2018.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 02/07/2018] [Indexed: 12/12/2022]
|
21
|
Thomas M, Davis T, Loos B, Sishi B, Huisamen B, Strijdom H, Engelbrecht AM. Autophagy is essential for the maintenance of amino acids and ATP levels during acute amino acid starvation in MDAMB231 cells. Cell Biochem Funct 2018; 36:65-79. [PMID: 29399832 DOI: 10.1002/cbf.3318] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/20/2017] [Accepted: 12/26/2017] [Indexed: 12/17/2022]
Abstract
Autophagy plays a major role in the adaptive metabolic response of cancer cells during adverse conditions such as nutrient deprivation. However, specific data that assess metabolite profiles in context with adenosine triphosphate (ATP) availability and cell death susceptibility remain limited. Human breast cancer cells, MDAMB231, and normal breast epithelial cells, MCF12A, were subjected to short-term amino acid starvation and the cellular apoptotic and autophagic responses assessed. The role of autophagy in the control of cellular amino acid, ATP, free fatty acid, and glucose levels during amino acid starvation were compared. We demonstrate that breast cancer cells have an increased metabolic demand contributing to significant amino acid and ATP depletion in a nutrient-poor environment. Upregulation of autophagy was important for the generation of amino acids and free fatty acids and maintenance of cellular ATP levels. In contrast to normal cells, breast cancer cells were unable to maintain the response after 12 hours of amino acid starvation. Regulation of autophagic activity in these environments had indirect consequences on cell death susceptibility. Overall, our data provide support for autophagy as an important survival mechanism capable of providing metabolic substrates when cancer cells are faced with nutrient-deprived environments. SIGNIFICANCE OF STUDY The results obtained in this study helps to expand our current knowledge on how cells respond to environmental changes; the biochemical and metabolic consequences and the physiological processes activated in response. The environmental stress applied in this study is relevant to tumour physiology, and results can be translated to cancer therapeutic and clinical research areas, ultimately assisting in the specific targeting of cancer cells while avoiding harm to normal cells.
Collapse
Affiliation(s)
- Mark Thomas
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Tanja Davis
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Balindiwe Sishi
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Barbara Huisamen
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,Diabetes Discovery Platform, Medical Research Council, Cape Town, South Africa
| | - Hans Strijdom
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
22
|
Computational Modeling of the Staphylococcal Enterotoxins and Their Interaction with Natural Antitoxin Compounds. Int J Mol Sci 2018; 19:ijms19010133. [PMID: 29301344 PMCID: PMC5796082 DOI: 10.3390/ijms19010133] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/26/2017] [Accepted: 12/27/2017] [Indexed: 01/08/2023] Open
Abstract
Staphylococcus aureus is an opportunistic bacterium that produces various types of toxins, resulting in serious food poisoning. Staphylococcal enterotoxins (SEs) are heat-stable and resistant to hydrolysis by digestive enzymes, representing a potential hazard for consumers worldwide. In the present study, we used amino-acid sequences encoding SEA and SEB-like to identify their respective template structure and build the three-dimensional (3-D) models using homology modeling method. Two natural compounds, Betulin and 28-Norolean-12-en-3-one, were selected for docking study on the basis of the criteria that they satisfied the Lipinski’s Rule-of-Five. A total of 14 and 13 amino-acid residues were present in the best binding site predicted in the SEA and SEB-like, respectively, using the Computer Atlas of Surface Topology of Proteins (CASTp). Among these residues, the docking study with natural compounds Betulin and 28-Norolean-12-en-3-one revealed that GLN43 and GLY227 in the binding site of the SEA, each formed a hydrogen-bond interaction with 28-Norolean-12-en-3-one; while GLY227 residue established a hydrogen bond with Betulin. In the case of SEB-like, the docking study demonstrated that ASN87 and TYR88 residues in its binding site formed hydrogen bonds with Betulin; whereas HIS59 in the binding site formed a hydrogen-bond interaction with 28-Norolean-12-en-3-one. Our results demonstrate that the toxic effects of these two SEs can be effectively treated with antitoxins like Betulin and 28-Norolean-12-en-3-one, which could provide an effective drug therapy for this pathogen.
Collapse
|
23
|
Zhang X, Hu X, Rao X. Apoptosis induced by Staphylococcus aureus toxins. Microbiol Res 2017; 205:19-24. [DOI: 10.1016/j.micres.2017.08.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/31/2017] [Accepted: 08/09/2017] [Indexed: 10/19/2022]
|
24
|
Pietrocola G, Nobile G, Rindi S, Speziale P. Staphylococcus aureus Manipulates Innate Immunity through Own and Host-Expressed Proteases. Front Cell Infect Microbiol 2017; 7:166. [PMID: 28529927 PMCID: PMC5418230 DOI: 10.3389/fcimb.2017.00166] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 04/18/2017] [Indexed: 01/29/2023] Open
Abstract
Neutrophils, complement system and skin collectively represent the main elements of the innate immune system, the first line of defense of the host against many common microorganisms. Bacterial pathogens have evolved strategies to counteract all these defense activities. Specifically, Staphylococcus aureus, a major human pathogen, secretes a variety of immune evasion molecules including proteases, which cleave components of the innate immune system or disrupt the integrity of extracellular matrix and intercellular connections of tissues. Additionally, S. aureus secretes proteins that can activate host zymogens which, in turn, target specific defense components. Secreted proteins can also inhibit the anti-bacterial function of neutrophils or complement system proteases, potentiating S. aureus chances of survival. Here, we review the current understanding of these proteases and modulators of host proteases in the functioning of innate immunity and describe the importance of these mechanisms in the pathology of staphylococcal diseases.
Collapse
Affiliation(s)
- Giampiero Pietrocola
- Unit of Biochemistry, Department of Molecular Medicine, University of PaviaPavia, Italy
| | - Giulia Nobile
- Unit of Biochemistry, Department of Molecular Medicine, University of PaviaPavia, Italy
| | - Simonetta Rindi
- Unit of Biochemistry, Department of Molecular Medicine, University of PaviaPavia, Italy
| | - Pietro Speziale
- Unit of Biochemistry, Department of Molecular Medicine, University of PaviaPavia, Italy
| |
Collapse
|
25
|
Vdovikova S, Luhr M, Szalai P, Nygård Skalman L, Francis MK, Lundmark R, Engedal N, Johansson J, Wai SN. A Novel Role of Listeria monocytogenes Membrane Vesicles in Inhibition of Autophagy and Cell Death. Front Cell Infect Microbiol 2017; 7:154. [PMID: 28516064 PMCID: PMC5413512 DOI: 10.3389/fcimb.2017.00154] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/12/2017] [Indexed: 11/13/2022] Open
Abstract
Bacterial membrane vesicle (MV) production has been mainly studied in Gram-negative species. In this study, we show that Listeria monocytogenes, a Gram-positive pathogen that causes the food-borne illness listeriosis, produces MVs both in vitro and in vivo. We found that a major virulence factor, the pore-forming hemolysin listeriolysin O (LLO), is tightly associated with the MVs, where it resides in an oxidized, inactive state. Previous studies have shown that LLO may induce cell death and autophagy. To monitor possible effects of LLO and MVs on autophagy, we performed assays for LC3 lipidation and LDH sequestration as well as analysis by confocal microscopy of HEK293 cells expressing GFP-LC3. The results revealed that MVs alone did not affect autophagy whereas they effectively abrogated autophagy induced by pure LLO or by another pore-forming toxin from Vibrio cholerae, VCC. Moreover, Listeria monocytogenes MVs significantly decreased Torin1-stimulated macroautophagy. In addition, MVs protected against necrosis of HEK293 cells caused by the lytic action of LLO. We explored the mechanisms of LLO-induced autophagy and cell death and demonstrated that the protective effect of MVs involves an inhibition of LLO-induced pore formation resulting in inhibition of autophagy and the lytic action on eukaryotic cells. Further, we determined that these MVs help bacteria to survive inside eukaryotic cells (mouse embryonic fibroblasts). Taken together, these findings suggest that intracellular release of MVs from L. monocytogenes may represent a bacterial strategy to survive inside host cells, by its control of LLO activity and by avoidance of destruction from the autophagy system during infection.
Collapse
Affiliation(s)
- Svitlana Vdovikova
- Department of Molecular Biology, Umeå UniversityUmeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Umeå Centre for Microbial Research, Umeå UniversityUmeå, Sweden
| | - Morten Luhr
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of OsloOslo, Norway
| | - Paula Szalai
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of OsloOslo, Norway
| | - Lars Nygård Skalman
- Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Department of Medical Biochemistry and Biophysics, Umeå UniversityUmeå, Sweden
| | - Monika K Francis
- Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Department of Medical Biochemistry and Biophysics, Umeå UniversityUmeå, Sweden
| | - Richard Lundmark
- Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Department of Medical Biochemistry and Biophysics, Umeå UniversityUmeå, Sweden.,Department of Integrative Medical Biology, Umeå UniversityUmeå, Sweden
| | - Nikolai Engedal
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of OsloOslo, Norway
| | - Jörgen Johansson
- Department of Molecular Biology, Umeå UniversityUmeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Umeå Centre for Microbial Research, Umeå UniversityUmeå, Sweden
| | - Sun N Wai
- Department of Molecular Biology, Umeå UniversityUmeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Umeå Centre for Microbial Research, Umeå UniversityUmeå, Sweden
| |
Collapse
|
26
|
Eiffler I, Behnke J, Ziesemer S, Müller C, Hildebrandt JP. Staphylococcus aureus α-toxin-mediated cation entry depolarizes membrane potential and activates p38 MAP kinase in airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2016; 311:L676-85. [PMID: 27496896 DOI: 10.1152/ajplung.00090.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/29/2016] [Indexed: 11/22/2022] Open
Abstract
Membrane potential (Vm)-, Na(+)-, or Ca(2+)-sensitive fluorescent dyes were used to analyze changes in Vm or intracellular ion concentrations in airway epithelial cells treated with Staphylococcus aureus α-toxin (Hla), a major virulence factor of pathogenic strains of these bacteria. Gramicidin, a channel-forming peptide causing membrane permeability to monovalent cations, a mutated form of Hla, rHla-H35L, which forms oligomers in the plasma membranes of eukaryotic cells but fails to form functional transmembrane pores, or the cyclodextrin-derivative IB201, a blocker of the Hla pore, were used to investigate the permeability of the pore. Na(+) as well as Ca(2+) ions were able to pass the Hla pore and accumulated in the cytosol. The pore-mediated influx of calcium ions was blocked by IB201. Treatment of cells with recombinant Hla resulted in plasma membrane depolarization as well as in increases in the phosphorylation levels of paxillin (signaling pathway mediating disruption of the actin cytoskeleton) and p38 MAP kinase (signaling pathway resulting in defensive actions). p38 MAP kinase phosphorylation, but not paxillin phosphorylation, was elicited by treatment of cells with gramicidin. Although treatment of cells with rHla-H35L resulted in the formation of membrane-associated heptamers, none of these cellular effects were observed in our experiments. This indicates that formation of functional Hla-transmembrane pores is required to induce the cell physiological changes mediated by α-toxin. Specifically, the changes in ion equilibria and plasma membrane potential are important activators of p38 MAP kinase, a signal transduction module involved in host cell defense.
Collapse
Affiliation(s)
- Ina Eiffler
- Animal Physiology and Biochemistry, Ernst Moritz Arndt-University, Greifswald, Germany
| | - Jane Behnke
- Animal Physiology and Biochemistry, Ernst Moritz Arndt-University, Greifswald, Germany
| | - Sabine Ziesemer
- Animal Physiology and Biochemistry, Ernst Moritz Arndt-University, Greifswald, Germany
| | - Christian Müller
- Animal Physiology and Biochemistry, Ernst Moritz Arndt-University, Greifswald, Germany
| | - Jan-Peter Hildebrandt
- Animal Physiology and Biochemistry, Ernst Moritz Arndt-University, Greifswald, Germany
| |
Collapse
|
27
|
Jagot-Lacoussiere L, Kotula E, Villoutreix BO, Bruzzoni-Giovanelli H, Poyet JL. A Cell-Penetrating Peptide Targeting AAC-11 Specifically Induces Cancer Cells Death. Cancer Res 2016; 76:5479-90. [DOI: 10.1158/0008-5472.can-16-0302] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/24/2016] [Indexed: 11/16/2022]
|
28
|
Kwok AHY, Wang Y, Ho WS. Cytotoxic and pro-oxidative effects of Imperata cylindrica aerial part ethyl acetate extract in colorectal cancer in vitro. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:558-565. [PMID: 27064015 DOI: 10.1016/j.phymed.2016.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 01/26/2016] [Accepted: 02/14/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer. Its global incidence and mortality have been on the rise. Recent strategy of therapies has involved the use of non-steroid anti-inflammatory drugs and cyclooxygenase-selective inhibitors. Aerial parts of Imperata cylindrical L. Raeusch (IMP) have been used as an anti-inflammatory agent in traditional Chinese medicine. HYPOTHESIS Asarachidonate acid cascadeis often involved in inflammation-related malignancy and IMP is an anti-inflammatory agent, hence it is hypothesized that IMP aerial part ethyl acetate extract exerts cytotoxic effects on colorectal cancer cells in vitro. STUDY DESIGN The HT-29 adenocarcinoma cell line was used to elucidate its pro-apoptotic activities. Flow cytometry and fluorescent microscopy were performed to assess cell cycle arrest and the accumulation of reactive oxygen species (ROS). The mRNA and hormone levels of arachidonate acid pathways were studied via quantitative reverse transcription PCR (qRT-PCR) and ELISA. RESULTS The 50% growth inhibitory effect (GI50) of the IMP extract on HT-29 was measured with a value of 14.5 µg/ml. Immuno-blot and caspase-3/7 activity assay showed the pro-apoptotic effect of IMP on the activation of caspase cascade. G2/M arrest was observed via flow cytometry. The ROS activity was modulated by the IMP extraction a concentration-dependent manner in HT-29 cells. The IMP extract increased PGE2 and PGF2α levels qRT-PCR revealed that transcripts of rate-limiting PGE2- and PGF2α-biosynthetic enzymes - COX-1, mPGES1 and AKR1C3 were notably up-regulated. Among the prostanoid receptors, EP1 and FP transcripts were up-regulated while EP4 transcripts decreased. The findings suggest that the proliferative effect of PGE2, which is generally believed to associate with heightened DNA synthesis and cross-talk with MAPK pathways, is likely triggered by the pro-apoptotic or -oxidative effects exerted by IMP extract in HT-29 cells. Concurring with this notion, indomethacin (COX-1/2-inhibitor) was demonstrated to potentiate the cytotoxic effect of IMP extract (GI50 ≦ 10.8 µg/ml). The results show that the cytotoxic effect of IMP extract predominates over the influence of proliferative prostanoids released by challenged colorectal cancer cells, and may present a potential source for development of novel anti-cancer drugs.
Collapse
Affiliation(s)
- Amy Ho Yan Kwok
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Yan Wang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Wing Shing Ho
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
29
|
Einsele-Scholz S, Malmsheimer S, Bertram K, Stehle D, Johänning J, Manz M, Daniel PT, Gillissen BF, Schulze-Osthoff K, Essmann F. Bok is a genuine multi-BH-domain protein that triggers apoptosis in the absence of Bax and Bak. J Cell Sci 2016; 129:2213-23. [PMID: 27076518 DOI: 10.1242/jcs.181727] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/11/2016] [Indexed: 12/15/2022] Open
Abstract
The pro-apoptotic multidomain Bcl-2 proteins Bax and Bak (also known as BAK1) are considered the gatekeepers of the intrinsic pathway of apoptosis by triggering the mitochondrial release of cytochrome c The role of the third Bax- and Bak-homologous multidomain protein Bok, however, is still unresolved. As cells doubly deficient for Bax and Bak are largely resistant to various apoptotic stimuli, it has been proposed that Bok is either dispensable for apoptosis or that its role is dependent on Bax and Bak. Here, we demonstrate, in several cell systems, that Bok efficiently induces cytochrome c release and apoptosis even in the complete absence of both Bak and Bax. Moreover, modulation of endogenous Bok levels affects the apoptosis response. By RNA interference and targeted deletion of the Bok gene, we demonstrate that Bok can significantly influence the apoptotic response to chemotherapeutic drugs in ovarian carcinoma cells. Hence, our results not only establish Bok as a Bak- and Bax-independent apoptosis inducer, but also suggest a potential impact of Bok expression in ovarian cancer therapy.
Collapse
Affiliation(s)
- Stephanie Einsele-Scholz
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard Karls University, Tübingen 72076, Germany
| | - Silke Malmsheimer
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard Karls University, Tübingen 72076, Germany
| | - Katrin Bertram
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard Karls University, Tübingen 72076, Germany
| | - Daniel Stehle
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard Karls University, Tübingen 72076, Germany
| | - Janina Johänning
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard Karls University, Tübingen 72076, Germany
| | - Marianne Manz
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard Karls University, Tübingen 72076, Germany
| | - Peter T Daniel
- Department of Hematology, Oncology and Tumor Immunology, University Medical Center Charité, Humboldt University, Berlin 13125, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Bernhard F Gillissen
- Department of Hematology, Oncology and Tumor Immunology, University Medical Center Charité, Humboldt University, Berlin 13125, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Klaus Schulze-Osthoff
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard Karls University, Tübingen 72076, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Frank Essmann
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard Karls University, Tübingen 72076, Germany German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| |
Collapse
|
30
|
Genome-wide CRISPR screen reveals novel host factors required for Staphylococcus aureus α-hemolysin-mediated toxicity. Sci Rep 2016; 6:24242. [PMID: 27066838 PMCID: PMC4828653 DOI: 10.1038/srep24242] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/23/2016] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus causes a wide variety of infections and antibiotic resistant strains are a major problem in hospitals. One of the best studied virulence factors of S. aureus is the pore-forming toxin alpha hemolysin (αHL) whose mechanism of action is incompletely understood. We performed a genome-wide loss-of-function screen using CRISPR/Cas9 technology to identify host targets required for αHL susceptibility in human myeloid cells. We found gRNAs for ten genes enriched after intoxication with αHL and focused on the top five hits. Besides a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10), the host receptor for αHL, we identified three proteins, Sys1 golgi trafficking protein (SYS1), ADP-ribosylation factor 1 (ARFRP1), and tetraspanin-14 (TSPAN14) which regulate the presentation of ADAM10 on the plasma membrane post-translationally. Interestingly, we also showed that cells lacking sphingomyelin synthase 1 (SGMS1) resist αHL intoxication, but have only a slightly reduced ADAM10 surface expression. SGMS1 regulates lipid raft formation, suggesting that αHL requires these membrane microdomains for attachment and cytotoxicity.
Collapse
|
31
|
Goldmann O, Tuchscherr L, Rohde M, Medina E. α-Hemolysin enhances Staphylococcus aureus internalization and survival within mast cells by modulating the expression of β1 integrin. Cell Microbiol 2016; 18:807-19. [PMID: 26595647 DOI: 10.1111/cmi.12550] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 11/06/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Abstract
Mast cells (MCs) are important sentinels of the host defence against invading pathogens. We previously reported that Staphylococcus aureus evaded the extracellular antimicrobial activities of MCs by promoting its internalization within these cells via β1 integrins. Here, we investigated the molecular mechanisms governing this process. We found that S. aureus responded to the antimicrobial mediators released by MCs by up-regulating the expression of α-hemolysin (Hla), fibronectin-binding protein A and several regulatory systems. We also found that S. aureus induced the up-regulation of β1 integrin expression on MCs and that this effect was mediated by Hla-ADAM10 (a disintegrin and metalloproteinase 10) interaction. Thus, deletion of Hla or inhibition of Hla-ADAM10 interaction significantly impaired S. aureus internalization within MCs. Furthermore, purified Hla but not the inactive HlaH35L induced up-regulation of β1 integrin expression in MCs in a dose-dependent manner. Our data support a model in which S. aureus counter-reacts the extracellular microbicidal mechanisms of MCs by increasing expression of fibronectin-binding proteins and by inducing Hla-ADAM10-mediated up-regulation of β1 integrin in MCs. The up-regulation of bacterial fibronectin-binding proteins, concomitantly with the increased expression of its receptor β1 integrin on the MCs, resulted in enhanced S. aureus internalization through the binding of fibronectin-binding proteins to integrin β1 via fibronectin.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Lorena Tuchscherr
- Institute of Medical Microbiology, University Hospital of Jena, Erlanger Allee 101, 07747, Jena, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| |
Collapse
|
32
|
Josse J, Velard F, Gangloff SC. Staphylococcus aureus vs. Osteoblast: Relationship and Consequences in Osteomyelitis. Front Cell Infect Microbiol 2015; 5:85. [PMID: 26636047 PMCID: PMC4660271 DOI: 10.3389/fcimb.2015.00085] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/10/2015] [Indexed: 12/11/2022] Open
Abstract
Bone cells, namely osteoblasts and osteoclasts work in concert and are responsible for bone extracellular matrix formation and resorption. This homeostasis is, in part, altered during infections by Staphylococcus aureus through the induction of various responses from the osteoblasts. This includes the over-production of chemokines, cytokines and growth factors, thus suggesting a role for these cells in both innate and adaptive immunity. S. aureus decreases the activity and viability of osteoblasts, by induction of apoptosis-dependent and independent mechanisms. The tight relationship between osteoclasts and osteoblasts is also modulated by S. aureus infection. The present review provides a survey of the relevant literature discussing the important aspects of S. aureus and osteoblast interaction as well as the ability for antimicrobial peptides to kill intra-osteoblastic S. aureus, hence emphasizing the necessity for new anti-infectious therapeutics.
Collapse
Affiliation(s)
- Jérôme Josse
- EA 4691 Biomatériaux et inflammation en site osseux, Pôle Santé, Université de Reims Champagne-Ardenne Reims, France
| | - Frédéric Velard
- EA 4691 Biomatériaux et inflammation en site osseux, Pôle Santé, Université de Reims Champagne-Ardenne Reims, France
| | - Sophie C Gangloff
- EA 4691 Biomatériaux et inflammation en site osseux, Pôle Santé, Université de Reims Champagne-Ardenne Reims, France
| |
Collapse
|
33
|
The ever-emerging complexity of α-toxin's interaction with host cells. Proc Natl Acad Sci U S A 2015; 112:14123-4. [PMID: 26542682 DOI: 10.1073/pnas.1519766112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
34
|
Rashidieh B, Etemadiafshar S, Memari G, Mirzaeichegeni M, Yazdi S, Farsimadan F, Alizadeh S. A molecular modeling based screening for potential inhibitors to alpha hemolysin from Staphylococcus aureus. Bioinformation 2015; 11:373-7. [PMID: 26420917 PMCID: PMC4574119 DOI: 10.6026/97320630011373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/01/2015] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus, a Gram-positive bacterium is pathogenic in nature. It is known that secreted toxins remain active after
antibiotic treatment. The alpha hemolysin or alpha toxin damages cell membrane and induces apoptosis and degradation of DNA.
The titer of alphahemolysin increases and causes hemostasis disturbances, thrombocytopenia, and pulmonary lesions during
staphylococcal infection. Therefore, it is of interest to inhibit alpha hemolysin using novel compounds. We used the structure of
alpha hemolysin(PDB: 7AHL) to screen structures for 100,000 compounds from the ZINC database using molecular docking with
AutoDock VINA. Nine (9) successive hits were then subjected for pharmacokinetic and toxicity properties by PROTOX (a
webserver for the prediction of oral toxicities of small molecules) and FAFDrugs (a tool for prediction of ADME and Toxicity). This
exercise further identified hit #1 ({[3a-(Dihydroxymethyl)-6-hydroxy-2,2-dimethyl-1,3,4-trioxatetrahydro-2H-pentalen-5-
yl]methyl}amino(9H-fluoren-9-yl)acetate with binding affinity: -10.3 kcal/mol) and hit #2 (6-(Dihydroxymethyl)-2-{2-[3-
(methylamino)propyl]-2-azatricyclo[9.4.0.03,8]pentadeca-1(11),3,5,7,12,14-hexaen-6-yloxy}tetrahydro-2H-pyran-3,4,5-triol with
binding affinity: -9.6 kcal/mol) with acceptable toxicity and ADME properties for potential predicted hemolysin inhibition. These
compounds should then be evaluated in vitro using inhibitory studies.
Collapse
|
35
|
Yang J, Liang X, Ji Y. The novel transcriptional regulator SA1804 Is involved in mediating the invasion and cytotoxicity of Staphylococcus aureus. Front Microbiol 2015; 6:174. [PMID: 25806024 PMCID: PMC4353350 DOI: 10.3389/fmicb.2015.00174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/16/2015] [Indexed: 11/24/2022] Open
Abstract
The two-component regulatory system, SaeRS, controls expression of important virulence factors, including toxins and invasins, which contribute to the pathogenicity of Staphylococcus aureus. Previously, we conducted a transcriptomics study for identification of SaeRS regulon and found that inactivation of SaeRS dramatically enhances the transcription of a novel transcriptional regulator (SA1804). This led us to question whether SA1804 is involved in bacterial pathogenicity by regulating the expression of virulence factors. To address this question, we created sa1804, saeRS, and sa1804/saeRS double deletion mutants in a USA300 community-acquired MRSA strain, 923, and determined their impact on the pathogenicity. The deletion of sa1804 dramatically increased the cytotoxicity and enhanced the capacity of bacteria to invade into the epithelial cells (A549), whereas the deletion of saeRS eliminated the cytotoxicity and abolished the bacterial ability to invade into the epithelial cells. Moreover, the double deletions of sa1804 and saeRS appeared a similar phenotype with the saeRS null mutation. Furthermore, we determined the regulatory mechanism of SA1804 using qPCR and gel-shift approaches. Our data indicate that the novel virulence repressor SA1804 is dependent on the regulation of SaeRS. This study sheds light on the regulatory mechanism of virulence factors and allows for us further elucidate the molecular pathogenesis of S. aureus.
Collapse
Affiliation(s)
- Junshu Yang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota Saint Paul, MN, USA
| | - Xudong Liang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota Saint Paul, MN, USA
| | - Yinduo Ji
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota Saint Paul, MN, USA
| |
Collapse
|
36
|
Cole J, Aberdein J, Jubrail J, Dockrell DH. The role of macrophages in the innate immune response to Streptococcus pneumoniae and Staphylococcus aureus: mechanisms and contrasts. Adv Microb Physiol 2014; 65:125-202. [PMID: 25476766 DOI: 10.1016/bs.ampbs.2014.08.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Macrophages are critical mediators of innate immune responses against bacteria. The Gram-positive bacteria Streptococcus pneumoniae and Staphylococcus aureus express a range of virulence factors, which challenge macrophages' immune competence. We review how macrophages respond to this challenge. Macrophages employ a range of strategies to phagocytose and kill each pathogen. When the macrophages capacity to clear bacteria is overwhelmed macrophages play important roles in orchestrating the inflammatory response through pattern recognition receptor-mediated responses. Macrophages also ensure the inflammatory response is tightly constrained, to avoid tissue damage, and play an important role in downregulating the inflammatory response once initial bacterial replication is controlled.
Collapse
Affiliation(s)
- Joby Cole
- Department of Infection and Immunity, University of Sheffield Medical School and Sheffield Teaching Hospitals, Sheffield, United Kingdom
| | - Jody Aberdein
- Department of Infection and Immunity, University of Sheffield Medical School and Sheffield Teaching Hospitals, Sheffield, United Kingdom
| | - Jamil Jubrail
- Department of Infection and Immunity, University of Sheffield Medical School and Sheffield Teaching Hospitals, Sheffield, United Kingdom
| | - David H Dockrell
- Department of Infection and Immunity, University of Sheffield Medical School and Sheffield Teaching Hospitals, Sheffield, United Kingdom.
| |
Collapse
|
37
|
Bacterial delivery of Staphylococcus aureus α-hemolysin causes regression and necrosis in murine tumors. Mol Ther 2014; 22:1266-1274. [PMID: 24590046 DOI: 10.1038/mt.2014.36] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 02/10/2014] [Indexed: 01/12/2023] Open
Abstract
Bacterial therapies, designed to manufacture therapeutic proteins directly within tumors, could eliminate cancers that are resistant to other therapies. To be effective, a payload protein must be secreted, diffuse through tissue, and efficiently kill cancer cells. To date, these properties have not been shown for a single protein. The gene for Staphylococcus aureus α-hemolysin (SAH), a pore-forming protein, was cloned into Escherichia coli. These bacteria were injected into tumor-bearing mice and volume was measured over time. The location of SAH relative to necrosis and bacterial colonies was determined by immunohistochemistry. In culture, SAH was released and killed 93% of cancer cells in 24 hours. Injection of SAH-producing bacteria reduced viable tissue to 9% of the original tumor volume. By inducing cell death, SAH moved the boundary of necrosis toward the tumor edge. SAH diffused 6.8 ± 0.3 µm into tissue, which increased the volume of affected tissue from 48.6 to 3,120 µm(3). A mathematical model of molecular transport predicted that SAH efficacy is primarily dependent on colony size and the rate of protein production. As a payload protein, SAH will enable effective bacterial therapy because of its ability to diffuse in tissue, kill cells, and expand tumor necrosis.
Collapse
|
38
|
Vivekananda J, Salgado C, Millenbaugh NJ. DNA aptamers as a novel approach to neutralize Staphylococcus aureus α-toxin. Biochem Biophys Res Commun 2014; 444:433-8. [PMID: 24472539 DOI: 10.1016/j.bbrc.2014.01.076] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 01/20/2014] [Indexed: 11/18/2022]
Abstract
Staphylococcus aureus is a versatile pathogen capable of causing a broad spectrum of diseases ranging from superficial skin infections to life threatening conditions such as endocarditis, septicemia, pneumonia and toxic shock syndrome. In vitro and in vivo studies identified an exotoxin, α-toxin, as a major cause of S. aureus toxicity. Because S. aureus has rapidly evolved resistance to a number of antibiotics, including methicillin, it is important to identify new therapeutic strategies, other than antibiotics, for inhibiting the harmful effects of this pathogen. Aptamers are single-stranded DNA or RNA oligonucleotides with three-dimensional folded conformations that bind with high affinity and selectivity to targets and modulate their biological functions. The goal of this study was to isolate DNA aptamers that specifically inhibit the cytotoxic activity of α-toxin. After 10 rounds of Systematic Evolution of Ligands by EXponential Enrichment (SELEX), 49 potential anti-α-toxin aptamers were identified. In vitro neutralization assays demonstrated that 4 of these 49 aptamers, AT-27, AT-33, AT-36, and AT-49, significantly inhibited α-toxin-mediated cell death in Jurkat T cells. Furthermore, RT-PCR analysis revealed that α-toxin increased the transcription of the inflammatory cytokines TNF-α and IL-17 and that anti-α-toxin aptamers AT-33 and AT-36 inhibited the upregulation of these genes. Collectively, the data suggest the feasibility of generating functionally effective aptamers against α-toxin for treatment of S. aureus infections.
Collapse
Affiliation(s)
- Jeevalatha Vivekananda
- Maxillofacial Injury and Disease Department, Craniofacial Health and Restorative Medicine, Naval Medical Research Unit San Antonio, JBSA-Fort Sam Houston, TX 78234, USA
| | - Christi Salgado
- Maxillofacial Injury and Disease Department, Craniofacial Health and Restorative Medicine, Naval Medical Research Unit San Antonio, JBSA-Fort Sam Houston, TX 78234, USA
| | - Nancy J Millenbaugh
- Maxillofacial Injury and Disease Department, Craniofacial Health and Restorative Medicine, Naval Medical Research Unit San Antonio, JBSA-Fort Sam Houston, TX 78234, USA.
| |
Collapse
|
39
|
YU FENGLING, LIU TINGTING, ZHU XIANG, YANG WENGXUAN, ZHANG TAO, LIN NA, LIU YONG, LIU CONGSEN, JIANG JIU, GUAN JUNCHANG. Staphylococcal enterotoxin B and α-toxin induce the apoptosis of ECV304 cells via similar mechanisms. Mol Med Rep 2013; 8:591-6. [DOI: 10.3892/mmr.2013.1550] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 06/13/2013] [Indexed: 11/06/2022] Open
|
40
|
Autheman D, Wyder M, Popoff M, D’Herde K, Christen S, Posthaus H. Clostridium perfringens beta-toxin induces necrostatin-inhibitable, calpain-dependent necrosis in primary porcine endothelial cells. PLoS One 2013; 8:e64644. [PMID: 23734212 PMCID: PMC3667183 DOI: 10.1371/journal.pone.0064644] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 04/17/2013] [Indexed: 11/30/2022] Open
Abstract
Clostridium perfringens β-toxin (CPB) is a β-barrel pore-forming toxin and an essential virulence factor of C. perfringens type C strains, which cause fatal hemorrhagic enteritis in animals and humans. We have previously shown that CPB is bound to endothelial cells within the intestine of affected pigs and humans, and that CPB is highly toxic to primary porcine endothelial cells (pEC) in vitro. The objective of the present study was to investigate the type of cell death induced by CPB in these cells, and to study potential host cell mechanisms involved in this process. CPB rapidly induced lactate dehydrogenase (LDH) release, propidium iodide uptake, ATP depletion, potassium efflux, a marked rise in intracellular calcium [Ca2+]i, release of high-mobility group protein B1 (HMGB1), and caused ultrastructural changes characteristic of necrotic cell death. Despite a certain level of caspase-3 activation, no appreciable DNA fragmentation was detected. CPB-induced LDH release and propidium iodide uptake were inhibited by necrostatin-1 and the two dissimilar calpain inhibitors PD150606 and calpeptin. Likewise, inhibition of potassium efflux, chelation of intracellular calcium and treatment of pEC with cyclosporin A also significantly inhibited CPB-induced LDH release. Our results demonstrate that rCPB primarily induces necrotic cell death in pEC, and that necrotic cell death is not merely a passive event caused by toxin-induced membrane disruption, but is propagated by host cell-dependent biochemical pathways activated by the rise in intracellular calcium and inhibitable by necrostatin-1, consistent with the emerging concept of programmed necrosis (“necroptosis”).
Collapse
Affiliation(s)
- Delphine Autheman
- Institute of Infectious Diseases, Medical Faculty, University of Bern, Bern, Switzerland
| | - Marianne Wyder
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Katharina D’Herde
- Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Stephan Christen
- Institute of Infectious Diseases, Medical Faculty, University of Bern, Bern, Switzerland
| | - Horst Posthaus
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
41
|
Bischofberger M, Iacovache I, van der Goot FG. Pathogenic pore-forming proteins: function and host response. Cell Host Microbe 2013; 12:266-75. [PMID: 22980324 DOI: 10.1016/j.chom.2012.08.005] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Organisms from all kingdoms produce pore-forming proteins, with the best-characterized being of bacterial origin. The last decade of research has revealed that the channels formed by these proteins can be very diverse, thus differentially affecting target cell-membrane permeability and consequent cellular outcome. The responses to these toxins are also extremely diverse due to multiple downstream effects of pore-induced changes in ion balance. Determining the secondary effects of pore-forming toxins is essential to understand their contribution to infection.
Collapse
Affiliation(s)
- Mirko Bischofberger
- Ecole Polytechnique Fédérale de Lausanne, Global Health Institute, Station 15, CH-1015 Lausanne, Switzerland
| | | | | |
Collapse
|
42
|
Fennessey CM, Ivie SE, McClain MS. Coenzyme depletion by members of the aerolysin family of pore-forming toxins leads to diminished ATP levels and cell death. MOLECULAR BIOSYSTEMS 2012; 8:2097-105. [PMID: 22688384 PMCID: PMC3759351 DOI: 10.1039/c2mb25142f] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent studies demonstrated that a variety of bacterial pore-forming toxins induce cell death through a process of programmed necrosis characterized by the rapid depletion of cellular ATP. However, events leading to the necrosis and depletion of ATP are not thoroughly understood. We demonstrate that ATP-depletion induced by two pore-forming toxins, the Clostridium perfringens epsilon-toxin and the Aeromonas hydrophila aerolysin toxin, is associated with decreased mitochondrial membrane potential and opening of the mitochondrial permeability transition pore. To gain further insight into the toxin-induced metabolic changes contributing to necrosis and depletion of ATP, we analyzed the biochemical profiles of 251 distinct compounds by GC/MS or LC/MS/MS following exposure of a human kidney cell line to the epsilon-toxin. As expected, numerous biochemicals were seen to increase or decrease in response to epsilon-toxin. However, the pattern of these changes was consistent with the toxin-induced disruption of major energy-producing pathways in the cell including disruptions to the beta-oxidation of lipids. In particular, treatment with epsilon-toxin led to decreased levels of key coenzymes required for energy production including carnitine, NAD (and NADH), and coenzyme A. Independent biochemical assays confirmed that epsilon-toxin and aerolysin induced the rapid decrease of these coenzymes or their synthetic precursors. Incubation of cells with NADH or carnitine-enriched medium helped protect cells from toxin-induced ATP depletion and cell death. Collectively, these results demonstrate that members of the aerolysin family of pore-forming toxins lead to decreased levels of essential coenzymes required for energy production. The resulting loss of energy substrates is expected to contribute to dissipation of the mitochondrial membrane potential, opening of the mitochondrial permeability transition pore, and ultimately cell death.
Collapse
Affiliation(s)
- Christine M. Fennessey
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Susan E. Ivie
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Mark S. McClain
- Division of Infectious Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| |
Collapse
|
43
|
Staphylococcus haemolyticus strains target mitochondria and induce caspase-dependent apoptosis of macrophages. Antonie van Leeuwenhoek 2012; 102:611-20. [PMID: 22660952 PMCID: PMC3474905 DOI: 10.1007/s10482-012-9756-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 05/22/2012] [Indexed: 11/30/2022]
Abstract
The aim of this study was to investigate the interaction of Staphylococcus haemolyticus strains with a macrophage cell line. Infection with the strains resulted in macrophage injury. All strains exhibited cytotoxic effects towards J774 cells. Moreover, the bacteria triggered apoptosis of the cells. The lowest apoptotic index did not exceed 21 %, whereas the highest reached 70 % at 24 h and 85 % at 48 h after infection. Incubation with the bacteria caused loss of mitochondrial membrane potential (ΔΨm) in macrophages. The pro-apoptotic activity of the strains was blocked by a pan-caspase inhibitor z-VAD-fmk, indicating the involvement of caspases in the bacteria-mediated cell death. We observed that the induction of macrophage apoptosis could constitute an important mechanism of pathogenesis by which S. haemolyticus strains evade host immune defences and cause disease.
Collapse
|
44
|
Fraunholz M, Sinha B. Intracellular Staphylococcus aureus: live-in and let die. Front Cell Infect Microbiol 2012; 2:43. [PMID: 22919634 PMCID: PMC3417557 DOI: 10.3389/fcimb.2012.00043] [Citation(s) in RCA: 277] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 03/15/2012] [Indexed: 12/30/2022] Open
Abstract
Staphylococcus aureus uses a plethora of virulence factors to accommodate a diversity of niches in its human host. Aside from the classical manifestations of S. aureus-induced diseases, the pathogen also invades and survives within mammalian host cells.The survival strategies of the pathogen are as diverse as strains or host cell types used. S. aureus is able to replicate in the phagosome or freely in the cytoplasm of its host cells. It escapes the phagosome of professional and non-professional phagocytes, subverts autophagy, induces cell death mechanisms such as apoptosis and pyronecrosis, and even can induce anti-apoptotic programs in phagocytes. The focus of this review is to present a guide to recent research outlining the variety of intracellular fates of S. aureus.
Collapse
Affiliation(s)
- Martin Fraunholz
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany.
| | | |
Collapse
|
45
|
Pathophysiological changes to the peritoneal membrane during PD-related peritonitis: the role of mesothelial cells. Mediators Inflamm 2012; 2012:484167. [PMID: 22577250 PMCID: PMC3337720 DOI: 10.1155/2012/484167] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 01/18/2012] [Accepted: 01/18/2012] [Indexed: 01/08/2023] Open
Abstract
The success of peritoneal dialysis (PD) is dependent on the structural and functional integrity of the
peritoneal membrane. The mesothelium lines the peritoneal membrane and is the first line of
defense against chemical and/or bacterial insult. Peritonitis remains a major complication of PD and
is a predominant cause of technique failure, morbidity and mortality amongst PD patients. With
appropriate antibiotic treatment, peritonitis resolves without further complications, but in some PD
patients excessive peritoneal inflammatory responses lead to mesothelial cell exfoliation and
thickening of the submesothelium, resulting in peritoneal fibrosis and sclerosis. The detrimental
changes in the peritoneal membrane structure and function correlate with the number and severity
of peritonitis episodes and the need for catheter removal. There is evidence that despite clinical
resolution of peritonitis, increased levels of inflammatory and fibrotic mediators may persist in the
peritoneal cavity, signifying persistent injury to the mesothelial cells. This review will describe the
structural and functional changes that occur in the peritoneal membrane during peritonitis and how
mesothelial cells contribute to these changes and respond to infection. The latter part of the review
discusses the potential of mesothelial cell transplantation and genetic manipulation in the
preservation of the peritoneal membrane.
Collapse
|
46
|
Staphylococcus aureus induces eosinophil cell death mediated by α-hemolysin. PLoS One 2012; 7:e31506. [PMID: 22355374 PMCID: PMC3280314 DOI: 10.1371/journal.pone.0031506] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Accepted: 01/12/2012] [Indexed: 01/21/2023] Open
Abstract
Staphylococcus aureus, a major human pathogen, exacerbates allergic disorders, including atopic dermatitis, nasal polyps and asthma, which are characterized by tissue eosinophilia. Eosinophils, via their destructive granule contents, can cause significant tissue damage, resulting in inflammation and further recruitment of inflammatory cells. We hypothesised that the relationship between S. aureus and eosinophils may contribute to disease pathology. We found that supernatants from S. aureus (SH1000 strain) cultures cause rapid and profound eosinophil necrosis, resulting in dramatic cell loss within 2 hours. This is in marked contrast to neutrophil granulocytes where no significant cell death was observed (at equivalent dilutions). Supernatants prepared from a strain deficient in the accessory gene regulator (agr) that produces reduced levels of many important virulence factors, including the abundantly produced α-hemolysin (Hla), failed to induce eosinophil death. The role of Hla in mediating eosinophil death was investigated using both an Hla deficient SH1000-modified strain, which did not induce eosinophil death, and purified Hla, which induced concentration-dependent eosinophil death via both apoptosis and necrosis. We conclude that S. aureus Hla induces aberrant eosinophil cell death in vitro and that this may increase tissue injury in allergic disease.
Collapse
|
47
|
Skals M, Leipziger J, Praetorius HA. Haemolysis induced by α-toxin from Staphylococcus aureus requires P2X receptor activation. Pflugers Arch 2011; 462:669-79. [PMID: 21847558 DOI: 10.1007/s00424-011-1010-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 07/27/2011] [Accepted: 08/01/2011] [Indexed: 11/26/2022]
Abstract
Recently, it was documented that α-haemolysin (HlyA) from Escherichia coli uses erythrocyte P2 receptors cause lysis. This finding was surprising as it appeared firmly established that HlyA-dependent pore formation per se is sufficient for full cell lysis. We discovered that HlyA induced a sequential process of shrinkage and swelling and that the final haemolysis is completely prevented by blockers of P2X receptors and pannexin channels. This finding has potential clinical relevance as it may offer specific pharmacological interference to ameliorate haemolysis inflicted by pore-forming bacterial toxins. In this context, it is essential to know whether this is specific to HlyA-induced cell damage or if other bacterial pore-forming toxins involve purinergic signals to orchestrate haemolysis. Here, we investigate if the haemolysis produced by α-toxin from Staphylococcus aureus involves P2 receptor activation. We observed that α-toxin-induced haemolysis is completely blocked by the unselective P2 receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid. Moreover, several selective blockers of P2X(1) and P2X(7) ionotropic receptors abolished haemolysis in murine and equine erythrocytes. Inhibitors of pannexin channels partially reduced the α-toxin induced lysis. Thus, we conclude that α-toxin, similar to HlyA from E. coli produces cell damage by specific activation of a purinergic signalling cascade. These data indicate that pore-forming toxins in general require purinergic signalling to elicit their toxicity.
Collapse
Affiliation(s)
- Marianne Skals
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, build. 1160, 8000, Aarhus C, Denmark
| | | | | |
Collapse
|
48
|
Medina E, Goldmann O. In vivo and ex vivo protocols for measuring the killing of extracellular pathogens by macrophages. ACTA ACUST UNITED AC 2011; Chapter 14:Unit 14.19.1-17. [PMID: 21400679 DOI: 10.1002/0471142735.im1419s92] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This unit describes a series of in vivo/ex vivo combined protocols for investigating the interactions (adhesion, phagocytosis, and killing) of extracellular bacteria with peritoneal murine macrophages. It includes steps needed for in vivo infection of murine peritoneal macrophages after intraperitoneal inoculation with the pathogen of interest, as well as the measurement of bacteria associated with or truly internalized by these phagocytic cells. Several protocols for the ex vivo measurement of the ability of peritoneal macrophages to kill the microorganisms that have been ingested during the in vivo infection assay are included.
Collapse
Affiliation(s)
- Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | |
Collapse
|
49
|
Helicobacter pylori VacA induces programmed necrosis in gastric epithelial cells. Infect Immun 2011; 79:2535-43. [PMID: 21482684 DOI: 10.1128/iai.01370-10] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori is a Gram-negative bacterium that colonizes the human stomach and contributes to the development of peptic ulcer disease and gastric cancer. The secreted pore-forming toxin VacA is one of the major virulence factors of H. pylori. In the current study, we show that AZ-521 human gastric epithelial cells are highly susceptible to VacA-induced cell death. Wild-type VacA causes death of these cells, whereas mutant VacA proteins defective in membrane channel formation do not. Incubation of AZ-521 cells with wild-type VacA results in cell swelling, poly(ADP-ribose) polymerase (PARP) activation, decreased intracellular ATP concentration, and lactate dehydrogenase (LDH) release. VacA-induced death of these cells is a caspase-independent process that results in cellular release of histone-binding protein high mobility group box 1 (HMGB1), a proinflammatory protein. These features are consistent with the occurrence of cell death through a programmed necrosis pathway and suggest that VacA can be included among the growing number of bacterial pore-forming toxins that induce cell death through programmed necrosis. We propose that VacA augments H. pylori-induced mucosal inflammation in the human stomach by causing programmed necrosis of gastric epithelial cells and subsequent release of proinflammatory proteins and may thereby contribute to the pathogenesis of gastric cancer and peptic ulceration.
Collapse
|
50
|
Liang X, Hall JW, Yang J, Yan M, Doll K, Bey R, Ji Y. Identification of single nucleotide polymorphisms associated with hyperproduction of alpha-toxin in Staphylococcus aureus. PLoS One 2011; 6:e18428. [PMID: 21494631 PMCID: PMC3072997 DOI: 10.1371/journal.pone.0018428] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 03/06/2011] [Indexed: 12/02/2022] Open
Abstract
The virulence factor α-toxin (hla) is needed by Staphylococcus aureus in order to cause infections in both animals and humans. Although the complicated regulation of hla expression has been well studied in human S. aureus isolates, the mechanisms of of hla regulation in bovine S. aureus isolates remain undefined. In this study, we found that many bovine S. aureus isolates, including the RF122 strain, generate dramatic amounts of α-toxin in vitro compared with human clinical S. aureus isolates, including MRSA WCUH29 and MRSA USA300. To elucidate potential regulatory mechanisms, we analyzed the hla promoter regions and identified predominant single nucleotide polymorphisms (SNPs) at positions −376, −483, and −484 from the start codon in α-toxin hyper-producing isolates. Using site-directed mutagenesis and hla promoter-gfp-luxABCDE dual reporter approaches, we demonstrated that the SNPs contribute to the differential control of hla expression among bovine and human S. aureus isolates. Using a DNA affinity assay, gel-shift assays and a null mutant, we identified and revealed that an hla positive regulator, SarZ, contributes to the involvement of the SNPs in mediating hla expression. In addition, we found that the bovine S. aureus isolate RF122 exhibits higher transcription levels of hla positive regulators, including agrA, saeR, arlR and sarZ, but a lower expression level of hla repressor rot compared to the human S. aureus isolate WCUH29. Our results indicate α-toxin hyperproduction in bovine S. aureus is a multifactorial process, influenced at both the genomic and transcriptional levels. Moreover, the identification of predominant SNPs in the hla promoter region may provide a novel method for genotyping the S. aureus isolates.
Collapse
Affiliation(s)
- Xudong Liang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Jeffrey W. Hall
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Junshu Yang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Meiying Yan
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Katherine Doll
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Russell Bey
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Yinduo Ji
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
- * E-mail:
| |
Collapse
|