1
|
Liang W, Xiao H, Chen JY, Chang YF, Cao SJ, Wen YP, Wu R, Du SY, Yan QG, Huang XB, Zhao Q. Immunogenicity and protective efficacy of a multi-epitope recombinant toxin antigen of Pasteurella multocida against virulent challenge in mice. Vaccine 2023; 41:2387-2396. [PMID: 36872144 DOI: 10.1016/j.vaccine.2023.02.070] [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: 10/26/2022] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
Pasteurella multocida (P. multocida) infection frequently results in porcine atrophic rhinitis and swine plague, leading to large economic losses for the swine industry worldwide. P. multocida toxin (PMT, 146 kDa) is a highly virulent key virulence factor that plays a vital role in causing lung and turbinate lesions. This study developed a multi-epitope recombinant antigen of PMT (rPMT) that showed excellent immunogenicity and protection in a mouse model. Using bioinformatics to analyse the dominant epitopes of PMT, we constructed and synthesized rPMT containing 10 B-cell epitopes, 8 peptides with multiple B-cell epitopes and 13 T-cell epitopes of PMT and a rpmt gene (1,974 bp) with multiple epitopes. The rPMT protein (97 kDa) was soluble and contained a GST tag protein. Immunization of mice with rPMT stimulated significantly elevated serum IgG titres and splenocyte proliferation, and serum IFN-γ and IL-12 were upregulated by 5-fold and 1.6-fold, respectively, but IL-4 was not. Furthermore, the rPMT immunization group exhibited alleviated lung tissue lesions and a significantly decreased degree of neutrophil infiltration compared with the control groups post-challenge. In the rPMT vaccination group, 57.1% (8/14) of the mice survived the challenge, similar to the bacterin HN06 group, while all the mice in the control groups succumbed to the challenge. Thus, rPMT could be a suitable candidate antigen for developing a subunit vaccine against toxigenic P. multocida infection.
Collapse
Affiliation(s)
- Wei Liang
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Hang Xiao
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Jia-Yong Chen
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - San-Jie Cao
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi-Ping Wen
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Rui Wu
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Sen-Yan Du
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Qi-Gui Yan
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Bo Huang
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Qin Zhao
- Research Center of Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China; National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
2
|
Kubatzky KF. Pasteurella multocida toxin - lessons learned from a mitogenic toxin. Front Immunol 2022; 13:1058905. [PMID: 36591313 PMCID: PMC9800868 DOI: 10.3389/fimmu.2022.1058905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
The gram-negative, zoonotic bacterium Pasteurella multocida was discovered in 1880 and found to be the causative pathogen of fowl cholera. Pasteurella-related diseases can be found in domestic and wild life animals such as buffalo, sheep, goat, deer and antelope, cats, dogs and tigers and cause hemorrhagic septicemia in cattle, rhinitis or pneumonia in rabbits or fowl cholera in poultry and birds. Pasteurella multocida does not play a major role in the immune-competent human host, but can be found after animal bites or in people with close contact to animals. Toxigenic strains are most commonly found in pigs and express a phage-encoded 146 kDa protein, the Pasteurella multocida toxin (PMT). Toxin-expressing strains cause atrophic rhinitis where nasal turbinate bones are destroyed through the inhibition of bone building osteoblasts and the activation of bone resorbing osteoclasts. After its uptake through receptor-mediated endocytosis, PMT specifically targets the alpha subunit of several heterotrimeric G proteins and constitutively activates them through deamidation of a glutamine residue to glutamate in the alpha subunit. This results in cytoskeletal rearrangement, proliferation, differentiation and survival of cells. Because of the toxin's mitogenic effects, it was suggested that it might have carcinogenic properties, however, no link between Pasteurella infections and cell transformation could be established, neither in tissue culture models nor through epidemiological data. In the recent years it was shown that the toxin not only affects bone, but also the heart as well as basically all cells of innate and adaptive immunity. During the last decade the focus of research shifted from signal transduction processes to understanding how the bacteria might benefit from a bone-destroying toxin. The primary function of PMT seems to be the modulation of immune cell activation which at the same time creates an environment permissive for osteoclast formation. While the disease is restricted to pigs, the implications of the findings from PMT research can be used to explore human diseases and have a high translational potential. In this review our current knowledge will be summarized and it will be discussed what can be learned from using PMT as a tool to understand human pathologies.
Collapse
Affiliation(s)
- Katharina F. Kubatzky
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
3
|
Pepanian A, Sommerfeld P, Kasprzyk R, Kühl T, Binbay FA, Hauser C, Löser R, Wodtke R, Bednarczyk M, Chrominski M, Kowalska J, Jemielity J, Imhof D, Pietsch M. Fluorescence Anisotropy Assay with Guanine Nucleotides Provides Access to Functional Analysis of Gαi1 Proteins. Anal Chem 2022; 94:14410-14418. [PMID: 36206384 DOI: 10.1021/acs.analchem.2c03176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Gα proteins as part of heterotrimeric G proteins are molecular switches essential for G protein-coupled receptor- mediated intracellular signaling. The role of the Gα subunits has been examined for decades with various guanine nucleotides to elucidate the activation mechanism and Gα protein-dependent signal transduction. Several approaches describe fluorescent ligands mimicking the GTP function, yet lack the efficient estimation of the proteins' GTP binding activity and the fraction of active protein. Herein, we report the development of a reliable fluorescence anisotropy-based method to determine the affinity of ligands at the GTP-binding site and to quantify the fraction of active Gαi1 protein. An advanced bacterial expression protocol was applied to produce active human Gαi1 protein, whose GTP binding capability was determined with novel fluorescently labeled guanine nucleotides acting as high-affinity Gαi1 binders compared to the commonly used BODIPY FL GTPγS. This study thus contributes a new method for future investigations of the characterization of Gαi and other Gα protein subunits, exploring their corresponding signal transduction systems and potential for biomedical applications.
Collapse
Affiliation(s)
- Anna Pepanian
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, 53121 Bonn, Germany
| | - Paul Sommerfeld
- Institutes I & II of Pharmacology, Center of Pharmacology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Renata Kasprzyk
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Toni Kühl
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, 53121 Bonn, Germany
| | - F Ayberk Binbay
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, 53121 Bonn, Germany
| | - Christoph Hauser
- Institutes I & II of Pharmacology, Center of Pharmacology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Reik Löser
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Robert Wodtke
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Marcelina Bednarczyk
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland.,Division of Biophysics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| | | | - Joanna Kowalska
- Division of Biophysics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| | - Jacek Jemielity
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, 53121 Bonn, Germany
| | - Markus Pietsch
- Institutes I & II of Pharmacology, Center of Pharmacology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
4
|
Pasteurella multocida Toxin Aggravates Ligatured-Induced Periodontal Bone Loss and Inflammation via NOD-Like Receptor Protein 3 Inflammasome. Cell Microbiol 2022. [DOI: 10.1155/2022/3305695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is reportedly involved in periodontal pathogenesis. Pasteurella multocida toxin (PMT) is the major virulence factor of Pasteurella multocida strains, which belongs to the nonoral gram-negative facultative rods (GNFR). The existence of GNFR and their toxin may aggravate periodontitis. Therefore, it is important to unclose the regulatory mechanisms of PMT in periodontitis. However, the involvement of NLRP3 inflammasome and PMT in periodontitis remain unclear. The results showed that NLRP3 expression was increased in periodontitis mice by immunohistochemical staining and quantitative reverse transcription polymerase chain reaction (qRT-PCR). Nlrp3-/- mice showed less periodontal bone loss and lower abundances of Pasteurella multocida by 16S rRNA sequencing. PMT promoted NLRP3 expressions by activating nuclear factor kappa light chain enhancer of B cells (NF-κB) pathway and activated NLRP3 inflammasome. This effect was reversed by NLRP3 inhibitor MCC950. Furthermore, PMT aggravated periodontal bone loss and inflammation in WT mice, while MCC950 attenuated periodontal bone loss and inflammation. The Nlrp3-/- periodontitis models with PMT local injection showed less bone loss and inflammation compared with WT periodontitis mice after PMT treatment. Taken together, our results showed that PMT aggravates periodontal response to the ligature by promoting NLRP3 expression and activating NLRP3 inflammasome, suggesting that NLRP3 may be an effective target for the treatment of periodontitis caused by GNFR and MCC950 may be a potential drug against this disease.
Collapse
|
5
|
Keen AC, Pedersen MH, Lemel L, Scott DJ, Canals M, Littler DR, Beddoe T, Ono Y, Shi L, Inoue A, Javitch JA, Lane JR. OZITX, a pertussis toxin-like protein for occluding inhibitory G protein signalling including Gα z. Commun Biol 2022; 5:256. [PMID: 35322196 PMCID: PMC8943041 DOI: 10.1038/s42003-022-03191-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/17/2022] [Indexed: 11/18/2022] Open
Abstract
Heterotrimeric G proteins are the main signalling effectors for G protein-coupled receptors. Understanding the distinct functions of different G proteins is key to understanding how their signalling modulates physiological responses. Pertussis toxin, a bacterial AB5 toxin, inhibits Gαi/o G proteins and has proven useful for interrogating inhibitory G protein signalling. Pertussis toxin, however, does not inhibit one member of the inhibitory G protein family, Gαz. The role of Gαz signalling has been neglected largely due to a lack of inhibitors. Recently, the identification of another Pertussis-like AB5 toxin was described. Here we show that this toxin, that we call OZITX, specifically inhibits Gαi/o and Gαz G proteins and that expression of the catalytic S1 subunit is sufficient for this inhibition. We identify mutations that render Gα subunits insensitive to the toxin that, in combination with the toxin, can be used to interrogate the signalling of each inhibitory Gα G protein.
Collapse
Affiliation(s)
- Alastair C Keen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham, UK
| | - Maria Hauge Pedersen
- Departments of Psychiatry and Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
- NNF Center for Basic Metabolic Research, Section for Metabolic Receptology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Laura Lemel
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham, UK
| | - Daniel J Scott
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3052, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, 3052, Australia
| | - Meritxell Canals
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham, UK
| | - Dene R Littler
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3052, Australia
| | - Travis Beddoe
- Department of Animal, Plant and Soil Science and Centre for AgriBioscience, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Yuki Ono
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Jonathan A Javitch
- Departments of Psychiatry and Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA.
| | - J Robert Lane
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK.
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham, UK.
| |
Collapse
|
6
|
Nubbemeyer B, Pepanian A, Paul George AA, Imhof D. Strategies towards Targeting Gαi/s Proteins: Scanning of Protein-Protein Interaction Sites To Overcome Inaccessibility. ChemMedChem 2021; 16:1696-1715. [PMID: 33615736 PMCID: PMC8252600 DOI: 10.1002/cmdc.202100039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Indexed: 12/16/2022]
Abstract
Heterotrimeric G proteins are classified into four subfamilies and play a key role in signal transduction. They transmit extracellular signals to intracellular effectors subsequent to the activation of G protein-coupled receptors (GPCRs), which are targeted by over 30 % of FDA-approved drugs. However, addressing G proteins as drug targets represents a compelling alternative, for example, when G proteins act independently of the corresponding GPCRs, or in cases of complex multifunctional diseases, when a large number of different GPCRs are involved. In contrast to Gαq, efforts to target Gαi/s by suitable chemical compounds has not been successful so far. Here, a comprehensive analysis was conducted examining the most important interface regions of Gαi/s with its upstream and downstream interaction partners. By assigning the existing compounds and the performed approaches to the respective interfaces, the druggability of the individual interfaces was ranked to provide perspectives for selective targeting of Gαi/s in the future.
Collapse
Affiliation(s)
- Britta Nubbemeyer
- Pharmaceutical Biochemistry and BioanalyticsPharmaceutical InstituteUniversity of BonnAn der Immenburg 453121BonnGermany
| | - Anna Pepanian
- Pharmaceutical Biochemistry and BioanalyticsPharmaceutical InstituteUniversity of BonnAn der Immenburg 453121BonnGermany
| | | | - Diana Imhof
- Pharmaceutical Biochemistry and BioanalyticsPharmaceutical InstituteUniversity of BonnAn der Immenburg 453121BonnGermany
| |
Collapse
|
7
|
Banu A, Lax AJ, Grigoriadis AE. In Vivo Targets of Pasteurella Multocida Toxin. Int J Mol Sci 2020; 21:ijms21082739. [PMID: 32326543 PMCID: PMC7215291 DOI: 10.3390/ijms21082739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 01/03/2023] Open
Abstract
Many Pasteurella multocida strains are carried as commensals, while some cause disease in animals and humans. Some type D strains cause atrophic rhinitis in pigs, where the causative agent is known to be the Pasteurella multocida toxin (PMT). PMT activates three families of G-proteins—Gq/11, G12/13, and Gi/o—leading to cellular mitogenesis and other sequelae. The effects of PMT on whole animals in vivo have been investigated previously, but only at the level of organ-specific pathogenesis. We report here the first study to screen all the organs targeted by the toxin by using the QE antibody that recognizes only PMT-modified G-proteins. Under our experimental conditions, short-term treatment of PMT is shown to have multiple in vivo targets, demonstrating G-alpha protein modification, stimulation of proliferation markers and expression of active β-catenin in a tissue- and cell-specific manner. This highlights the usefulness of PMT as an important tool for dissecting the specific roles of different G-alpha proteins in vivo.
Collapse
Affiliation(s)
- Arshiya Banu
- Department of Microbiology, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Centre for Craniofacial and Regenerative Biology, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Alistair J. Lax
- Department of Microbiology, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Agamemnon E. Grigoriadis
- Centre for Craniofacial and Regenerative Biology, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Correspondence: ; Tel.: +44-(0)20-7188-1807
| |
Collapse
|
8
|
Li J, Ge Y, Huang JX, Strømgaard K, Zhang X, Xiong XF. Heterotrimeric G Proteins as Therapeutic Targets in Drug Discovery. J Med Chem 2019; 63:5013-5030. [PMID: 31841625 DOI: 10.1021/acs.jmedchem.9b01452] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heterotrimeric G proteins are molecular switches in GPCR signaling pathways and regulate a plethora of physiological and pathological processes. GPCRs are efficient drug targets, and more than 30% of the drugs in use target them. However, selectively targeting an individual GPCR may be undesirable in various multifactorial diseases in which multiple receptors are involved. In addition, abnormal activation or expression of G proteins is frequently associated with diseases. Furthermore, G proteins harboring mutations often result in malignant diseases. Thus, targeting G proteins instead of GPCRs might provide alternative approaches for combating these diseases. In this review, we discuss the biochemistry of heterotrimeric G proteins, describe the G protein-associated diseases, and summarize the currently known modulators that can regulate the activities of G proteins. The outlook for targeting G proteins to treat diverse diseases is also included in this manuscript.
Collapse
Affiliation(s)
- Jian Li
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Yang Ge
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Jun-Xiang Huang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Xiaolei Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Xiao-Feng Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| |
Collapse
|
9
|
Selheim F, Aasebø E, Ribas C, Aragay AM. An Overview on G Protein-coupled Receptor-induced Signal Transduction in Acute Myeloid Leukemia. Curr Med Chem 2019; 26:5293-5316. [PMID: 31032748 DOI: 10.2174/0929867326666190429153247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 03/22/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Acute Myeloid Leukemia (AML) is a genetically heterogeneous disease characterized by uncontrolled proliferation of precursor myeloid-lineage cells in the bone marrow. AML is also characterized by patients with poor long-term survival outcomes due to relapse. Many efforts have been made to understand the biological heterogeneity of AML and the challenges to develop new therapies are therefore enormous. G Protein-coupled Receptors (GPCRs) are a large attractive drug-targeted family of transmembrane proteins, and aberrant GPCR expression and GPCR-mediated signaling have been implicated in leukemogenesis of AML. This review aims to identify the molecular players of GPCR signaling, focusing on the hematopoietic system, which are involved in AML to help developing novel drug targets and therapeutic strategies. METHODS We undertook an exhaustive and structured search of bibliographic databases for research focusing on GPCR, GPCR signaling and expression in AML. RESULTS AND CONCLUSION Many scientific reports were found with compelling evidence for the involvement of aberrant GPCR expression and perturbed GPCR-mediated signaling in the development of AML. The comprehensive analysis of GPCR in AML provides potential clinical biomarkers for prognostication, disease monitoring and therapeutic guidance. It will also help to provide marker panels for monitoring in AML. We conclude that GPCR-mediated signaling is contributing to leukemogenesis of AML, and postulate that mass spectrometrybased protein profiling of primary AML cells will accelerate the discovery of potential GPCR related biomarkers for AML.
Collapse
Affiliation(s)
- Frode Selheim
- The Proteomics Unit at the University of Bergen, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Elise Aasebø
- The Proteomics Unit at the University of Bergen, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.,Department of Clinical Science, University of Bergen, Jonas Lies vei 87, 5021 Bergen, Norway
| | - Catalina Ribas
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029 Madrid, Spain
| | - Anna M Aragay
- Departamento de Biologia Celular. Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Spanish National Research Council (CSIC), Baldiri i Reixac, 15, 08028 Barcelona, Spain
| |
Collapse
|
10
|
Ebner JK, König GM, Kostenis E, Siegert P, Aktories K, Orth JHC. Activation of G q signaling by Pasteurella multocida toxin inhibits the osteoblastogenic-like actions of Activin A in C2C12 myoblasts, a cell model of fibrodysplasia ossificans progressiva. Bone 2019; 127:592-601. [PMID: 31376533 DOI: 10.1016/j.bone.2019.07.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 01/15/2023]
Abstract
The human disease fibrodysplasia ossificans progressiva (FOP) is a rare and highly disabling disorder of extensive heterotopic bone growth that is caused by a point mutation (R206H) in the activation domain of Alk2, a BMP (bone morphogenic protein) type 1 receptor. The mutation leads to extensive BMP-signaling induced by Activin A, which is normally an antagonist for wildtype receptors, resulting in excessive and uncontrolled bone formation. Here, we studied the effects of Pasteurella multocida toxin (PMT), which activates osteoclasts and inhibits osteoblast activity, in C2C12 myoblasts expressing the mutant Alk2(R206H) receptor as model of FOP. In our study, we mainly used alkaline phosphatase (ALP) activity as marker to determine osteoblast differentiation. BMP-4 stimulated an increase in ALP activity in C2C12-Alk2wt and C2C12-Alk2(R206H) cells. By contrast, Activin A only induced ALP activity in C2C12-Alk2(R206H) cells. In both cases, PMT acted as a potent inhibitor of ALP activity. PMT-induced inhibition of ALP activity was paralleled by a constitutive activation of the heterotrimeric Gq protein. Expression of a permanently active Gαq blocked Activin A/Alk2(R206H)-dependent increase in ALP activity. Inactivation of Gq by specific inhibitor FR900359 blocked the PMT effect. Similarly, canonical second messengers and effectors of Gαq (e.g. ionophore A23187-induced increase in intracellular Ca2+ and activation of PKC by PMA (phorbol 12-myristate 13-acetate)) inhibited Alk2(R206H)-mediated induction of ALP activity. Notably, Activin A-induced increase in ALP activity in C2C12-Alk2(R206H) cells was also inhibited by stimulation of the α1A-adrenoceptor, which couples to Gαq, by phenylephrine. PMT did not alter tail phosphorylation of the major downstream effectors of the Alk2 receptor, Smad1/5/9; neither did the toxin affect nuclear translocation of the Smad-complex. However, PMT diminished BMP responsive element-induced gene expression. The data indicate that PMT potently inhibits the induction of osteoblast markers in a FOP model via activation of G proteins. Moreover, our findings indicate that activation of G protein-coupled receptors and of G protein signaling might be a rationale for pharmacological therapy of FOP.
Collapse
Affiliation(s)
- Julia K Ebner
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, Schänzlestr. 1, 79104 Freiburg, Germany; Spemann Graduate School for Biology and Medicine, University of Freiburg, Albertstr. 19A, 79104 Freiburg, Germany
| | - Gabriele M König
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Peter Siegert
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| | - Klaus Aktories
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany; Spemann Graduate School for Biology and Medicine, University of Freiburg, Albertstr. 19A, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany.
| | - Joachim H C Orth
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| |
Collapse
|
11
|
Banu A, Liu KJ, Lax AJ, Grigoriadis AE. G-Alpha Subunit Abundance and Activity Differentially Regulate β-Catenin Signaling. Mol Cell Biol 2019; 39:MCB.00422-18. [PMID: 30559307 PMCID: PMC6379582 DOI: 10.1128/mcb.00422-18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/27/2018] [Indexed: 11/29/2022] Open
Abstract
Heterotrimeric G proteins are signal transduction proteins involved in regulating numerous signaling events. In particular, previous studies have demonstrated a role for G-proteins in regulating β-catenin signaling. However, the link between G-proteins and β-catenin signaling is controversial and appears to depend on G-protein specificity. We describe a detailed analysis of a link between specific G-alpha subunits and β-catenin using G-alpha subunit genetic knockout and knockdown approaches. The Pasteurella multocida toxin was utilized as a unique tool to activate G-proteins, with LiCl treatment serving as a β-catenin signaling agonist. The results show that Pasteurella multocida toxin (PMT) significantly enhanced LiCl-induced active β-catenin levels in HEK293T cells and mouse embryo fibroblasts. Evaluation of the effect of specific G-alpha proteins on the regulation of β-catenin showed that Gq/11 and G12/13 knockout cells had significantly higher levels of active and total β-catenin than wild-type cells. The stimulation of active β-catenin by PMT and LiCl was lost upon both constitutive and transient knockdown of G12 and G13 but not Gq Based on our results, we conclude that endogenous G-alpha proteins are negative regulators of active β-catenin; however, PMT-activated G-alpha subunits positively regulate LiCl-induced β-catenin expression in a G12/13-dependent manner. Hence, G-alpha subunit regulation of β-catenin is context dependent.
Collapse
Affiliation(s)
- Arshiya Banu
- Department of Microbiology, King's College London, Guy's Hospital, London, United Kingdom
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, United Kingdom
| | - Alistair J Lax
- Department of Microbiology, King's College London, Guy's Hospital, London, United Kingdom
| | - Agamemnon E Grigoriadis
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
12
|
Salter SJ, Scott P, Page AJ, Tracey A, de Goffau MC, Cormie C, Ochoa-Montaño B, Ling CL, Tangmanakit J, Turner P, Parkhill J. 'Candidatus Ornithobacterium hominis': insights gained from draft genomes obtained from nasopharyngeal swabs. Microb Genom 2019; 5. [PMID: 30720420 PMCID: PMC6421346 DOI: 10.1099/mgen.0.000247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
'Candidatus Ornithobacterium hominis' represents a new member of the Flavobacteriaceae detected in 16S rRNA gene surveys of people from South-East Asia, Africa and Australia. It frequently colonizes the infant nasopharynx at high proportional abundance, and we demonstrate its presence in 42 % of nasopharyngeal swabs from 12-month-old children in the Maela refugee camp in Thailand. The species, a Gram-negative bacillus, has not yet been cultured, but the cells can be identified in mixed samples by fluorescent hybridization. Here, we report seven genomes assembled from metagenomic data, two to improved draft standard. The genomes are approximately 1.9 Mb, sharing 62 % average amino acid identity with the only other member of the genus, the bird pathogen Ornithobacterium rhinotracheale. The draft genomes encode multiple antibiotic-resistance genes, competition factors, Flavobacterium johnsoniae-like gliding motility genes and a homologue of the Pasteurella multocida mitogenic toxin. Intra- and inter-host genome comparison suggests that colonization with this bacterium is both persistent and strain exclusive.
Collapse
Affiliation(s)
| | - Paul Scott
- 1Pathogen Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Andrew J Page
- 1Pathogen Genomics, Wellcome Sanger Institute, Hinxton, UK.,†Present address: Quadram Institute Bioscience, Norwich, UK
| | - Alan Tracey
- 1Pathogen Genomics, Wellcome Sanger Institute, Hinxton, UK
| | | | - Claire Cormie
- 1Pathogen Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Bernardo Ochoa-Montaño
- 2Department of Biochemistry, University of Cambridge, Cambridge, UK.,‡Present address: Illumina Cambridge Ltd, Little Chesterford, UK
| | - Clare L Ling
- 3Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand.,4Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jiraporn Tangmanakit
- 3Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Paul Turner
- 4Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,5Cambodia-Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, Cambodia
| | | |
Collapse
|
13
|
Toomey CB, Fraser K, Thorson JA, Goldbaum MH, Lin JH. GNAQ and PMS1 Mutations Associated with Uveal Melanoma, Ocular Surface Melanosis, and Nevus of Ota. Ocul Oncol Pathol 2019; 5:267-272. [PMID: 31367589 DOI: 10.1159/000495508] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/13/2018] [Indexed: 12/23/2022] Open
Abstract
G protein mutations are common in uveal melanomas, and the vast majority target amino acid residue Q209 in either GNAQ or GNA11. The GNAQ R183Q mutation is found in a small fraction of uveal melanomas. We report a patient with an unusual presentation of uveal melanoma arising at an early age in the setting of congenital skin and ocular surface melanosis. A 34-year-old Hispanic female with congenital bilateral nevus of Ota and ocular surface melanosis presented with progressive loss of visual acuity and was found to have a juxtapapillary uveal melanoma. She was treated with brachytherapy, but the tumor relapsed. She underwent enucleation that revealed mixed spindle and epithelioid uveal melanoma cells with no extraocular or lymphovascular spread. Next-generation sequencing performed on DNA isolated from the enucleation specimen identified a GNAQ R183Q mutation and a PMS1 truncation mutation. Cytogenetic profiling revealed no monosomy 3. These findings raise the possibility that uveal melanomas bearing G protein R183 mutations may have distinct clinicopathologic profiles compared to those with Q209 mutations. Furthermore, this is the first reported case of a mutation in the mismatch repair gene PMS1 associated with uveal melanoma.
Collapse
Affiliation(s)
- Christopher B Toomey
- Department of Ophthalmology, UC San Diego, La Jolla, California, USA.,VA San Diego Healthcare System, San Diego, California, USA
| | - Kyle Fraser
- Department of Pathology, UC San Diego, La Jolla, California, USA
| | - John A Thorson
- Department of Pathology, UC San Diego, La Jolla, California, USA
| | | | - Jonathan H Lin
- Department of Ophthalmology, UC San Diego, La Jolla, California, USA.,Department of Pathology, UC San Diego, La Jolla, California, USA.,VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
14
|
Biancucci M, Minasov G, Banerjee A, Herrera A, Woida PJ, Kieffer MB, Bindu L, Abreu-Blanco M, Anderson WF, Gaponenko V, Stephen AG, Holderfield M, Satchell KJF. The bacterial Ras/Rap1 site-specific endopeptidase RRSP cleaves Ras through an atypical mechanism to disrupt Ras-ERK signaling. Sci Signal 2018; 11:eaat8335. [PMID: 30279169 PMCID: PMC6309442 DOI: 10.1126/scisignal.aat8335] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Ras-extracellular signal-regulated kinase pathway is critical for controlling cell proliferation, and its aberrant activation drives the growth of various cancers. Because many pathogens produce toxins that inhibit Ras activity, efforts to develop effective Ras inhibitors to treat cancer could be informed by studies of Ras inhibition by pathogens. Vibrio vulnificus causes fatal infections in a manner that depends on multifunctional autoprocessing repeats-in-toxin, a toxin that releases bacterial effector domains into host cells. One such domain is the Ras/Rap1-specific endopeptidase (RRSP), which site-specifically cleaves the Switch I domain of the small GTPases Ras and Rap1. We solved the crystal structure of RRSP and found that its backbone shares a structural fold with the EreA/ChaN-like superfamily of enzymes. Unlike other proteases in this family, RRSP is not a metalloprotease. Through nuclear magnetic resonance analysis and nucleotide exchange assays, we determined that the processing of KRAS by RRSP did not release any fragments or cause KRAS to dissociate from its bound nucleotide but instead only locally affected its structure. However, this structural alteration of KRAS was sufficient to disable guanine nucleotide exchange factor-mediated nucleotide exchange and prevent KRAS from binding to RAF. Thus, RRSP is a bacterial effector that represents a previously unrecognized class of protease that disconnects Ras from its signaling network while inducing limited structural disturbance in its target.
Collapse
Affiliation(s)
- Marco Biancucci
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - George Minasov
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Structural Genomics of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Avik Banerjee
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Alfa Herrera
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Patrick J Woida
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Matthew B Kieffer
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lakshman Bindu
- National Cancer Institute-RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Maria Abreu-Blanco
- National Cancer Institute-RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Wayne F Anderson
- Center for Structural Genomics of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Andrew G Stephen
- National Cancer Institute-RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Matthew Holderfield
- National Cancer Institute-RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Karla J F Satchell
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
- Center for Structural Genomics of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
15
|
Brink T, Leiss V, Siegert P, Jehle D, Ebner JK, Schwan C, Shymanets A, Wiese S, Nürnberg B, Hensel M, Aktories K, Orth JHC. Salmonella Typhimurium effector SseI inhibits chemotaxis and increases host cell survival by deamidation of heterotrimeric Gi proteins. PLoS Pathog 2018; 14:e1007248. [PMID: 30102745 PMCID: PMC6107295 DOI: 10.1371/journal.ppat.1007248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/23/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022] Open
Abstract
Salmonella enterica serotype Typhimurium (S. Typhimurium) is one of the most frequent causes of food-borne illness in humans and usually associated with acute self-limiting gastroenteritis. However, in immunocompromised patients, the pathogen can disseminate and lead to severe systemic diseases. S. Typhimurium are facultative intracellular bacteria. For uptake and intracellular life, Salmonella translocate numerous effector proteins into host cells using two type-III secretion systems (T3SS), which are encoded within Salmonella pathogenicity islands 1 (SPI-1) and 2 (SPI-2). While SPI-1 effectors mainly promote initial invasion, SPI-2 effectors control intracellular survival and proliferation. Here, we elucidate the mode of action of Salmonella SPI-2 effector SseI, which is involved in control of systemic dissemination of S. Typhimurium. SseI deamidates a specific glutamine residue of heterotrimeric G proteins of the Gαi family, resulting in persistent activation of the G protein. Gi activation inhibits cAMP production and stimulates PI3-kinase γ by Gαi-released Gβγ subunits, resulting in activation of survival pathways by phosphorylation of Akt and mTOR. Moreover, SseI-induced deamidation leads to non-polarized activation of Gαi and, thereby, to loss of directed migration of dendritic cells. Salmonella Typhimurium is one of the most common causes of gastroenteritis in humans. In immunocompromised patients, the pathogen can cause systemic infections. Crucial virulence factors are encoded on two Salmonella pathogenicity islands SPI-1 and SPI-2. While SPI-1 encodes virulence factors essential for host cell invasion, intracellular proliferation of the pathogen depends mainly on SPI-2 effectors. Here, we elucidate the mode of action of Salmonella SPI-2 effector SseI. SseI activates heterotrimeric G proteins of the Gαi family by deamidation of a specific glutamine residue. Deamidation blocks GTP hydrolysis by Gαi, resulting in a persistently active G protein. Gi activation inhibits cAMP production and stimulates PI3Kγ by Gαi-released Gβγ subunits, resulting in activation of survival pathways by phosphorylation of Akt and mTOR. Moreover, deamidation of Gαi leads to a loss of directed migration in dendritic cells. The data offers a new perspective in the understanding of the actions of SseI.
Collapse
Affiliation(s)
- Thorsten Brink
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Veronika Leiss
- Abteilung für Pharmakologie und Experimentelle Therapie, Medizinische Fakultät und ICePhA, Eberhard-Karls-Universität Tübingen, Germany
| | - Peter Siegert
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Doris Jehle
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Julia K. Ebner
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- Fakultät für Biologie, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Carsten Schwan
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Aliaksei Shymanets
- Abteilung für Pharmakologie und Experimentelle Therapie, Medizinische Fakultät und ICePhA, Eberhard-Karls-Universität Tübingen, Germany
| | - Sebastian Wiese
- Zentrum für Biosystemanalyse, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Bernd Nürnberg
- Abteilung für Pharmakologie und Experimentelle Therapie, Medizinische Fakultät und ICePhA, Eberhard-Karls-Universität Tübingen, Germany
| | - Michael Hensel
- Abteilung Mikrobiologie, Fachbereich Biologie/Chemie, Universität Osnabrück, Osnabrück, Germany
| | - Klaus Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- * E-mail:
| | - Joachim H. C. Orth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| |
Collapse
|
16
|
Heni H, Ebner JK, Schmidt G, Aktories K, Orth JHC. Involvement of Osteocytes in the Action of Pasteurella multocida Toxin. Toxins (Basel) 2018; 10:toxins10080328. [PMID: 30104531 PMCID: PMC6115833 DOI: 10.3390/toxins10080328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 01/24/2023] Open
Abstract
Pasteurella multocida toxin (PMT) causes progressive atrophic rhinitis with severe turbinate bone degradation in pigs. It has been reported that the toxin deamidates and activates heterotrimeric G proteins, resulting in increased differentiation of osteoclasts and blockade of osteoblast differentiation. So far, the action of PMT on osteocytes, which is the most abundant cell type in bone tissue, is not known. In MLO-Y4 osteocytes, PMT deamidated heterotrimeric G proteins, resulting in loss of osteocyte dendritic processes, stress fiber formation, cell spreading and activation of RhoC but not of RhoA. Moreover, the toxin caused processing of membrane-bound receptor activator of NF-κB ligand (RANKL) to release soluble RANKL and enhanced the secretion of osteoclastogenic TNF-α. In a co-culture model of osteocytes and bone marrow cells, PMT-induced osteoclastogenesis was largely increased as compared to the mono-culture model. The enhancement of osteoclastogenesis observed in the co-culture was blocked by sequestering RANKL with osteoprotegerin and by an antibody against TNF-α indicating involvement of release of the osteoclastogenic factors from osteocytes. Data support the crucial role of osteocytes in bone metabolism and osteoclastogenesis and identify osteocytes as important target cells of PMT in progressive atrophic rhinitis.
Collapse
Affiliation(s)
- Hannah Heni
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany.
- Hermann-Staudinger-Graduiertenschule, Universität Freiburg, 79104 Freiburg, Germany.
| | - Julia K Ebner
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany.
- Spemann Graduate School of Biology and Medicine (SGBM), Universität Freiburg, 79104 Freiburg, Germany.
- Faculty of Biology, Universität Freiburg, 79104 Freiburg, Germany.
| | - Gudula Schmidt
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany.
| | - Klaus Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany.
- BIOSS Centre for Biological Signalling Studies, Universität Freiburg, 79104 Freiburg, Germany.
| | - Joachim H C Orth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany.
| |
Collapse
|
17
|
Cytosolic Delivery of Multidomain Cargos by the N Terminus of Pasteurella multocida Toxin. Infect Immun 2018; 86:IAI.00248-18. [PMID: 29784857 DOI: 10.1128/iai.00248-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/11/2018] [Indexed: 12/25/2022] Open
Abstract
The zoonotic pathogen Pasteurella multocida produces a 146-kDa modular toxin (PMT) that enters host cells and manipulates intracellular signaling through action on its Gα protein targets. The N terminus of PMT (PMT-N) mediates cellular uptake through receptor-mediated endocytosis, followed by the delivery of the C-terminal catalytic domain from acidic endosomes into the cytosol. The putative native cargo of PMT consists of a 710-residue polypeptide with three distinct modular subdomains (C1-C2-C3), where C1 contains a membrane localization domain (MLD), C2 has an as-yet-undefined function, and C3 catalyzes the deamidation of a specific active-site glutamine residue in Gα protein targets. However, whether the three cargo subdomains are delivered intact or undergo further proteolytic processing during or after translocation from the late endosome is unclear. Here, we demonstrate that PMT-N mediates the delivery of its native C-terminal cargo as a single polypeptide, corresponding to C1-C2-C3, including the MLD, with no evidence of cleavage between subdomains. We show that PMT-N also delivers nonnative green fluorescent protein (GFP) cargo into the cytosol, further supporting that the receptor-binding and translocation functions reside within PMT-N. Our findings further show that PMT-N can deliver C1-C2 alone but that the presence of C1-C2 is important for the cytosolic delivery of the catalytic C3 subdomain by PMT-N. In addition, we further refine the minimum C3 domain required for intracellular activity as comprising residues 1105 to 1278. These findings reinforce that PMT-N serves as the cytosolic delivery vehicle for C-terminal cargo and demonstrate that its native cargo is delivered intact as C1-C2-C3.
Collapse
|
18
|
Ng MY, Gan YH, Hagen T. Characterisation of cellular effects of Burkholderia pseudomallei cycle inhibiting factor (Cif). Biol Open 2018; 7:bio.028225. [PMID: 29848489 PMCID: PMC6078346 DOI: 10.1242/bio.028225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cycle inhibiting factors (Cifs) are type III secretion system effectors produced by some Gram-negative pathogenic bacteria including Burkholderia pseudomallei. Through their deamidase activity, Cifs inhibit the activity of Cullin RING E3 ubiquitin ligases (CRL). CRL inhibition induces the accumulation of cell cycle inhibitors p21 and p27, thereby leading to host cell cycle arrest. However, whether Cif exerts additional effects on host cells that are important in bacterial pathogenesis is currently poorly understood. In this study, we found that Cif exerts a bimodal effect on NF-κB signalling. Cif increases basal NF-κB activity. This effect is dependent on Cif-mediated activation of ERK MAPK. On the other hand, Cif inhibits NF-κB activation by TNFα and Burkholderia thailandensis infection. This inhibitory effect on NF-κB activity is partially mediated by Cif-dependent inhibition of CRLs. We also found that Cif only has a modest effect in stimulating the intracellular replication of the B. pseudomallei surrogate, B. thailandensis. The observed Cif-dependent stimulation of B. thailandensis intracellular replication was not, or was only partially, due to CRL inhibition. Furthermore, the increased B. thailandensis replication induced by Cif was independent of ERK MAPK activation. Our findings suggest that Cif likely exerts additional cellular effects through novel targets. Summary: Cycle inhibiting factor (Cif) is a Burkholderia pseudomallei virulence factor and is shown to exert both Cullin RING E3 ligase dependent and independent effects on host cells.
Collapse
Affiliation(s)
- Mei Ying Ng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yunn-Hwen Gan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Thilo Hagen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
19
|
Eisenhardt AE, Sprenger A, Röring M, Herr R, Weinberg F, Köhler M, Braun S, Orth J, Diedrich B, Lanner U, Tscherwinski N, Schuster S, Dumaz N, Schmidt E, Baumeister R, Schlosser A, Dengjel J, Brummer T. Phospho-proteomic analyses of B-Raf protein complexes reveal new regulatory principles. Oncotarget 2018; 7:26628-52. [PMID: 27034005 PMCID: PMC5042004 DOI: 10.18632/oncotarget.8427] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 03/07/2016] [Indexed: 12/19/2022] Open
Abstract
B-Raf represents a critical physiological regulator of the Ras/RAF/MEK/ERK-pathway and a pharmacological target of growing clinical relevance, in particular in oncology. To understand how B-Raf itself is regulated, we combined mass spectrometry with genetic approaches to map its interactome in MCF-10A cells as well as in B-Raf deficient murine embryonic fibroblasts (MEFs) and B-Raf/Raf-1 double deficient DT40 lymphoma cells complemented with wildtype or mutant B-Raf expression vectors. Using a multi-protease digestion approach, we identified a novel ubiquitination site and provide a detailed B-Raf phospho-map. Importantly, we identify two evolutionary conserved phosphorylation clusters around T401 and S419 in the B-Raf hinge region. SILAC labelling and genetic/biochemical follow-up revealed that these clusters are phosphorylated in the contexts of oncogenic Ras, sorafenib induced Raf dimerization and in the background of the V600E mutation. We further show that the vemurafenib sensitive phosphorylation of the T401 cluster occurs in trans within a Raf dimer. Substitution of the Ser/Thr-residues of this cluster by alanine residues enhances the transforming potential of B-Raf, indicating that these phosphorylation sites suppress its signaling output. Moreover, several B-Raf phosphorylation sites, including T401 and S419, are somatically mutated in tumors, further illustrating the importance of phosphorylation for the regulation of this kinase.
Collapse
Affiliation(s)
- Anja E Eisenhardt
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Adrian Sprenger
- Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Institute for Experimental and Clinical Pharmacology and Toxicology, ALU, Freiburg, Germany.,INSERM U976 and Universitéi Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Michael Röring
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Ricarda Herr
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Florian Weinberg
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Martin Köhler
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Sandra Braun
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Joachim Orth
- Institute for Experimental and Clinical Pharmacology and Toxicology, ALU, Freiburg, Germany
| | - Britta Diedrich
- Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Department of Dermatology, University Medical Centre, ALU, Freiburg, Germany
| | - Ulrike Lanner
- Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Natalja Tscherwinski
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Simon Schuster
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Nicolas Dumaz
- INSERM U976 and Universitéi Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Enrico Schmidt
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany
| | - Ralf Baumeister
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Freiburg Institute for Advanced Studies (FRIAS), ALU, Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany
| | - Andreas Schlosser
- Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Jörn Dengjel
- Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Department of Dermatology, University Medical Centre, ALU, Freiburg, Germany.,Freiburg Institute for Advanced Studies (FRIAS), ALU, Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany.,Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University (ALU), Freiburg, Germany.,Institute of Biology III, Faculty of Biology, ALU, Freiburg, Germany.,Centre for Biological Systems Analysis (ZBSA), Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany
| |
Collapse
|
20
|
Mohammadipanah F, Salimi F. Potential biological targets for bioassay development in drug discovery of Sturge-Weber syndrome. Chem Biol Drug Des 2017; 91:359-369. [PMID: 28941044 DOI: 10.1111/cbdd.13112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 06/10/2017] [Accepted: 09/18/2017] [Indexed: 01/02/2023]
Abstract
Sturge-Weber Syndrome (SWS) is a neurocutaneous disease with clinical manifestations including ocular (glaucoma), cutaneous (port-wine birthmark), neurologic (seizures), and vascular problems. Molecular mechanisms of SWS pathogenesis are initiated by the somatic mutation in GNAQ. Therefore, no definite treatments exist for SWS and treatment options only mitigate the intensity of its clinical manifestations. Biological assay design for drug discovery against this syndrome demands comprehensive knowledge on mechanisms which are involved in its pathogenesis. By analysis of the interrelated molecular targets of SWS, some in vitro bioassay systems can be allotted for drug screening against its progression. Development of such platforms of bioassay can bring along the implementation of high-throughput screening of natural or synthetic compounds in drug discovery programs. Regarding the fact that study of molecular targets and their integration in biological assay design can facilitate the process of effective drug discovery; some potential biological targets and their respective biological assay for SWS drug discovery are propounded in this review. For this purpose, some biological targets for SWS drug discovery such as acetylcholinesterase, alkaline phosphatase, GABAergic receptors, Hypoxia-Inducible Factor (HIF)-1α and 2α are suggested.
Collapse
Affiliation(s)
- Fatemeh Mohammadipanah
- Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Salimi
- Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
21
|
Castonguay J, Orth JHC, Müller T, Sleman F, Grimm C, Wahl-Schott C, Biel M, Mallmann RT, Bildl W, Schulte U, Klugbauer N. The two-pore channel TPC1 is required for efficient protein processing through early and recycling endosomes. Sci Rep 2017; 7:10038. [PMID: 28855648 PMCID: PMC5577145 DOI: 10.1038/s41598-017-10607-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/11/2017] [Indexed: 02/06/2023] Open
Abstract
Two-pore channels (TPCs) are localized in endo-lysosomal compartments and assumed to play an important role for vesicular fusion and endosomal trafficking. Recently, it has been shown that both TPC1 and 2 were required for host cell entry and pathogenicity of Ebola viruses. Here, we investigate the cellular function of TPC1 using protein toxins as model substrates for distinct endosomal processing routes. Toxin uptake and activation through early endosomes but not processing through other compartments were reduced in TPC1 knockout cells. Detailed co-localization studies with subcellular markers confirmed predominant localization of TPC1 to early and recycling endosomes. Proteomic analysis of native TPC1 channels finally identified direct interaction with a distinct set of syntaxins involved in fusion of intracellular vesicles. Together, our results demonstrate a general role of TPC1 for uptake and processing of proteins in early and recycling endosomes, likely by providing high local Ca2+ concentrations required for SNARE-mediated vesicle fusion.
Collapse
Affiliation(s)
- Jan Castonguay
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University, Albertstrasse 25, 79104, Freiburg, Germany
| | - Joachim H C Orth
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University, Albertstrasse 25, 79104, Freiburg, Germany
| | - Thomas Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University, Albertstrasse 25, 79104, Freiburg, Germany
| | - Faten Sleman
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University, Albertstrasse 25, 79104, Freiburg, Germany
| | - Christian Grimm
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-University, Munich, Germany
| | - Christian Wahl-Schott
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-University, Munich, Germany
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-University, Munich, Germany
| | - Robert Theodor Mallmann
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University, Albertstrasse 25, 79104, Freiburg, Germany
| | - Wolfgang Bildl
- Institute of Physiology II, Faculty of Medicine, Albert-Ludwigs-University, Hermann-Herder-Strasse 7, 79104, Freiburg, Germany
| | - Uwe Schulte
- Institute of Physiology II, Faculty of Medicine, Albert-Ludwigs-University, Hermann-Herder-Strasse 7, 79104, Freiburg, Germany.,Logopharm GmbH, Schlossstrasse 14, 79232, March-Buchheim, Germany.,Center for Biological Signaling Studies (BIOSS), Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Norbert Klugbauer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University, Albertstrasse 25, 79104, Freiburg, Germany.
| |
Collapse
|
22
|
Huang Z, Li Y, Zhao Z, Hu J, Tong X, Chen X, Liu S, Xu X, Tao Y, Wang T, Cheng X, Dai Y, Gui Y, Wu J. GNAQ mutation R183Q as a potential cause of familial Sturge-Weber syndrome: A case report. Oncol Lett 2017; 13:2665-2669. [PMID: 28454448 PMCID: PMC5403265 DOI: 10.3892/ol.2017.5791] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/06/2016] [Indexed: 02/05/2023] Open
Abstract
Sturge-Weber syndrome (SWS) is a rare neurocutaneous disorder whose etiology remains unclear. To investigate the genetic contribution underlying this disease, the genetic variants of a 4-generation family with a history of SWS was analyzed in the present study. SWS was diagnosed in 3 of the family members (II-1, III-11 and IV-6). Sanger sequencing was performed to identify mutations in G protein subunit αq (GNAQ) and RAS p21 protein activator 1 exons in the 3 patients with SWS and other unaffected family members. Notably, a non-synonymous single-nucleotide variant at codon 183 on exon 4 of the GNAQ gene was identified as the only pathogenic site. This variant generated a substitution of arginine (R) with glutamine and resulted in a change of function of the encoded protein. Evolutionary conservation analysis revealed that the mutated residue 183 (R) of GNAQ is highly conserved across several vertebrate species. Furthermore, an immunofluorescence staining assay demonstrated that the substitution of arginine with glutamine resulted in a change in the sub-cellular localization of the GNAQ recombinant protein in vitro. These findings may aid in the development of novel diagnostic markers and/or therapeutic targets for the treatment of patients with familial SWS.
Collapse
Affiliation(s)
- Zhengyi Huang
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Yuchi Li
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen Peking University and Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Zengxia Zhao
- Department of Neurology, The Shenzhen Longhua New District Central Hospital, Shenzhen, Guangdong 511180, P.R. China
| | - Jun Hu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Xiaoxin Tong
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Shuyun Liu
- Department of Neurology, The Shenzhen Longhua New District Central Hospital, Shenzhen, Guangdong 511180, P.R. China
| | - Xiaonan Xu
- Department of Neurology, Shenzhen Shekou People's Hospital, Shenzhen, Guangdong 518000, P.R. China
| | - Yongjun Tao
- Department of Neurology, Taizhou Municipal Hospital, Taizhou, Zhejiang 318000, P.R. China
| | - Tingting Wang
- Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xin Cheng
- Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yangyang Dai
- Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| | - Yaoting Gui
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen Peking University and Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
23
|
Hisao GS, Brothers MC, Ho M, Wilson BA, Rienstra CM. The membrane localization domains of two distinct bacterial toxins form a 4-helix-bundle in solution. Protein Sci 2017; 26:497-504. [PMID: 27977897 PMCID: PMC5326565 DOI: 10.1002/pro.3097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 11/12/2022]
Abstract
Membrane localization domain (MLD) was first proposed for a 4-helix-bundle motif in the crystal structure of the C1 domain of Pasteurella multocida toxin (PMT). This structure motif is also found in the crystal structures of several clostridial glycosylating toxins (TcdA, TcdB, TcsL, and TcnA). The Ras/Rap1-specific endopeptidase (RRSP) module of the multifunctional autoprocessing repeats-in-toxins (MARTX) toxin produced by Vibrio vulnificus has sequence homology to the C1-C2 domains of PMT, including a putative MLD. We have determined the solution structure for the MLDs in PMT and in RRSP using solution state NMR. We conclude that the MLDs in these two toxins assume a 4-helix-bundle structure in solution.
Collapse
Affiliation(s)
- Grant S. Hisao
- Department of ChemistryUniversity of Illinois at Urbana‐ChampaignIllinois
| | | | - Mengfei Ho
- Department of MicrobiologyUniversity of Illinois at Urbana‐ChampaignIllinois
| | - Brenda A. Wilson
- Department of MicrobiologyUniversity of Illinois at Urbana‐ChampaignIllinois
| | - Chad M. Rienstra
- Department of ChemistryUniversity of Illinois at Urbana‐ChampaignIllinois
- Department of BiochemistryUniversity of Illinois at Urbana‐ChampaignIllinois
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana‐ChampaignIllinois
| |
Collapse
|
24
|
Chakraborty S, Kloos B, Harre U, Schett G, Kubatzky KF. Pasteurella multocida Toxin Triggers RANKL-Independent Osteoclastogenesis. Front Immunol 2017; 8:185. [PMID: 28289415 PMCID: PMC5327351 DOI: 10.3389/fimmu.2017.00185] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/09/2017] [Indexed: 01/15/2023] Open
Abstract
Bone remodeling is a continuous process to retain the structural integrity and function of the skeleton. A tight coupling is maintained between osteoclast-mediated resorption of old or damaged bones and osteoblast-mediated formation of new bones for bone homeostasis. While osteoblasts differentiate from mesenchymal stem cells, osteoclasts are hematopoietic in origin and derived from myeloid precursor cells. Osteoclast differentiation is driven by two cytokines, cytokine receptor activator of NF-κB ligand (RANKL), and macrophage colony-stimulating factor. Imbalances in the activity of osteoblasts and osteoclasts result in the development of bone disorders. Bacterially caused porcine atrophic rhinitis is characterized by a loss of nasal ventral conche bones and a distortion of the snout. While Bordetella bronchiseptica strains cause mild and reversible symptoms, infection of pigs with toxigenic Pasteurella multocida strains causes a severe and irreversible decay. The responsible virulence factor Pasteurella multocida toxin (PMT) contains a deamidase activity in its catalytical domain that constitutively activates specific heterotrimeric G proteins to induce downstream signaling cascades. While osteoblasts are inhibited by the toxin, osteoclasts are activated, thus skewing bone remodeling toward excessive bone degradation. Still, the mechanism by which PMT interferes with bone homeostasis, and the reason for this unusual target tissue is not yet well understood. Here, we show that PMT has the potential to differentiate bone marrow-derived macrophages into functional osteoclasts. This toxin-mediated differentiation process is independent of RANKL, a cytokine believed to be indispensable for triggering osteoclastogenesis, as addition of osteoprotegerin to PMT-treated macrophages does not show any effect on PMT-induced osteoclast formation. Although RANKL is not a prerequisite, toxin-primed macrophages show enhanced responsiveness to low concentrations of RANKL, suggesting that the PMT-generated microenvironment offers conditions where low concentrations of RANKL lead to an increase in the number of osteoclasts resulting in increased resorption. PMT-mediated release of the osteoclastogenic cytokines such as IL-6 and TNF-α, but not IL-1, supports the differentiation process. Although the production of cytokines and the subsequent activation of signaling cascades are necessary for PMT-mediated differentiation into osteoclasts, they are not sufficient and PMT-induced activation of G protein signaling is essential for efficient osteoclastogenesis.
Collapse
Affiliation(s)
- Sushmita Chakraborty
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg , Heidelberg , Germany
| | - Bianca Kloos
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg , Heidelberg , Germany
| | - Ulrike Harre
- Department of Internal Medicine 3, Institute of Clinical Immunology, University of Erlangen-Nuremberg , Erlangen , Germany
| | - Georg Schett
- Department of Internal Medicine 3, Institute of Clinical Immunology, University of Erlangen-Nuremberg , Erlangen , Germany
| | - Katharina F Kubatzky
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg , Heidelberg , Germany
| |
Collapse
|
25
|
Carle S, Brink T, Orth JHC, Aktories K, Barth H. Auranofin Inhibits the Enzyme Activity of Pasteurella multocida Toxin PMT in Human Cells and Protects Cells from Intoxication. Toxins (Basel) 2017; 9:toxins9010032. [PMID: 28098782 PMCID: PMC5308264 DOI: 10.3390/toxins9010032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/23/2016] [Accepted: 01/10/2017] [Indexed: 11/16/2022] Open
Abstract
The AB-type protein toxin from Pasteurella multocida (PMT) contains a functionally important disulfide bond within its catalytic domain, which must be cleaved in the host cell cytosol to render the catalytic domain of PMT into its active conformation. Here, we found that the reductive potential of the cytosol of target cells, and more specifically, the activity of the thioredoxin reductase (TrxR) is crucial for this process. This was demonstrated by the strong inhibitory effect of the pharmacological TrxR inhibitor auranofin, which inhibited the intoxication of target cells with PMT, as determined by analyzing the PMT-catalyzed deamidation of GTP-binding proteins (G-proteins) in the cytosol of cells. The amount of endogenous substrate levels modified by PMT in cells pretreated with auranofin was reduced compared to cells treated with PMT alone. Auranofin had no inhibitory effect on the activity of the catalytic domain of constitutively active PMT in vitro, demonstrating that auranofin did not directly inhibit PMT activity, but interferes with the mode of action of PMT in cells. In conclusion, the results show that TrxR is crucial for the mode of action of PMT in mammalian cells, and that the drug auranofin can serve as an efficient inhibitor, which might be a starting point for novel therapeutic options against toxin-associated diseases.
Collapse
Affiliation(s)
- Stefan Carle
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, Ulm 89081, Germany.
| | - Thorsten Brink
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg 79104, Germany.
| | - Joachim H C Orth
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg 79104, Germany.
| | - Klaus Aktories
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg 79104, Germany.
- Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg 79104, Germany.
| | - Holger Barth
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, Ulm 89081, Germany.
| |
Collapse
|
26
|
|
27
|
The Type IV Secretion System Effector Protein CirA Stimulates the GTPase Activity of RhoA and Is Required for Virulence in a Mouse Model of Coxiella burnetii Infection. Infect Immun 2016; 84:2524-33. [PMID: 27324482 DOI: 10.1128/iai.01554-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/14/2016] [Indexed: 01/08/2023] Open
Abstract
Coxiella burnetii, the etiological agent of Q fever in humans, is an intracellular pathogen that replicates in an acidified parasitophorous vacuole derived from host lysosomes. Generation of this replicative compartment requires effectors delivered into the host cell by the Dot/Icm type IVb secretion system. Several effectors crucial for C. burnetii intracellular replication have been identified, but the host pathways coopted by these essential effectors are poorly defined, and very little is known about how spacious vacuoles are formed and maintained. Here we demonstrate that the essential type IVb effector, CirA, stimulates GTPase activity of RhoA. Overexpression of CirA in mammalian cells results in cell rounding and stress fiber disruption, a phenotype that is rescued by overexpression of wild-type or constitutively active RhoA. Unlike other effector proteins that subvert Rho GTPases to modulate uptake, CirA is the first effector identified that is dispensable for uptake and instead recruits Rho GTPase to promote biogenesis of the bacterial vacuole. Collectively our results highlight the importance of CirA in coopting host Rho GTPases for establishment of Coxiella burnetii infection and virulence in mammalian cell culture and mouse models of infection.
Collapse
|
28
|
Selective Membrane Redistribution and Depletion of Gαq-Protein by Pasteurella multocida Toxin. Toxins (Basel) 2016; 8:toxins8080233. [PMID: 27490568 PMCID: PMC4999849 DOI: 10.3390/toxins8080233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 02/03/2023] Open
Abstract
Pasteurella multocida toxin (PMT), the major virulence factor responsible for zoonotic atrophic rhinitis, is a protein deamidase that activates the alpha subunit of heterotrimeric G proteins. Initial activation of G alpha-q-coupled phospholipase C-beta-1 signaling by PMT is followed by uncoupling of G alpha-q-dependent signaling, causing downregulation of downstream calcium and mitogenic signaling pathways. Here, we show that PMT decreases endogenous and exogenously expressed G alpha-q protein content in host cell plasma membranes and in detergent resistant membrane (DRM) fractions. This membrane depletion of G alpha-q protein was dependent upon the catalytic activity of PMT. Results indicate that PMT-modified G alpha-q redistributes within the host cell membrane from the DRM fraction into the soluble membrane and cytosolic fractions. In contrast, PMT had no affect on G alpha-s or G beta protein levels, which are not substrate targets of PMT. PMT also had no affect on G alpha-11 levels, even though G alpha-11 can serve as a substrate for deamidation by PMT, suggesting that membrane depletion of PMT-modified G-alpha-q has specificity.
Collapse
|
29
|
Rho-modifying bacterial protein toxins from Photorhabdus species. Toxicon 2016; 116:17-22. [DOI: 10.1016/j.toxicon.2015.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/15/2015] [Accepted: 05/26/2015] [Indexed: 02/04/2023]
|
30
|
Aubert DF, Xu H, Yang J, Shi X, Gao W, Li L, Bisaro F, Chen S, Valvano MA, Shao F. A Burkholderia Type VI Effector Deamidates Rho GTPases to Activate the Pyrin Inflammasome and Trigger Inflammation. Cell Host Microbe 2016; 19:664-74. [PMID: 27133449 DOI: 10.1016/j.chom.2016.04.004] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 03/17/2016] [Accepted: 03/30/2016] [Indexed: 10/21/2022]
Abstract
Burkholderia cenocepacia is an opportunistic pathogen of the cystic fibrosis lung that elicits a strong inflammatory response. B. cenocepacia employs a type VI secretion system (T6SS) to survive in macrophages by disarming Rho-type GTPases, causing actin cytoskeletal defects. Here, we identified TecA, a non-VgrG T6SS effector responsible for actin disruption. TecA and other bacterial homologs bear a cysteine protease-like catalytic triad, which inactivates Rho GTPases by deamidating a conserved asparagine in the GTPase switch-I region. RhoA deamidation induces caspase-1 inflammasome activation, which is mediated by the familial Mediterranean fever disease protein Pyrin. In mouse infection, the deamidase activity of TecA is necessary and sufficient for B. cenocepacia-triggered lung inflammation and also protects mice from lethal B. cenocepacia infection. Therefore, Burkholderia TecA is a T6SS effector that modifies a eukaryotic target through an asparagine deamidase activity, which in turn elicits host cell death and inflammation through activation of the Pyrin inflammasome.
Collapse
Affiliation(s)
- Daniel F Aubert
- Department of Microbiology and Immunology, University of Western Ontario, London N6A 5C1, Canada
| | - Hao Xu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Jieling Yang
- National Institute of Biological Sciences, Beijing 102206, China; National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xuyan Shi
- National Institute of Biological Sciences, Beijing 102206, China
| | - Wenqing Gao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Lin Li
- National Institute of Biological Sciences, Beijing 102206, China
| | - Fabiana Bisaro
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, UK
| | - She Chen
- National Institute of Biological Sciences, Beijing 102206, China
| | - Miguel A Valvano
- Department of Microbiology and Immunology, University of Western Ontario, London N6A 5C1, Canada; Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, UK.
| | - Feng Shao
- National Institute of Biological Sciences, Beijing 102206, China; National Institute of Biological Sciences, Beijing, Collaborative Innovation Center for Cancer Medicine, Beijing 102206, China.
| |
Collapse
|
31
|
Urtatiz O, Van Raamsdonk CD. Gnaq and Gna11 in the Endothelin Signaling Pathway and Melanoma. Front Genet 2016; 7:59. [PMID: 27148356 PMCID: PMC4837292 DOI: 10.3389/fgene.2016.00059] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/01/2016] [Indexed: 12/21/2022] Open
Abstract
In this article, we first briefly outline the function of G protein coupled receptors in cancer, and then specifically examine the roles of the seven transmembrane G protein coupled Endothelin B receptor (Ednrb) and the G proteins, GNAQ and GNA11, in both melanocyte development and melanoma. Ednrb plays an essential role in melanocyte development. GNAQ and GNA11 are oncogenes when mutated in certain types of melanocytic lesions, being extremely frequent in uveal melanoma, which forms from melanocytes located in the eye. Previously, we reported that in mice, Schwann cell precursor derived melanocytes colonize the dermis and hair follicles, while the inter-follicular epidermis is populated by other melanocytes. A pattern has emerged whereby melanocytes whose activities are affected by gain-of-function mutations of the Endothelin 3 ligand and Gαq/11 are the same subset that arise from Schwann cell precursors. Furthermore, the forced expression of the constitutively active human GNAQQ209L oncogene in mouse melanocytes only causes hyper-proliferation in the subset that arise from Schwann cell precursors. This has led us to hypothesize that in Schwann cell precursor derived melanocytes, Ednrb signals through Gαq/11. Ednrb is promiscuous and may signal through other G protein alpha subunits in melanomas located in the inter-follicular epidermis.
Collapse
Affiliation(s)
- Oscar Urtatiz
- Department of Medical Genetics, University of British Columbia Vancouver, BC, Canada
| | | |
Collapse
|
32
|
Abstract
Protein deamidation has been considered a nonenzymatic process associated with protein functional decay or "aging." Recent studies implicate protein deamidation in regulating signal transduction in fundamental biological processes, such as innate immune responses. Work investigating gammaherpesviruses and bacterial pathogens indicates that microbial pathogens deploy deamidases or enzyme-deficient homologues (pseudoenzymes) to induce deamidation of key signaling components and evade host immune responses. Here, we review studies on protein deamidation in innate immune signaling and present several imminent questions concerning the roles of protein deamidation in infection and immunity.
Collapse
|
33
|
Teper D, Burstein D, Salomon D, Gershovitz M, Pupko T, Sessa G. Identification of novel Xanthomonas euvesicatoria type III effector proteins by a machine-learning approach. MOLECULAR PLANT PATHOLOGY 2016; 17:398-411. [PMID: 26104875 PMCID: PMC6638362 DOI: 10.1111/mpp.12288] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The Gram-negative bacterium Xanthomonas euvesicatoria (Xcv) is the causal agent of bacterial spot disease in pepper and tomato. Xcv pathogenicity depends on a type III secretion (T3S) system that delivers effector proteins into host cells to suppress plant immunity and promote disease. The pool of known Xcv effectors includes approximately 30 proteins, most identified in the 85-10 strain by various experimental and computational techniques. To identify additional Xcv 85-10 effectors, we applied a genome-wide machine-learning approach, in which all open reading frames (ORFs) were scored according to their propensity to encode effectors. Scoring was based on a large set of features, including genomic organization, taxonomic dispersion, hypersensitive response and pathogenicity (hrp)-dependent expression, 5' regulatory sequences, amino acid composition bias and GC content. Thirty-six predicted effectors were tested for translocation into plant cells using the hypersensitive response (HR)-inducing domain of AvrBs2 as a reporter. Seven proteins (XopAU, XopAV, XopAW, XopAP, XopAX, XopAK and XopAD) harboured a functional translocation signal and their translocation relied on the HrpF translocon, indicating that they are bona fide T3S effectors. Remarkably, four belong to novel effector families. Inactivation of the xopAP gene reduced the severity of disease symptoms in infected plants. A decrease in cell death and chlorophyll content was observed in pepper leaves inoculated with the xopAP mutant when compared with the wild-type strain. However, populations of the xopAP mutant in infected leaves were similar in size to those of wild-type bacteria, suggesting that the reduction in virulence was not caused by impaired bacterial growth.
Collapse
Affiliation(s)
- Doron Teper
- Department of Molecular Biology and Ecology of Plants, Tel Aviv University, Tel Aviv, 69978, Israel
| | - David Burstein
- Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Dor Salomon
- Department of Molecular Biology and Ecology of Plants, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Michael Gershovitz
- Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tal Pupko
- Department of Earth and Planetary Science, UC Berkeley, Berkeley, CA, 94720, USA
| | - Guido Sessa
- Department of Molecular Biology and Ecology of Plants, Tel Aviv University, Tel Aviv, 69978, Israel
| |
Collapse
|
34
|
Jank T, Belyi Y, Aktories K. Bacterial glycosyltransferase toxins. Cell Microbiol 2015; 17:1752-65. [DOI: 10.1111/cmi.12533] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 12/28/2022]
Affiliation(s)
- Thomas Jank
- Institute for Experimental and Clinical Pharmacology and Toxicology; Albert-Ludwigs University of Freiburg; Freiburg Germany
| | - Yury Belyi
- Gamaleya Research Institute; Moscow 123098 Russia
- Freiburg Institute for Advanced Studies (FRIAS); Albert-Ludwigs University of Freiburg; Freiburg Germany
| | - Klaus Aktories
- Institute for Experimental and Clinical Pharmacology and Toxicology; Albert-Ludwigs University of Freiburg; Freiburg Germany
- Freiburg Institute for Advanced Studies (FRIAS); Albert-Ludwigs University of Freiburg; Freiburg Germany
| |
Collapse
|
35
|
Zuverink M, Barbieri JT. From GFP to β-lactamase: advancing intact cell imaging for toxins and effectors. Pathog Dis 2015; 73:ftv097. [PMID: 26500183 DOI: 10.1093/femspd/ftv097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2015] [Indexed: 11/13/2022] Open
Abstract
Canonical reporters such as green fluorescent protein (GFP) and luciferase have assisted researchers in probing cellular pathways and processes. Prior research in pathogenesis depended on sensitivity of biochemical and biophysical techniques to identify effectors and elucidate entry mechanisms. Recently, the β-lactamase (βlac) reporter system has advanced toxin and effector reporting by permitting measurement of βlac delivery into the cytosol or host βlac expression in intact cells. βlac measurement in cells was facilitated by the development of the fluorogenic substrate, CCF2-AM, to identify novel effectors, target cells, and domains involved in bacterial pathogenesis. The assay is also adaptable for high-throughput screening of small molecule inhibitors against toxins, providing information on mechanism and potential therapeutic agents. The versatility and limitations of the βlac reporter system as applied to toxins and effectors are discussed in this review.
Collapse
Affiliation(s)
- Madison Zuverink
- Medical College of Wisconsin, Microbiology and Molecular Genetics, Milwaukee, WI 53226, USA
| | - Joseph T Barbieri
- Medical College of Wisconsin, Microbiology and Molecular Genetics, Milwaukee, WI 53226, USA
| |
Collapse
|
36
|
Kloos B, Chakraborty S, Lindner SG, Noack K, Harre U, Schett G, Krämer OH, Kubatzky KF. Pasteurella multocida toxin- induced osteoclastogenesis requires mTOR activation. Cell Commun Signal 2015; 13:40. [PMID: 26369790 PMCID: PMC4570759 DOI: 10.1186/s12964-015-0117-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/04/2015] [Indexed: 11/10/2022] Open
Abstract
Background Pasteurella multocida toxin (PMT) is a potent inducer of osteoclast formation. Pigs suffering from an infection with toxigenic Pasteurella multocida strains develop atrophic rhinitis characterised by a loss of turbinate bones and conchae. However, on the molecular level the process of bone loss remains largely uncharacterised. Results Recently it was found that PMT activates the serine/threonine kinase mammalian target of rapamycin (mTOR) in fibroblasts. Using RAW264.7 macrophages, we investigated the role of the mTOR complex 1 (mTORC1) in PMT-mediated osteoclast formation. PMT induces the differentiation of RAW264.7 macrophages into multinucleated, tartrate resistant acid phosphatase (TRAP) positive osteoclasts that are capable to resorb bone. In the presence of the mTORC1 inhibitor rapamycin, PMT was significantly less able to induce the formation of TRAP-positive osteoclasts. Accordingly, the resulting resorption of bone was strongly reduced. A major target of mTOR is the 70 kDa ribosomal protein S6 kinase 1 (p70 S6K1). Activated p70 S6K1 decreases the expression of programmed cell death protein 4 (PDCD4), a negative transcriptional regulator of osteoclastogenesis, at the protein and gene level. Ultimately this results in the activation of c-Jun, a component of the activator protein 1 (AP-1) complex, which is a major transcription factor for the induction of osteoclast-specific genes. We now demonstrate that c-Jun and its downstream target, the osteoclast-specific bone degrading protease cathepsin K, are upregulated upon PMT treatment in an mTOR-dependent manner. Conclusions Activation of mTOR signalling plays a central role in the formation of osteoclasts through the bacterial toxin PMT. On the molecular level, PMT-induced activation of mTOR leads to down regulation of PDCD4, a known repressor of AP-1 complex, culminating in the activation of c-Jun, an essential transcription factor for triggering osteoclastogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0117-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bianca Kloos
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Sushmita Chakraborty
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Sonja G Lindner
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Katrin Noack
- Center for Molecular Biomedicine (CMB), Department of Biochemistry, University of Jena, Hans Knöll Str. 2, 07745, Jena, Germany.,Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Erlanger Allee 101, 07747, Jena, Germany
| | - Ulrike Harre
- Department of Internal Medicine 3 and Institute of Clinical Immunology, University of Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 and Institute of Clinical Immunology, University of Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Oliver H Krämer
- Institute of Toxicology, Medical Center of the University Mainz, Obere Zahlbacher Str. 67, 55131, Mainz, Germany
| | - Katharina F Kubatzky
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| |
Collapse
|
37
|
Lin P, Cheng T, Jin S, Wu Y, Fu B, Long R, Zhao P, Xia Q. PC, a Novel Oral Insecticidal Toxin from Bacillus bombysepticus Involved in Host Lethality via APN and BtR-175. Sci Rep 2015; 5:11101. [PMID: 26057951 PMCID: PMC4460869 DOI: 10.1038/srep11101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 05/13/2015] [Indexed: 01/31/2023] Open
Abstract
Insect pests have developed resistance to chemical insecticides, insecticidal toxins as bioinsecticides or genetic protection built into crops. Consequently, novel, orally active insecticidal toxins would be valuable biological alternatives for pest control. Here, we identified a novel insecticidal toxin, parasporal crystal toxin (PC), from Bacillus bombysepticus (Bb). PC shows oral pathogenic activity and lethality towards silkworms and Cry1Ac-resistant Helicoverpa armigera strains. In vitro assays, PC after activated by trypsin binds to BmAPN4 and BtR-175 by interacting with CR7 and CR12 fragments. Additionally, trypsin-activated PC demonstrates cytotoxicity against Sf9 cells expressing BmAPN4, revealing that BmAPN4 serves as a functional receptor that participates in Bb and PC pathogenicity. In vivo assay, knocking out BtR-175 increased the resistance of silkworms to PC. These data suggest that PC is the first protein with insecticidal activity identified in Bb that is capable of causing silkworm death via receptor interactions, representing an important advance in our understanding of the toxicity of Bb and the contributions of interactions between microbial pathogens and insects to disease pathology. Furthermore, the potency of PC as an insecticidal protein makes it a good candidate for inclusion in integrated agricultural pest management systems.
Collapse
Affiliation(s)
- Ping Lin
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Tingcai Cheng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Shengkai Jin
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Yuqian Wu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Bohua Fu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Renwen Long
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Ping Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| |
Collapse
|
38
|
Weise M, Vettel C, Spiger K, Gilsbach R, Hein L, Lorenz K, Wieland T, Aktories K, Orth JHC. A systemic Pasteurella multocida toxin aggravates cardiac hypertrophy and fibrosis in mice. Cell Microbiol 2015; 17:1320-31. [PMID: 25759205 DOI: 10.1111/cmi.12436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 02/20/2015] [Accepted: 03/06/2015] [Indexed: 11/30/2022]
Abstract
Pasteurella multocida toxin (PMT) persistently activates heterotrimeric G proteins of the Gαq/11 , Gα12/13 and Gαi family without interaction with G protein-coupled receptors (GPCRs). We show that PMT acts on heart tissue in vivo and on cardiomyocytes and cardiac fibroblasts in vitro by deamidation of heterotrimeric G proteins. Increased normalized ventricle weights and fibrosis were detected after intraperitoneal administration of PMT in combination with the GPCR agonist phenylephrine. In neonatal rat cardiomyocytes, PMT stimulated the mitogen-activated protein kinase pathway, which is crucial for the development of cellular hypertrophy. The toxin induced phosphorylation of the canonical phosphorylation sites of the extracellular-regulated kinase 1/2 and, additionally, caused phosphorylation of the recently recognized autophosphorylation site, which appears to be important for the development of cellular hypertrophy. Moreover, PMT stimulated the small GTPases Rac1 and RhoA. Both switch proteins are involved in cardiomyocyte hypertrophy. In addition, PMT stimulated RhoA and Rac1 in neonatal rat cardiac fibroblasts. RhoA and Rac1 have been implicated in the regulation of connective tissue growth factor (CTGF) secretion and expression. Accordingly, we show that PMT treatment increased secretion and expression of CTGF in cardiac fibroblasts. Altogether, the data indicate that PMT is an inducer of pathological remodelling of cardiac cells and identifies the toxin as a promising tool for studying heterotrimeric G protein-dependent signalling in cardiac cells.
Collapse
Affiliation(s)
- Markus Weise
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. I, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, Freiburg, 79104, Germany
| | - Christiane Vettel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Katharina Spiger
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ralf Gilsbach
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. II, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Lutz Hein
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. II, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Klaus Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. I, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, Freiburg, 79104, Germany.,BIOSS Centre for Biological Signalling Studies, Universität Freiburg, Freiburg, Germany
| | - Joachim H C Orth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Dept. I, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, Freiburg, 79104, Germany
| |
Collapse
|
39
|
Nowland MH, Brammer DW, Garcia A, Rush HG. Biology and Diseases of Rabbits. LABORATORY ANIMAL MEDICINE 2015. [PMCID: PMC7150064 DOI: 10.1016/b978-0-12-409527-4.00010-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Beginning in 1931, an inbred rabbit colony was developed at the Phipps Institute for the Study, Treatment and Prevention of Tuberculosis at the University of Pennsylvania. This colony was used to study natural resistance to infection with tuberculosis (Robertson et al., 1966). Other inbred colonies or well-defined breeding colonies were also developed at the University of Illinois College of Medicine Center for Genetics, the Laboratories of the International Health Division of The Rockefeller Foundation, the University of Utrecht in the Netherlands, and Jackson Laboratories. These colonies were moved or closed in the years to follow. Since 1973, the U.S. Department of Agriculture has reported the total number of certain species of animals used by registered research facilities (1997). In 1973, 447,570 rabbits were used in research. There has been an overall decrease in numbers of rabbits used. This decreasing trend started in the mid-1990s. In 2010, 210,172 rabbits were used in research. Despite the overall drop in the number used in research, the rabbit is still a valuable model and tool for many disciplines.
Collapse
|
40
|
Noncanonical G-protein-dependent modulation of osteoclast differentiation and bone resorption mediated by Pasteurella multocida toxin. mBio 2014; 5:e02190. [PMID: 25389180 PMCID: PMC4235216 DOI: 10.1128/mbio.02190-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pasteurella multocida toxin (PMT) induces atrophic rhinitis in animals, which is characterized by a degradation of nasal turbinate bones, indicating an effect of the toxin on bone cells such as osteoblasts and osteoclasts. The underlying molecular mechanism of PMT was defined as a persistent activation of heterotrimeric G proteins by deamidation of a specific glutamine residue. Here, we show that PMT acts directly on osteoclast precursor cells such as bone marrow-derived CD14+ monocytes and RAW246.7 cells to induce osteoclastogenesis as measured by expression of osteoclast-specific markers such as tartrate-resistant acid phosphatase and bone resorption activity. Treatment performed solely with PMT stimulates osteoclast differentiation, showing a receptor activator of nuclear factor-κB ligand (RANKL)-independent action of the toxin. The underlying signal transduction pathway was defined as activation of the heterotrimeric G proteins Gαq/11 leading to the transactivation of Ras and the mitogen-activated protein kinase pathway. Gαq/11 transactivates Ras via its effector phospholipase Cβ-protein kinase C (PKC) involving proline-rich tyrosine kinase 2 (Pyk2). PMT-induced activation of the mitogen-activated protein kinase pathway results in stimulation of the osteoclastogenic transcription factors AP-1, NF-κB, and NFATc1. In addition, Ca2+-dependent calcineurin activation of NFAT is crucial for PMT-induced osteoclastogenesis. The data not only elucidate a rationale for PMT-dependent bone loss during atrophic rhinitis but also highlight a noncanonical, G-protein-dependent pathway toward bone resorption that is distinct from the RANKL-RANK pathway but mimics it. We define heterotrimeric G proteins as as-yet-underestimated entities/players in the maturation of osteoclasts which might be of pharmacological relevance. Pasteurella multocida toxin (PMT) induces degradation of nasal turbinate bones, leading to the syndrome of atrophic rhinitis. Recently, the molecular mechanism and substrate specificity of PMT were identified. The toxin activates heterotrimeric G proteins by a covalent modification. However, the mechanism by which PMT induces bone degradation is poorly understood. Our report demonstrates a direct effect of PMT on osteoclast precursor cells, leading to maturation of bone-degrading osteoclasts. Interestingly, PMT stimulates osteoclastogenesis independently of the cytokine RANKL, which is a key factor in induction of osteoclast differentiation. This implicates a noncanonical osteoclastogenic signaling pathway induced by PMT. The elucidated Gαq/11-dependent osteoclastogenic signal transduction pathway ends in osteoclastogenic NFAT signaling. The noncanonical, heterotrimeric G protein-dependent osteoclast differentiation process may be of pharmacological relevance, as members of this pathway are highly druggable. In particular, modulation of G protein-coupled receptor activity in osteoclast progenitors by small molecules might be of specific interest.
Collapse
|
41
|
Nakashima M, Miyajima M, Sugano H, Iimura Y, Kato M, Tsurusaki Y, Miyake N, Saitsu H, Arai H, Matsumoto N. The somatic GNAQ mutation c.548G>A (p.R183Q) is consistently found in Sturge-Weber syndrome. J Hum Genet 2014; 59:691-3. [PMID: 25374402 DOI: 10.1038/jhg.2014.95] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/13/2014] [Accepted: 10/10/2014] [Indexed: 02/04/2023]
Abstract
Sturge-Weber syndrome (SWS) is a neurocutaneous disorder characterized by capillary malformation (port-wine stains), and choroidal and leptomeningeal vascular malformations. Previously, the recurrent somatic mutation c.548G>A (p.R183Q) in the G-α q gene (GNAQ) was identified as causative in SWS and non-syndromic port-wine stain patients using whole-genome sequencing. In this study, we investigated somatic mutations in GNAQ by next-generation sequencing. We first performed targeted amplicon sequencing of 15 blood-brain-paired samples in sporadic SWS and identified the recurrent somatic c.548G>A mutation in 80% of patients (12 of 15). The percentage of mutant alleles in brain tissues of these 12 patients ranged from 3.6 to 8.9%. We found no other somatic mutations in any of the seven GNAQ exons in the remaining three patients without c.548G>A. These findings suggest that the recurrent somatic GNAQ mutation c.548G>A is the major determinant genetic factor for SWS and imply that other mutated candidate gene(s) may exist in SWS.
Collapse
Affiliation(s)
- Mitsuko Nakashima
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masakazu Miyajima
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hidenori Sugano
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasushi Iimura
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yoshinori Tsurusaki
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hirotomo Saitsu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hajime Arai
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
42
|
Hildebrand D, Bode KA, Rieß D, Cerny D, Waldhuber A, Römmler F, Strack J, Korten S, Orth JHC, Miethke T, Heeg K, Kubatzky KF. Granzyme A produces bioactive IL-1β through a nonapoptotic inflammasome-independent pathway. Cell Rep 2014; 9:910-7. [PMID: 25437548 DOI: 10.1016/j.celrep.2014.10.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/22/2014] [Accepted: 09/29/2014] [Indexed: 01/21/2023] Open
Abstract
Bacterial components are recognized by the immune system through activation of the inflammasome, eventually causing processing of the proinflammatory cytokine interleukin-1? (IL-1?), a pleiotropic cytokine and one of the most important mediators of inflammation, through the protease caspase-1. Synthesis of the precursor protein and processing into its bioactive form are tightly regulated, given that disturbed control of IL-1? release can cause severe autoinflammatory diseases or contribute to cancer development. We show that the bacterial Pasteurella multocida toxin (PMT) triggers Il1b gene transcription in macrophages independently of Toll-like receptor signaling through RhoA/Rho-kinase-mediated NF-?? activation. Furthermore, PMT mediates signal transducer and activator of transcription (STAT) protein-controlled granzyme A (a serine protease) expression in macrophages. The exocytosed granzyme A enters target cells and mediates IL-1? maturation independently of caspase-1 and without inducing cytotoxicity. These findings show that macrophages can induce an IL-1?-initiated immune response independently of inflammasome activity.
Collapse
Affiliation(s)
- Dagmar Hildebrand
- Department für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Konrad A Bode
- Department für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - David Rieß
- Department für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Daniela Cerny
- Department für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Anna Waldhuber
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Trogerstraße 30, 81675 München, Germany
| | - Franziska Römmler
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Trogerstraße 30, 81675 München, Germany
| | - Julia Strack
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Simone Korten
- Labor Lademannbogen MVZ GmbH, Lademannbogen 61-63, 22339 Hamburg, Germany
| | - Joachim H C Orth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Thomas Miethke
- Institut für Medizinische Mikrobiologie und Hygiene, Medizinische Fakultät Mannheim der Universität Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Klaus Heeg
- Department für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Katharina F Kubatzky
- Department für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| |
Collapse
|
43
|
Antic I, Biancucci M, Satchell KJF. Cytotoxicity of the Vibrio vulnificus MARTX toxin effector DUF5 is linked to the C2A subdomain. Proteins 2014; 82:2643-56. [PMID: 24935440 DOI: 10.1002/prot.24628] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/02/2014] [Accepted: 06/11/2014] [Indexed: 12/19/2022]
Abstract
The multifunctional-autoprocessing repeats-in-toxin (MARTX) toxins are bacterial protein toxins that serve as delivery platforms for cytotoxic effector domains. The domain of unknown function in position 5 (DUF5) effector domain is present in at least six different species' MARTX toxins and as a hypothetical protein in Photorhabdus spp. Its presence increases the potency of the Vibrio vulnificus MARTX toxin in mouse virulence studies, indicating DUF5 directly contributes to pathogenesis. In this work, DUF5 is shown to be cytotoxic when transiently expressed in HeLa cells. DUF5 localized to the plasma membrane dependent upon its C1 domain and the cells become rounded dependent upon its C2 domain. Both full-length DUF5 and the C2 domain caused growth inhibition when expressed in Saccharomyces cerevisiae. A structural model of DUF5 was generated based on the structure of Pasteurella multocida toxin facilitating localization of the cytotoxic activity to a 186 amino acid subdomain termed C2A. Within this subdomain, an alanine scanning mutagenesis revealed aspartate-3721 and arginine-3841 as residues critical for cytotoxicity. These residues were also essential for HeLa cell intoxication when purified DUF5 fused to anthrax toxin lethal factor was delivered cytosolically. Thermal shift experiments indicated that these conserved residues are important to maintain protein structure, rather than for catalysis. The Aeromonas hydrophila MARTX toxin DUF5(Ah) domain was also cytotoxic, while the weakly conserved C1-C2 domains from P. multocida toxin were not. Overall, this study is the first demonstration that DUF5 as found in MARTX toxins has cytotoxic activity that depends on conserved residues in the C2A subdomain.
Collapse
Affiliation(s)
- Irena Antic
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, 60611
| | | | | |
Collapse
|
44
|
What a difference a Dalton makes: bacterial virulence factors modulate eukaryotic host cell signaling systems via deamidation. Microbiol Mol Biol Rev 2014; 77:527-39. [PMID: 24006474 DOI: 10.1128/mmbr.00013-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pathogenic bacteria commonly deploy enzymes to promote virulence. These enzymes can modulate the functions of host cell targets. While the actions of some enzymes can be very obvious (e.g., digesting plant cell walls), others have more subtle activities. Depending on the lifestyle of the bacteria, these subtle modifications can be crucially important for pathogenesis. In particular, if bacteria rely on a living host, subtle mechanisms to alter host cellular function are likely to dominate. Several bacterial virulence factors have evolved to use enzymatic deamidation as a subtle posttranslational mechanism to modify the functions of host protein targets. Deamidation is the irreversible conversion of the amino acids glutamine and asparagine to glutamic acid and aspartic acid, respectively. Interestingly, all currently characterized bacterial deamidases affect the function of the target protein by modifying a single glutamine residue in the sequence. Deamidation of target host proteins can disrupt host signaling and downstream processes by either activating or inactivating the target. Despite the subtlety of this modification, it has been shown to cause dramatic, context-dependent effects on host cells. Several crystal structures of bacterial deamidases have been solved. All are members of the papain-like superfamily and display a cysteine-based catalytic triad. However, these proteins form distinct structural subfamilies and feature combinations of modular domains of various functions. Based on the diverse pathogens that use deamidation as a mechanism to promote virulence and the recent identification of multiple deamidases, it is clear that this enzymatic activity is emerging as an important and widespread feature in bacterial pathogenesis.
Collapse
|
45
|
Boularan C, Kehrl JH. Implications of non-canonical G-protein signaling for the immune system. Cell Signal 2014; 26:1269-82. [PMID: 24583286 DOI: 10.1016/j.cellsig.2014.02.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 02/22/2014] [Indexed: 01/13/2023]
Abstract
Heterotrimeric guanine nucleotide-binding proteins (G proteins), which consist of three subunits α, β, and γ, function as molecular switches to control downstream effector molecules activated by G protein-coupled receptors (GPCRs). The GTP/GDP binding status of Gα transmits information about the ligand binding state of the GPCR to intended signal transduction pathways. In immune cells heterotrimeric G proteins impact signal transduction pathways that directly, or indirectly, regulate cell migration, activation, survival, proliferation, and differentiation. The cells of the innate and adaptive immune system abundantly express chemoattractant receptors and lesser amounts of many other types of GPCRs. But heterotrimeric G-proteins not only function in classical GPCR signaling, but also in non-canonical signaling. In these pathways the guanine exchange factor (GEF) exerted by a GPCR in the canonical pathway is replaced or supplemented by another protein such as Ric-8A. In addition, other proteins such as AGS3-6 can compete with Gβγ for binding to GDP bound Gα. This competition can promote Gβγ signaling by freeing Gβγ from rapidly rebinding GDP bound Gα. The proteins that participate in these non-canonical signaling pathways will be briefly described and their role, or potential one, in cells of the immune system will be highlighted.
Collapse
Affiliation(s)
- Cédric Boularan
- B-cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - John H Kehrl
- B-cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
46
|
Sánchez-Fernández G, Cabezudo S, García-Hoz C, Benincá C, Aragay AM, Mayor F, Ribas C. Gαq signalling: the new and the old. Cell Signal 2014; 26:833-48. [PMID: 24440667 DOI: 10.1016/j.cellsig.2014.01.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/09/2014] [Indexed: 01/25/2023]
Abstract
In the last few years the interactome of Gαq has expanded considerably, contributing to improve our understanding of the cellular and physiological events controlled by this G alpha subunit. The availability of high-resolution crystal structures has led the identification of an effector-binding region within the surface of Gαq that is able to recognise a variety of effector proteins. Consequently, it has been possible to ascribe different Gαq functions to specific cellular players and to identify important processes that are triggered independently of the canonical activation of phospholipase Cβ (PLCβ), the first identified Gαq effector. Novel effectors include p63RhoGEF, that provides a link between G protein-coupled receptors and RhoA activation, phosphatidylinositol 3-kinase (PI3K), implicated in the regulation of the Akt pathway, or the cold-activated TRPM8 channel, which is directly inhibited upon Gαq binding. Recently, the activation of ERK5 MAPK by Gq-coupled receptors has also been described as a novel PLCβ-independent signalling axis that relies upon the interaction between this G protein and two novel effectors (PKCζ and MEK5). Additionally, the association of Gαq with different regulatory proteins can modulate its effector coupling ability and, therefore, its signalling potential. Regulators include accessory proteins that facilitate effector activation or, alternatively, inhibitory proteins that downregulate effector binding or promote signal termination. Moreover, Gαq is known to interact with several components of the cytoskeleton as well as with important organisers of membrane microdomains, which suggests that efficient signalling complexes might be confined to specific subcellular environments. Overall, the complex interaction network of Gαq underlies an ever-expanding functional diversity that puts forward this G alpha subunit as a major player in the control of physiological functions and in the development of different pathological situations.
Collapse
Affiliation(s)
- Guzmán Sánchez-Fernández
- Departamento de Biología Molecular and Centro de Biologia Molecular "Severo Ochoa", CSIC-UAM, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Sofía Cabezudo
- Departamento de Biología Molecular and Centro de Biologia Molecular "Severo Ochoa", CSIC-UAM, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Carlota García-Hoz
- Departamento de Biología Molecular and Centro de Biologia Molecular "Severo Ochoa", CSIC-UAM, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | | | - Anna M Aragay
- Department of Cell Biology, Molecular Biology Institute of Barcelona, Spain
| | - Federico Mayor
- Departamento de Biología Molecular and Centro de Biologia Molecular "Severo Ochoa", CSIC-UAM, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Catalina Ribas
- Departamento de Biología Molecular and Centro de Biologia Molecular "Severo Ochoa", CSIC-UAM, Universidad Autónoma de Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain.
| |
Collapse
|
47
|
A bacterial toxin catalyzing tyrosine glycosylation of Rho and deamidation of Gq and Gi proteins. Nat Struct Mol Biol 2013; 20:1273-80. [PMID: 24141704 DOI: 10.1038/nsmb.2688] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 09/11/2013] [Indexed: 12/30/2022]
Abstract
Entomopathogenic Photorhabdus asymbiotica is an emerging pathogen in humans. Here, we identified a P. asymbiotica protein toxin (PaTox), which contains a glycosyltransferase and a deamidase domain. PaTox mono-O-glycosylates Y32 (or Y34) of eukaryotic Rho GTPases by using UDP-N-acetylglucosamine (UDP-GlcNAc). Tyrosine glycosylation inhibits Rho activation and prevents interaction with downstream effectors, resulting in actin disassembly, inhibition of phagocytosis and toxicity toward insects and mammalian cells. The crystal structure of the PaTox glycosyltransferase domain in complex with UDP-GlcNAc determined at 1.8-Å resolution represents a canonical GT-A fold and is the smallest glycosyltransferase toxin known. (1)H-NMR analysis identifies PaTox as a retaining glycosyltransferase. The glutamine-deamidase domain of PaTox blocks GTP hydrolysis of heterotrimeric Gαq/11 and Gαi proteins, thereby activating RhoA. Thus, PaTox hijacks host GTPase signaling in a bidirectional manner by deamidation-induced activation and glycosylation-induced inactivation of GTPases.
Collapse
|
48
|
Kubatzky KF, Kloos B, Hildebrand D. Signaling cascades of Pasteurella multocida toxin in immune evasion. Toxins (Basel) 2013; 5:1664-81. [PMID: 24064721 PMCID: PMC3798879 DOI: 10.3390/toxins5091664] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/17/2013] [Accepted: 09/17/2013] [Indexed: 12/11/2022] Open
Abstract
Pasteurella multocida toxin (PMT) is a protein toxin found in toxigenic strains of Pasteurella multocida. PMT is the causative agent for atrophic rhinitis in pigs, a disease characterized by loss of nasal turbinate bones due to an inhibition of osteoblast function and an increase in osteoclast activity and numbers. Apart from this, PMT acts as a strong mitogen, protects from apoptosis and has an impact on the differentiation and function of immune cells. Many signaling pathways have been elucidated, however, the effect of these signaling cascades as a means to subvert the host’s immune system are just beginning to unravel.
Collapse
Affiliation(s)
- Katharina F Kubatzky
- Medical Microbiology and Hygiene, Department of Infectious Diseases, University Hospital Heidelberg, Im Neuenheimer Feld 324, Heidelberg 69120, Germany.
| | | | | |
Collapse
|
49
|
The actions of Pasteurella multocida toxin on neuronal cells. Neuropharmacology 2013; 77:9-18. [PMID: 24055502 PMCID: PMC3878393 DOI: 10.1016/j.neuropharm.2013.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 02/06/2023]
Abstract
Pasteurella multocida toxin (PMT) activates the G-proteins Gαi(1-3), Gαq, Gα11, Gα12 and Gα13 by deamidation of specific glutamine residues. A number of these alpha subunits have signalling roles in neurones. Hence we studied the action of this toxin on rat superior cervical ganglion (SCG) neurones and NG108-15 neuronal cells. Both Gαq and Gα11 could be identified in SCGs with immunocytochemistry. PMT had no direct action on Kv7 or Cav2 channels in SCGs. However PMT treatment enhanced muscarinic receptor mediated inhibition of M-current (Kv7.2 + 7. 3) as measured by a 19-fold leftward shift in the oxotremorine-M concentration–inhibition curve. Agonists of other receptors, such as bradykinin or angiotensin, that inhibit M-current did not produce this effect. However the amount of PIP2 hydrolysis could be enhanced by PMT for all three agonists. In a transduction system in SCGs that is unlikely to be affected by PMT, Go mediated inhibition of calcium current, PMT was ineffective whereas the response was blocked by pertussis toxin as expected. M1 muscarinic receptor evoked calcium mobilisation in transformed NG108-15 cells was enhanced by PMT. The calcium rises evoked by uridine triphosphate acting on endogenous P2Y2 receptors in NG108-15 cells were enhanced by PMT. The time and concentration dependence of the PMT effect was different for the resting calcium compared to the calcium rise produced by activation of P2Y2 receptors. PMT's action on these neuronal cells would suggest that if it got into the brain, symptoms of a hyperexcitable nature would be seen, such as seizures. Pasteurella multocida toxin (PMT) activates a range of G-protein alpha subunits. PMT increased muscarinic receptor mediated suppression of Kv7 potassium current in sympathetic neurones. PMT enhances both muscarinic and purinergic receptor mediated calcium mobilisation in NG108-15 cells. Both these events are mediated by the G-proteins Gq or G11. We would predict that the symptoms of central nervous system PMT toxicity would be hyperexcitable events such as seizures.
Collapse
|
50
|
Abstract
The human colon plays host to a diverse and metabolically complex community of microorganisms. While the colonic microbiome has been suggested to contribute to the development of colorectal cancer (CRC), a definitive link has not been made. The role in which the colon microflora could contribute to the initiation and/or progression of CRC is explored in this review. Potential mechanisms of bacterial oncogenesis are presented, along with lines of evidence derived from animal models of microbially induced CRC. Particular focus is given to the oncogenic capabilities of enterotoxigenic Bacteroides fragilis. Recent progress in defining the microbiome of CRC in the human population is evaluated, and the future challenges of linking specific etiologic agents to CRC are emphasized.
Collapse
Affiliation(s)
- Christine Dejea
- Department of Molecular Microbiology & Immunology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | | | | |
Collapse
|