1
|
Bonacquisti EE, Ferguson SW, Wadsworth GM, Jasiewicz NE, Wang J, Chaudhari AP, Kussatz CC, Nogueira AT, Keeley DP, Itano MS, Bolton ML, Hahn KM, Banerjee PR, Nguyen J. Fluorogenic RNA-based biomaterials for imaging and tracking the cargo of extracellular vesicles. J Control Release 2024; 374:349-368. [PMID: 39111600 PMCID: PMC11550487 DOI: 10.1016/j.jconrel.2024.07.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/25/2024]
Abstract
Extracellular vesicles (EVs), or exosomes, play important roles in physiological and pathological cellular communication and have gained substantial traction as biological drug carriers. EVs contain both short and long non-coding RNAs that regulate gene expression and epigenetic processes. To fully capitalize on the potential of EVs as drug carriers, it is important to study and understand the intricacies of EV function and EV RNA-based communication. Here we developed a genetically encodable RNA-based biomaterial, termed EXO-Probe, for tracking EV RNAs. The EXO-Probe comprises an EV-loading RNA sequence (EXO-Code), fused to a fluorogenic RNA Mango aptamer for RNA imaging. This fusion construct allowed the visualization and tracking of EV RNA and colocalization with markers of multivesicular bodies; imaging RNA within EVs, and non-destructive quantification of EVs. Overall, the new RNA-based biomaterial provides a useful and versatile means to interrogate the role of EVs in cellular communication via RNA trafficking to EVs and to study cellular sorting decisions. The system will also help lay the foundation to further improve the therapeutic efficacy of EVs as drug carriers.
Collapse
Affiliation(s)
- Emily E Bonacquisti
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Scott W Ferguson
- Department of Pharmaceutical Sciences, University at Buffalo, USA
| | - Gable M Wadsworth
- Department of Physics, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Natalie E Jasiewicz
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jinli Wang
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Ameya P Chaudhari
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Caden C Kussatz
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ana T Nogueira
- Department of Pharmacology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Daniel P Keeley
- UNC Neuroscience Microscopy Core, Carolina Institute for Developmental Disabilities, UNC Neuroscience Center, University of North Carolina at Chapel Hill, NC 25799, USA
| | - Michelle S Itano
- UNC Neuroscience Microscopy Core, Carolina Institute for Developmental Disabilities, UNC Neuroscience Center, University of North Carolina at Chapel Hill, NC 25799, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Matthew L Bolton
- Department of Systems and Information Engineering, University of Virginia, Charlottesville, 22903, USA
| | - Klaus M Hahn
- Department of Pharmacology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Priya R Banerjee
- Department of Physics, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
2
|
Aggarwal R, Mahajan P, Pandiya S, Bajaj A, Verma SK, Yadav P, Kharat AS, Khan AU, Dua M, Johri AK. Antibiotic resistance: a global crisis, problems and solutions. Crit Rev Microbiol 2024; 50:896-921. [PMID: 38381581 DOI: 10.1080/1040841x.2024.2313024] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/09/2024] [Accepted: 01/28/2024] [Indexed: 02/23/2024]
Abstract
Healthy state is priority in today's world which can be achieved using effective medicines. But due to overuse and misuse of antibiotics, a menace of resistance has increased in pathogenic microbes. World Health Organization (WHO) has announced ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) as the top priority pathogens as these have developed resistance against certain antibiotics. To combat such a global issue, it is utmost important to identify novel therapeutic strategies/agents as an alternate to such antibiotics. To name certain antibiotic adjuvants including: inhibitors of beta-lactamase, efflux pumps and permeabilizers for outer membrane can potentially solve the antibiotic resistance problems. In this regard, inhibitors of lytic domain of lytic transglycosylases provide a novel way to not only act as an alternate to antibiotics but also capable of restoring the efficiency of previously resistant antibiotics. Further, use of bacteriophages is another promising strategy to deal with antibiotic resistant pathogens. Taking in consideration the alternatives of antibiotics, a green synthesis nanoparticle-based therapy exemplifies a good option to combat microbial resistance. As horizontal gene transfer (HGT) in bacteria facilitates the evolution of new resistance strains, therefore identifying the mechanism of resistance and development of inhibitors against it can be a novel approach to combat such problems. In our perspective, host-directed therapy (HDT) represents another promising strategy in combating antimicrobial resistance (AMR). This approach involves targeting specific factors within host cells that pathogens rely on for their survival, either through replication or persistence. As many new drugs are under clinical trials it is advisable that more clinical data and antimicrobial stewardship programs should be conducted to fully assess the clinical efficacy and safety of new therapeutic agents.
Collapse
Affiliation(s)
- Rupesh Aggarwal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Pooja Mahajan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sameeksha Pandiya
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Aayushi Bajaj
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Shailendra Kumar Verma
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Puja Yadav
- Department of Microbiology, Central University of Haryana, Mahendergarh, India
| | - Arun S Kharat
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Asad Ullah Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Meenakshi Dua
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Atul Kumar Johri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
3
|
Chen P, Cao XW, Dong JW, Zhao J, Wang FJ. Saponin and Ribosome-Inactivating Protein Synergistically Trigger Lysosome-Dependent Apoptosis by Inhibiting Lysophagy: Potential to Become a New Antitumor Strategy. Mol Pharm 2024; 21:2993-3005. [PMID: 38722865 DOI: 10.1021/acs.molpharmaceut.4c00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The susceptibility of lysosomal membranes in tumor cells to cationic amphiphilic drugs (CADs) enables CADs to induce lysosomal membrane permeabilization (LMP) and trigger lysosome-dependent cell death (LDCD), suggesting a potential antitumor therapeutic approach. However, the existence of intrinsic lysosomal damage response mechanisms limits the display of the pharmacological activity of CADs. In this study, we report that low concentrations of QS-21, a saponin with cationic amphiphilicity extracted from Quillaja Saponaria tree, can induce LMP but has nontoxicity to tumor cells. QS-21 and MAP30, a type I ribosome-inactivating protein, synergistically induce apoptosis in tumor cells at low concentrations of both. Mechanistically, QS-21-induced LMP helps MAP30 escape from endosomes or lysosomes and subsequently enter the endoplasmic reticulum, where MAP30 downregulates the expression of autophagy-associated LC3 proteins, thereby inhibiting lysophagy. The inhibition of lysophagy results in the impaired clearance of damaged lysosomes, leading to the leakage of massive lysosomal contents such as cathepsins into the cytoplasm, ultimately triggering LDCD. In summary, our study showed that coadministration of QS-21 and MAP30 amplified the lysosomal disruption and can be a new synergistic LDCD-based antitumor therapy.
Collapse
Affiliation(s)
- Piao Chen
- Department of Applied Biology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Xue-Wei Cao
- Department of Applied Biology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang, Zhejiang 322100, People's Republic of China
| | - Jing-Wen Dong
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang, Zhejiang 322100, People's Republic of China
| | - Jian Zhao
- Department of Applied Biology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Fu-Jun Wang
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang, Zhejiang 322100, People's Republic of China
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| |
Collapse
|
4
|
Kidner RQ, Goldstone EB, Rodefeld HJ, Brokaw LP, Gonzalez AM, Ros-Rocher N, Gerdt JP. Exogenous lipid vesicles induce endocytosis-mediated cellular aggregation in a close unicellular relative of animals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.593945. [PMID: 38798415 PMCID: PMC11118469 DOI: 10.1101/2024.05.14.593945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Capsaspora owczarzaki is a protozoan that may both reveal aspects of animal evolution and also curtail the spread of schistosomiasis, a neglected tropical disease. Capsaspora exhibits a chemically regulated aggregative behavior that resembles cellular aggregation in some animals. This behavior may have played a key role in the evolution of animal multicellularity. Additionally, this aggregative behavior may be important for Capsaspora 's ability to colonize the intermediate host of parasitic schistosomes and potentially prevent the spread of schistosomiasis. Both applications demand elucidation of the molecular mechanism of Capsaspora aggregation. Toward this goal, we first determined the necessary chemical properties of lipid cues that activate aggregation. We found that a wide range of abundant zwitterionic lipids induced aggregation, revealing that the aggregative behavior could be activated by diverse lipid-rich conditions. Furthermore, we demonstrated that aggregation in Capsaspora requires clathrin-mediated endocytosis, highlighting the potential significance of endocytosis-linked cellular signaling in recent animal ancestors. Finally, we found that aggregation was initiated by post-translational activation of cell-cell adhesion-not transcriptional regulation of cellular adhesion machinery. Our findings illuminate the chemical, molecular and cellular mechanisms that regulate Capsaspora aggregative behavior-with implications for the evolution of animal multicellularity and the transmission of parasites.
Collapse
|
5
|
Boesze-Battaglia K, Cohen GH, Bates PF, Walker LM, Zekavat A, Shenker BJ. Cellugyrin (synaptogyrin-2) dependent pathways are used by bacterial cytolethal distending toxin and SARS-CoV-2 virus to gain cell entry. Front Cell Infect Microbiol 2024; 14:1334224. [PMID: 38698905 PMCID: PMC11063343 DOI: 10.3389/fcimb.2024.1334224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/19/2024] [Indexed: 05/05/2024] Open
Abstract
Aggregatibacter actinomycetemcomitans cytolethal distending toxin (Cdt) is capable of intoxicating lymphocytes macrophages, mast cells and epithelial cells. Following Cdt binding to cholesterol, in the region of membrane lipid rafts, the CdtB and CdtC subunits are internalized and traffic to intracellular compartments. These events are dependent upon, cellugyrin, a critical component of synaptic like microvesicles (SLMVCg+). Target cells, such as Jurkat cells, rendered unable to express cellugyrin are resistant to Cdt-induced toxicity. Similar to Cdt, SARS-CoV-2 entry into host cells is initiated by binding to cell surface receptors, ACE-2, also associated with cholesterol-rich lipid rafts; this association leads to fusion and/or endocytosis of viral and host cell membranes and intracellular trafficking. The similarity in internalization pathways for both Cdt and SARS-CoV-2 led us to consider the possibility that cellugyrin was a critical component in both processes. Cellugyrin deficient Calu-3 cells (Calu-3Cg-) were prepared using Lentiviral particles containing shRNA; these cells were resistant to infection by VSV/SARS-CoV-2-spike pseudotype virus and partially resistant to VSV/VSV-G pseudotype virus. Synthetic peptides representing various regions of the cellugyrin protein were prepared and assessed for their ability to bind to Cdt subunits using surface plasmon resonance. Cdt was capable of binding to a region designated the middle outer loop (MOL) which corresponds to a region extending into the cytoplasmic surface of the SLMVCg+. SARS-CoV-2 spike proteins were assessed for their ability to bind to cellugyrin peptides; SARS-CoV-2 full length spike protein preferentially binds to a region within the SLMVCg+ lumen, designated intraluminal loop 1A. SARS-CoV-2-spike protein domain S1, which contains the receptor binding domains, binds to cellugyrin N-terminus which extends out from the cytoplasmic surface of SLMV. Binding specificity was further analyzed using cellugyrin scrambled peptide mutants. We propose that SLMVCg+ represent a component of a common pathway that facilitates pathogen and/or pathogen-derived toxins to gain host cell entry.
Collapse
Affiliation(s)
- Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Gary H. Cohen
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Paul F. Bates
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Lisa M. Walker
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ali Zekavat
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Bruce J. Shenker
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
6
|
Jungbauer-Groznica M, Wiese K, Fischer I, Markus J, Chang TH, Gösler I, Kowalski H, Blaas D, Real-Hohn A. Aichivirus A1 replicates in human intestinal epithelium and bronchial tissue: Lung-gut axis? Virus Res 2024; 342:199338. [PMID: 38373599 PMCID: PMC10901855 DOI: 10.1016/j.virusres.2024.199338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/21/2024]
Abstract
The role of aichivirus A1 (AiV-A1) in acute gastroenteritis remains controversial and in vitro data illustrating its pathogenesis in suitable human models are scarce. Here, we demonstrate that AiV-A1 isolate A846/88 replicates in ApoA1- (absorptive) and Ki-67-positive (proliferative) enterocytes in stem cell-derived human small intestinal epithelium (HIE) as well as in patient biopsy samples, but not in any of the tested human cell lines. The infection did not result in tissue damage and did not trigger type I and type III interferon (IFN) signalling, whereas the control, human coxsackievirus B3 (strain Nancy), triggered both IFNs. To investigate the tissue tropism, we infected a human tracheal/bronchial epithelium model (HTBE) with AiV-A1 isolates A846/88 and kvgh99012632/2010 and, as a control, with rhinovirus A2 (RV-A2). AiV-A1 isolate kvgh99012632/2010, but not isolate A846/88, replicated in HTBE and induced type III IFN and ISGs signalling. By using various pharmacological inhibitors, we elaborated that cellular entry of AiV-A1 depends on clathrin, dynamin, and lipid rafts and is strongly reliant on endosome acidification. Viral particles co-localised with Rab5a-positive endosomes and promoted leakage of endosomal content. Our data shed light on the early events of AiV-A1 infection and reveal that different isolates exhibit distinct tissue tropism. This supports its clinical importance as a human pathogen with the potential to evolve toward broader tissue specificity.
Collapse
Affiliation(s)
- Martin Jungbauer-Groznica
- Center for Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria; Virus and Immunity Unit, Institute Pasteur, Université Paris Cité, Paris, France
| | - Konstantin Wiese
- Center for Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Irmgard Fischer
- Histology Facility, Vienna Biocenter, Max Perutz Laboratories, Vienna, Austria
| | - Jan Markus
- MatTek In Vitro Life Science Laboratories, Bratislava, Slovakia
| | - Tsung-Hsien Chang
- National Defense Medical Center, Department of Microbiology and Immunology, Taipei, Taiwan
| | - Irene Gösler
- Center for Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Heinrich Kowalski
- Center for Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria.
| | - Dieter Blaas
- Center for Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Antonio Real-Hohn
- Center for Medical Biochemistry, Vienna Biocenter, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Minns LA, Sausman KT, Brown AP, York RA, McCall JR. Karenia brevis Extract Induces Cellular Entry through Distinct Mechanisms in Phagocytic RAW 264.7 Macrophages versus Non-Phagocytic Vero Cells. Mar Drugs 2023; 22:4. [PMID: 38276642 PMCID: PMC10820030 DOI: 10.3390/md22010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Marine algae extracts are an important area of potential drug discovery; however, nearly all studies to date have used non-fluorescent-based methods to determine changes in target cell activity. Many of the most robust immunological and cellular analyses rely on fluorescent probes and readouts, which can be problematic when the algae extract is fluorescent itself. In this study, we identified the fluorescent spectrum of an isolated extract from the marine dinoflagellate Karenia brevis, which included two fluorescing components: chlorophyll α and pheophytin α. When excited at 405 nm and 664 nm, the extract emitted fluorescence at 676 nm and 696 nm, respectively. The extract and its fluorescing components, chlorophyll α and pheophytin α, entered phagocytic RAW 264.7 macrophages and non-phagocytic Vero kidney cells through distinct mechanisms. When incubated with the extract and its main components, both the RAW 264.7 macrophages and the Vero cells accumulated fluorescence as early as 30 min and continued through 48 h. Vero kidney cells accumulated the K. brevis fluorescent extract through a dynamin-independent and acidified endosomal-dependent mechanism. RAW 264.7 macrophages accumulated fluorescent extract through a dynamin-independent, acidified endosomal-independent mechanism, which supports accumulation through phagocytosis. Furthermore, RAW 264.7 macrophages downregulated cell-surface expression of CD206 in response to extract stimulation indicating activation of phagocytic responses and potential immunosuppression of these immune cells. This study represents the first characterization of the cellular update of K. brevis extracts in phagocytic versus non-phagocytic cells. The data suggest the importance of understanding cellular uptake of fluorescing algae extracts and their mechanism of action for future drug discovery efforts.
Collapse
Affiliation(s)
- Laurie A. Minns
- School of Nursing, College of Health and Human Services, University of North Carolina Wilmington, 601 S. College Road, Wilmington, NC 28403, USA; (L.A.M.)
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| | - Kathryn T. Sausman
- School of Nursing, College of Health and Human Services, University of North Carolina Wilmington, 601 S. College Road, Wilmington, NC 28403, USA; (L.A.M.)
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| | - Ariel P. Brown
- School of Nursing, College of Health and Human Services, University of North Carolina Wilmington, 601 S. College Road, Wilmington, NC 28403, USA; (L.A.M.)
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| | - Robert A. York
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
- Algal Resources Collection, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| | - Jennifer R. McCall
- School of Nursing, College of Health and Human Services, University of North Carolina Wilmington, 601 S. College Road, Wilmington, NC 28403, USA; (L.A.M.)
- Center for Marine Science, University of North Carolina Wilmington, 5600 Marvin K Moss Lane, Wilmington, NC 28409, USA
| |
Collapse
|
8
|
Wiest MJ, Baert L, Gu C, Gayler KM, Ham H, Gorvel L, Keddis MT, Griffing LW, Joo H, Gorvel JP, Billadeau DD, Kane RR, Oh S. Endosomal trafficking inhibitor EGA can control TLR7-mediated IFNα expression by human plasmacytoid dendritic cells. Front Immunol 2023; 14:1202197. [PMID: 38077311 PMCID: PMC10704457 DOI: 10.3389/fimmu.2023.1202197] [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: 04/07/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Plasmacytoid dendritic cells (pDC) are the major producer of type 1 IFN in response to TLR7 agonists. Aberrant TLR7 activation and type 1 IFN expression by pDCs are linked to the pathogenesis of certain types of autoimmune diseases, including systemic lupus erythematosus (SLE). This study investigated the underlying mechanisms for TLR7-mediated cytokine expression by pDCs using a late endosome trafficking inhibitor, EGA (4-bromobenzaldehyde N-(2,6-dimethylphenyl) semicarbazone). We found that EGA treatment decreased IFNα expression by pDCs stimulated with imiquimod (R837), single-stranded RNA40, and influenza virus. EGA also decreased TNFα expression and secretion by R837-stimulated pDCs. Mechanistically, EGA treatment decreased phosphorylation of IKKα/β, STAT1, and p38, and prolonged degradation of IκBα. Furthermore, EGA treatment decreased the colocalization of 3F, a substituted adenine TLR7 agonist, with LAMP1+ compartments in pDCs. EGA was also capable of diminishing IFNα expression by SLE pDCs treated with R837 or live PR8/A/34 influenza viruses. Therefore, we concluded that trafficking of TLR7 agonists to LAMP1+ compartments is important for IFNα expression by pDCs. Data from this study support additional examinations of the potential benefits of EGA in treating type 1 IFN-associated inflammatory diseases in the future.
Collapse
Affiliation(s)
- Matthew J. Wiest
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Laurie Baert
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Chao Gu
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Kevin M. Gayler
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, United States
| | - Hyoungjun Ham
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Laurent Gorvel
- CRCM, Aix Marseille Universite, INSERM, Marseille, France
| | - Mira T. Keddis
- Department of Nephrology, Mayo Clinic, Scottsdale, AZ, United States
| | - Leroy W. Griffing
- Department of Rheumatology, Mayo Clinic, Scottsdale, AZ, United States
| | - HyeMee Joo
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, United States
| | | | | | - Robert R. Kane
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, United States
| | - SangKon Oh
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, United States
| |
Collapse
|
9
|
Enslin LE, Purkait K, Pozza MD, Saubamea B, Mesdom P, Visser HG, Gasser G, Schutte-Smith M. Rhenium(I) Tricarbonyl Complexes of 1,10-Phenanthroline Derivatives with Unexpectedly High Cytotoxicity. Inorg Chem 2023; 62:12237-12251. [PMID: 37489813 PMCID: PMC10410611 DOI: 10.1021/acs.inorgchem.3c00730] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Indexed: 07/26/2023]
Abstract
Eight rhenium(I) tricarbonyl aqua complexes with the general formula fac-[Re(CO)3(N,N'-bid)(H2O)][NO3] (1-8), where N,N'-bid is (2,6-dimethoxypyridyl)imidazo[4,5-f]1,10-phenanthroline (L1), (indole)imidazo[4,5-f]1,10-phenanthroline (L2), (5-methoxyindole)-imidazo[4,5-f]1,10-phenanthroline (L3), (biphenyl)imidazo[4,5-f]1,10-phenanthroline (L4), (fluorene)imidazo[4,5-f]1,10-phenanthroline (L5), (benzo[b]thiophene)imidazo[4,5-f]1,10-phenanthroline (L6), (5-bromothiazole)imidazo[4,5-f]1,10-phenanthroline (L7), and (4,5-dimethylthiophene)imidazo[4,5-f]1,10-phenanthroline (L8), were synthesized and characterized using 1H and 13C{1H} NMR, FT-IR, UV/Vis absorption spectroscopy, and ESI-mass spectrometry, and their purity was confirmed by elemental analysis. The stability of the complexes in aqueous buffer solution (pH 7.4) was confirmed by UV/Vis spectroscopy. The cytotoxicity of the complexes (1-8) was then evaluated on prostate cancer cells (PC3), showing a low nanomolar to low micromolar in vitro cytotoxicity. Worthy of note, three of the Re(I) tricarbonyl complexes showed very low (IC50 = 30-50 nM) cytotoxic activity against PC3 cells and up to 26-fold selectivity over normal human retinal pigment epithelial-1 (RPE-1) cells. The cytotoxicity of both complexes 3 and 6 was lowered under hypoxic conditions in PC3 cells. However, the compounds were still 10 times more active than cisplatin in these conditions. Additional biological experiments were then performed on the most selective complexes (complexes 3 and 6). Cell fractioning experiments followed by ICP-MS studies revealed that 3 and 6 accumulate mostly in the mitochondria and nucleus, respectively. Despite the respective mitochondrial and nuclear localization of 3 and 6, 3 did not trigger the apoptosis pathways for cell killing, whereas 6 can trigger apoptosis but not as a major pathway. Complex 3 induced a paraptosis pathway for cell killing while 6 did not induce any of our other tested pathways, namely, necrosis, paraptosis, and autophagy. Both complexes 3 and 6 were found to be involved in mitochondrial dysfunction and downregulated the ATP production of PC3 cells. To the best of our knowledge, this report presents some of the most cytotoxic Re(I) carbonyl complexes with exceptionally low nanomolar cytotoxic activity toward prostate cancer cells, demonstrating further the future viability of utilizing rhenium in the fight against cancer.
Collapse
Affiliation(s)
- Lucy E. Enslin
- Department
of Chemistry, University of the Free State, Bloemfontein 9301, South Africa
| | - Kallol Purkait
- Chimie
ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for
Inorganic Chemistry, F-75005 Paris, France
| | - Maria Dalla Pozza
- Chimie
ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for
Inorganic Chemistry, F-75005 Paris, France
| | - Bruno Saubamea
- Plateforme
Imagerie Cellulaire et Moléculaire, Faculté de Pharmacie, Université Paris Cité, F-75270 Paris, France
| | - Pierre Mesdom
- Chimie
ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for
Inorganic Chemistry, F-75005 Paris, France
| | - Hendrik G. Visser
- Department
of Chemistry, University of the Free State, Bloemfontein 9301, South Africa
| | - Gilles Gasser
- Chimie
ParisTech, PSL University, CNRS, Institute of Chemistry of Life and Health Sciences, Laboratory for
Inorganic Chemistry, F-75005 Paris, France
| | | |
Collapse
|
10
|
Roa-Linares VC, Escudero-Flórez M, Vicente-Manzanares M, Gallego-Gómez JC. Host Cell Targets for Unconventional Antivirals against RNA Viruses. Viruses 2023; 15:v15030776. [PMID: 36992484 PMCID: PMC10058429 DOI: 10.3390/v15030776] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/12/2023] [Accepted: 02/28/2023] [Indexed: 03/31/2023] Open
Abstract
The recent COVID-19 crisis has highlighted the importance of RNA-based viruses. The most prominent members of this group are SARS-CoV-2 (coronavirus), HIV (human immunodeficiency virus), EBOV (Ebola virus), DENV (dengue virus), HCV (hepatitis C virus), ZIKV (Zika virus), CHIKV (chikungunya virus), and influenza A virus. With the exception of retroviruses which produce reverse transcriptase, the majority of RNA viruses encode RNA-dependent RNA polymerases which do not include molecular proofreading tools, underlying the high mutation capacity of these viruses as they multiply in the host cells. Together with their ability to manipulate the immune system of the host in different ways, their high mutation frequency poses a challenge to develop effective and durable vaccination and/or treatments. Consequently, the use of antiviral targeting agents, while an important part of the therapeutic strategy against infection, may lead to the selection of drug-resistant variants. The crucial role of the host cell replicative and processing machinery is essential for the replicative cycle of the viruses and has driven attention to the potential use of drugs directed to the host machinery as therapeutic alternatives to treat viral infections. In this review, we discuss small molecules with antiviral effects that target cellular factors in different steps of the infectious cycle of many RNA viruses. We emphasize the repurposing of FDA-approved drugs with broad-spectrum antiviral activity. Finally, we postulate that the ferruginol analog (18-(phthalimide-2-yl) ferruginol) is a potential host-targeted antiviral.
Collapse
Affiliation(s)
- Vicky C Roa-Linares
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Manuela Escudero-Flórez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain
| | - Juan C Gallego-Gómez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| |
Collapse
|
11
|
Polakowski N, Sarker MAK, Hoang K, Boateng G, Rushing AW, Kendle W, Pique C, Green PL, Panfil AR, Lemasson I. HBZ upregulates myoferlin expression to facilitate HTLV-1 infection. PLoS Pathog 2023; 19:e1011202. [PMID: 36827461 PMCID: PMC9994761 DOI: 10.1371/journal.ppat.1011202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/08/2023] [Accepted: 02/10/2023] [Indexed: 02/26/2023] Open
Abstract
The complex retrovirus, human T-cell leukemia virus type 1 (HTLV-1), primarily infects CD4+ T-cells in vivo. Infectious spread within this cell population requires direct contact between virally-infected and target cells. The HTLV-1 accessory protein, HBZ, was recently shown to enhance HTLV-1 infection by activating intracellular adhesion molecule 1 (ICAM-1) expression, which promotes binding of infected cells to target cells and facilitates formation of a virological synapse. In this study we show that HBZ additionally enhances HTLV-1 infection by activating expression of myoferlin (MyoF), which functions in membrane fusion and repair and vesicle transport. Results from ChIP assays and quantitative reverse transcriptase PCR indicate that HBZ forms a complex with c-Jun or JunB at two enhancer sites within the MYOF gene and activates transcription through recruitment of the coactivator p300/CBP. In HTLV-1-infected T-cells, specific inhibition of MyoF using the drug, WJ460, or shRNA-mediated knockdown of MyoF reduced infection efficiency. This effect was associated with a decrease in cell adhesion and an intracellular reduction in the abundance of HTLV-1 envelope (Env) surface unit (SU) and transmembrane domain (TM). Lysosomal protease inhibitors partially restored SU levels in WJ460-treated cells, and SU localization to LAMP-2 sites was increased by MyoF knockdown, suggesting that MyoF restricts SU trafficking to lysosomes for degradation. Consistent with these effects, less SU was associated with cell-free virus particles. Together, these data suggest that MyoF contributes to HTLV-1 infection through modulation of Env trafficking and cell adhesion.
Collapse
Affiliation(s)
- Nicholas Polakowski
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina, United States of America
| | - Md Abu Kawsar Sarker
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina, United States of America
| | - Kimson Hoang
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina, United States of America
| | - Georgina Boateng
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina, United States of America
| | - Amanda W. Rushing
- Catawba College, Department of Biology, Salisbury, North Carolina, United States of America
| | - Wesley Kendle
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina, United States of America
| | - Claudine Pique
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Patrick L. Green
- Center for Retrovirus Research and Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Amanda R. Panfil
- Center for Retrovirus Research and Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Isabelle Lemasson
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
12
|
Pham TT, Chen H, Nguyen PHD, Jayasinghe MK, Le AH, Le MT. Endosomal escape of nucleic acids from extracellular vesicles mediates functional therapeutic delivery. Pharmacol Res 2023; 188:106665. [PMID: 36657503 DOI: 10.1016/j.phrs.2023.106665] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/06/2023] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
Extracellular vesicles hold great promise as a drug delivery platform for RNA-based therapeutics. However, there is a lack of experimental evidence for the intracellular trafficking of nucleic acid cargos, specifically, whether they are capable of escaping from the endolysosomal confinement in the recipient cells to be released into the cytosol and hence, interact with their cytoplasmic targets. Here, we demonstrated how red blood cell-derived extracellular vesicles (RBCEVs) release their therapeutic RNA/DNA cargos at specific intracellular compartments characteristic of late endosomes and lysosomes. The released cargos were functional and capable of knocking down genes of interest in recipient cells, resulting in tumor suppression in vitro and in an acute myeloid leukemia murine model without causing significant toxicity. Notably, surface functionalization of RBCEVs with an anti-human CXCR4 antibody facilitated their specific uptake by CXCR4+ leukemic cells, leading to enhanced gene silencing efficiency. Our results provide insights into the cellular uptake mechanisms and endosomal escape routes of nucleic acid cargos delivered by RBCEVs which have important implications for further improvements of the RBCEV-based delivery system.
Collapse
Affiliation(s)
- Tuan Thach Pham
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Huan Chen
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Phuong Hoang Diem Nguyen
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Migara Kavishka Jayasinghe
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anh Hong Le
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Minh Tn Le
- Department of Pharmacology and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
13
|
Wiest MJ, Gu C, Ham H, Gorvel L, Keddis MT, Griffing LW, Joo H, Gorvel JP, Billadeau DD, Oh S. Disruption of endosomal trafficking with EGA alters TLR9 cytokine response in human plasmacytoid dendritic cells. Front Immunol 2023; 14:1144127. [PMID: 37020542 PMCID: PMC10067882 DOI: 10.3389/fimmu.2023.1144127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/10/2023] [Indexed: 04/07/2023] Open
Abstract
Plasmacytoid dendritic cells (pDCs) exhibit bifurcated cytokine responses to TLR9 agonists, an IRF7-mediated type 1 IFN response or a pro-inflammatory cytokine response via the activation of NF-κB. This bifurcated response has been hypothesized to result from either distinct signaling endosomes or endo-lysosomal trafficking delay of TLR9 agonists allowing for autocrine signaling to affect outcomes. Utilizing the late endosome trafficking inhibitor, EGA, we assessed the bifurcated cytokine responses of pDCs to TLR9 stimulation. EGA treatment of pDCs diminished both IFNα and pro-inflammatory cytokine expression induced by CpG DNAs (D- and K-type), CpG-DNAs complexed with DOTAP, and genomic DNAs complexed with LL37. Mechanistically, EGA suppressed phosphorylation of IKKα/β, STAT1, Akt, and p38, and decreased colocalization of CpG oligodeoxynucleotides with LAMP+ endo-lysosomes. EGA also diminished type 1 IFN expression by pDCs from systemic lupus erythematosus patients. Therefore, our findings help understand mechanisms for the bifurcated cytokine responses by pDCs and support future examination of the potential benefit of EGA in treating type 1 IFN-associated inflammatory diseases in the future.
Collapse
Affiliation(s)
- Matthew J. Wiest
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Baylor Institute of Biomedical Studies, Baylor University, Waco, TX, United States
| | - Chao Gu
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Hyoungjun Ham
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Laurent Gorvel
- CRCM, Aix Marseille Universite, INSERM, Marseille, France
| | - Mira T. Keddis
- Department of Nephrology, Mayo Clinic, Scottsdale, AZ, United States
| | - Leroy W. Griffing
- Department of Rheumatology, Mayo Clinic, Scottsdale, AZ, United States
| | - HyeMee Joo
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Baylor Institute of Biomedical Studies, Baylor University, Waco, TX, United States
| | | | | | - SangKon Oh
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Baylor Institute of Biomedical Studies, Baylor University, Waco, TX, United States
- *Correspondence: SangKon Oh,
| |
Collapse
|
14
|
LAMP3/CD63 Expression in Early and Late Endosomes in Human Vaginal Epithelial Cells Is Associated with Enhancement of HSV-2 Infection. J Virol 2022; 96:e0155322. [PMID: 36350153 PMCID: PMC9749459 DOI: 10.1128/jvi.01553-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Herpes simplex virus 2 (HSV-2) is a lifelong sexually transmitted virus that disproportionately infects women through heterosexual transmission in the vaginal tract. The vaginal epithelium is known to be highly susceptible to HSV-2 infection; however, the cellular mechanism of HSV-2 uptake and replication in vaginal epithelium has not been extensively studied. Previously, we observed that lysosomal-associated membrane protein-3 (LAMP3/CD63) was among the highly upregulated genes during HSV-2 infection of human vaginal epithelial cell line VK2, leading us to posit that LAMP3/CD63 may play a role in HSV-2 infection. Consequently, we generated two gene-altered VK2-derived cell lines, a LAMP3-overexpressed (OE) line and a LAMP3 knockout (KO) line. The wild-type VK2 and the LAMP3 OE and KO cell lines were grown in air-liquid interface (ALI) cultures for 7 days and infected with HSV-2. Twenty-four hours postinfection, LAMP3 OE cells produced and released significantly higher numbers of HSV-2 virions than wild-type VK2 cells, while virus production was greatly attenuated in LAMP3 KO cells, indicating a functional association between LAMP3/CD63 expression and HSV-2 replication. Fluorescence microscopy of HSV-2-infected cells revealed that HSV-2 colocalized with LAMP3 in both early endosomes and lysosomal compartments. In addition, blocking endosomal maturation or late endosomal/lysosomal fusion using specific inhibitors resulted in reduced HSV-2 replication in VK2 cells. Similarly, LAMP3 KO cells exhibited very low viral entry and association with endosomes, while LAMP3 OE cells demonstrated large amounts of virus that colocalized with LAMP3/CD63 in endosomes and lysosomes. IMPORTANCE Collectively, these results showed that HSV-2 is taken up by human vaginal epithelial cells through an endosomal-lysosomal pathway in association with LAMP3, which plays a crucial role in the enhancement of HSV-2 replication. These findings provide the basis for the future design of antiviral agents for prophylactic measures against HSV-2 infection.
Collapse
|
15
|
Acid-induced disassembly of the Haemophilus ducreyi cytolethal distending toxin. Biochem Biophys Res Commun 2022; 636:57-63. [DOI: 10.1016/j.bbrc.2022.10.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 11/21/2022]
|
16
|
Liu X, Clemens DL, Lee BY, Yang X, Zhou ZH, Horwitz MA. Atomic Structure of IglD Demonstrates Its Role as a Component of the Baseplate Complex of the Francisella Type VI Secretion System. mBio 2022; 13:e0127722. [PMID: 36036641 PMCID: PMC9600919 DOI: 10.1128/mbio.01277-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/25/2022] [Indexed: 11/29/2022] Open
Abstract
Francisella tularensis, a Tier 1 select agent of bioterrorism, contains a type VI secretion system (T6SS) encoded within the Francisella pathogenicity island (FPI), which is critical for its pathogenesis. Among the 18 proteins encoded by FPI is IglD, which is essential to Francisella's intracellular growth and virulence, but neither its location within T6SS nor its functional role has been established. Here, we present the cryoEM structure of IglD from Francisella novicida and show that the Francisella IglD forms a homotrimer that is structurally homologous to the T6SS baseplate protein TssK in Escherichia coli. Each IglD monomer consists of an N-terminal β-sandwich domain, a 4-helix bundle domain, and a flexible C-terminal domain. While the overall folds of IglD and TssK are similar, the two structures differ in three aspects: the relative orientation between their β-sandwich and the 4-helix bundle domains; two insertion loops present in TssK's β-sandwich domain; and, consequently, a lack of subunit-subunit interaction between insertion loops in the IglD trimer. Phylogenetic analysis indicates that IglD is genetically remote from the TssK orthologs in other T6SSs. While the other components of the Francisella baseplate are unknown, we conducted pulldown assays showing IglJ interacts with IglD and IglH, pointing to a model wherein IglD, IglH, and IglJ form the baseplate of the Francisella T6SS. Alanine substitution mutagenesis further established that IglD's hydrophobic pocket in the N-terminal β-sandwich domain interacts with two loops of IglJ, reminiscent of the TssK-TssG interaction. These results form a framework for understanding the hitherto unexplored Francisella T6SS baseplate. IMPORTANCE Francisella tularensis is a facultatively intracellular Gram-negative bacterium that causes the serious and potentially fatal zoonotic illness, tularemia. Because of its extraordinarily high infectivity and mortality to humans, especially when inhaled, F. tularensis is considered a potential bioterrorism agent and is classified as a Tier 1 select agent. The type VI secretion system (T6SS) encoded within the Francisella pathogenicity island (FPI) is critical to its pathogenesis, but its baseplate components are largely unknown. Here, we report the cryoEM structure of IglD from Francisella novicida and demonstrate its role as a component of the baseplate complex of the Francisella T6SS. We further show that IglD interacts with IglJ and IglH, and propose a model in which these proteins interact to form the Francisella T6SS baseplate. Elucidation of the structure and composition of the Francisella baseplate should facilitate the design of strategies to prevent and treat infections caused by F. tularensis.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- The California NanoSystems Institute (CNSI), UCLA, Los Angeles, California, USA
| | | | - Bai-Yu Lee
- Department of Medicine, UCLA, Los Angeles, California, USA
| | - Xue Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- The California NanoSystems Institute (CNSI), UCLA, Los Angeles, California, USA
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Z. Hong Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- The California NanoSystems Institute (CNSI), UCLA, Los Angeles, California, USA
| | - Marcus A. Horwitz
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- Department of Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
17
|
Wu Y, Mahtal N, Paillares E, Swistak L, Sagadiev S, Acharya M, Demeret C, Werf SVD, Guivel-Benhassine F, Schwartz O, Petracchini S, Mettouchi A, Caramelle L, Couvineau P, Thai R, Barbe P, Keck M, Brodin P, Machelart A, Sencio V, Trottein F, Sachse M, Chicanne G, Payrastre B, Ville F, Kreis V, Popoff MR, Johannes L, Cintrat JC, Barbier J, Gillet D, Lemichez E. C910 chemical compound inhibits the traffiking of several bacterial AB toxins with cross-protection against influenza virus. iScience 2022; 25:104537. [PMID: 35769882 PMCID: PMC9234246 DOI: 10.1016/j.isci.2022.104537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/20/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022] Open
Abstract
The development of anti-infectives against a large range of AB-like toxin-producing bacteria includes the identification of compounds disrupting toxin transport through both the endolysosomal and retrograde pathways. Here, we performed a high-throughput screening of compounds blocking Rac1 proteasomal degradation triggered by the Cytotoxic Necrotizing Factor-1 (CNF1) toxin, which was followed by orthogonal screens against two toxins that hijack the endolysosomal (diphtheria toxin) or retrograde (Shiga-like toxin 1) pathways to intoxicate cells. This led to the identification of the molecule C910 that induces the enlargement of EEA1-positive early endosomes associated with sorting defects of CNF1 and Shiga toxins to their trafficking pathways. C910 protects cells against eight bacterial AB toxins and the CNF1-mediated pathogenic Escherichia coli invasion. Interestingly, C910 reduces influenza A H1N1 and SARS-CoV-2 viral infection in vitro. Moreover, parenteral administration of C910 to mice resulted in its accumulation in lung tissues and a reduction in lethal influenza infection.
Collapse
Affiliation(s)
- Yu Wu
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
| | - Nassim Mahtal
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SCBM, 91191 Gif-sur-Yvette, France
| | - Eléa Paillares
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
- Université Paris Cité, 75006 Paris, France
| | - Léa Swistak
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
- Université Paris Cité, 75006 Paris, France
| | - Sara Sagadiev
- Seattle Children’s Research Institute, Jack R MacDonald Building, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Mridu Acharya
- Seattle Children’s Research Institute, Jack R MacDonald Building, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Caroline Demeret
- Unité Génétique Moléculaire des Virus à ARN, UMR 3569 CNRS, Université de Paris, Département de Virologie, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Sylvie Van Der Werf
- Unité Génétique Moléculaire des Virus à ARN, UMR 3569 CNRS, Université de Paris, Département de Virologie, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Florence Guivel-Benhassine
- Unité virus et immunité, Département de Virologie, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Olivier Schwartz
- Unité virus et immunité, Département de Virologie, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Serena Petracchini
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
- Université Paris Cité, 75006 Paris, France
| | - Amel Mettouchi
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
| | - Lucie Caramelle
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Pierre Couvineau
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Robert Thai
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Peggy Barbe
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Mathilde Keck
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Priscille Brodin
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 9017, University of Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Arnaud Machelart
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 9017, University of Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Valentin Sencio
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 9017, University of Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - François Trottein
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 9017, University of Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Martin Sachse
- Unité Technologie et service BioImagerie Ultrastructurale, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Gaëtan Chicanne
- Inserm, UMR1297 and Université Toulouse III Paul Sabatier, I2MC, 31024 Toulouse, France
| | - Bernard Payrastre
- Inserm, UMR1297 and Université Toulouse III Paul Sabatier, I2MC, 31024 Toulouse, France
| | - Florian Ville
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SCBM, 91191 Gif-sur-Yvette, France
| | - Victor Kreis
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Michel-Robert Popoff
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
| | - Ludger Johannes
- Institut Curie, PSL Research University, Cellular and Chemical Biology unit, Endocytic Trafficking and Intracellular Delivery team, U1143 INSERM, UMR3666 CNRS, 26 rue d'Ulm, 75248 Paris, France
| | - Jean-Christophe Cintrat
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SCBM, 91191 Gif-sur-Yvette, France
| | - Julien Barbier
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Daniel Gillet
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Emmanuel Lemichez
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
| |
Collapse
|
18
|
Abstract
![]()
The paradigm of antivirulence
therapy dictates that bacterial pathogens
are specifically disarmed but not killed by neutralizing their virulence
factors. Clearance of the invading pathogen by the immune system is
promoted. As compared to antibiotics, the pathogen-selective antivirulence
drugs hold promise to minimize collateral damage to the beneficial
microbiome. Also, selective pressure for resistance is expected to
be lower because bacterial viability is not directly affected. Antivirulence
drugs are being developed for stand-alone prophylactic and therapeutic
treatments but also for combinatorial use with antibiotics. This Review
focuses on drug modalities that target bacterial exotoxins after the
secretion or release-upon-lysis. Exotoxins have a significant and
sometimes the primary role as the disease-causing virulence factor,
and thereby they are attractive targets for drug development. We describe
the key pre-clinical and clinical trial data that have led to the
approval of currently used exotoxin-targeted drugs, namely the monoclonal
antibodies bezlotoxumab (toxin B/TcdB, Clostridioides difficile), raxibacumab (anthrax toxin, Bacillus anthracis), and obiltoxaximab (anthrax toxin, Bacillus anthracis), but also to challenges with some of the promising leads. We also
highlight the recent developments in pre-clinical research sector
to develop exotoxin-targeted drug modalities, i.e., monoclonal antibodies,
antibody fragments, antibody mimetics, receptor analogs, neutralizing
scaffolds, dominant-negative mutants, and small molecules. We describe
how these exotoxin-targeted drug modalities work with high-resolution
structural knowledge and highlight their advantages and disadvantages
as antibiotic alternatives.
Collapse
Affiliation(s)
- Moona Sakari
- Institute of Biomedicine, Research Unit for Infection and Immunity, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Arttu Laisi
- Institute of Biomedicine, Research Unit for Infection and Immunity, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Arto T. Pulliainen
- Institute of Biomedicine, Research Unit for Infection and Immunity, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| |
Collapse
|
19
|
Zoledronic acid-loaded cationic methylcellulose polyplex nanoparticles for enhanced gene delivery efficiency and breast cancer cell killing effect. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-021-02127-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Hyeon B, Nguyen MK, Do Heo W. Optogenetic Control of Membrane Trafficking Using Light-Activated Reversible Inhibition by Assembly Trap of Intracellular Membranes (IM-LARIAT). Methods Mol Biol 2022; 2473:309-331. [PMID: 35819773 DOI: 10.1007/978-1-0716-2209-4_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Intracellular membrane trafficking is a dynamic and complex cellular process. To study membrane trafficking with a high spatiotemporal resolution, we present an optogenetic method based on a blue-light inducible oligomerization of Rab GTPases, termed light-activated reversible inhibition by assembly trap of intracellular membranes (IM-LARIAT). In this chapter, we focus on the optical disruption of the dynamics and functions of previously studied intracellular membrane trafficking events, including transferrin recycling and growth cone regulation in relation to specific Rab GTPases. To aid general application, we provide a detailed description of transfection, imaging with a confocal microscope, and analysis of data.
Collapse
Affiliation(s)
- Bobae Hyeon
- Department of Life Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Mai Khanh Nguyen
- Abcam Fremont Technology Development Custom Solution, Fremont, CA, USA
| | - Won Do Heo
- Department of Life Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.
| |
Collapse
|
21
|
Zhao H, Yuen KY. Broad-spectrum Respiratory Virus Entry Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:137-153. [DOI: 10.1007/978-981-16-8702-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
22
|
Loss of E-Cadherin Leads to Druggable Vulnerabilities in Sphingolipid Metabolism and Vesicle Trafficking. Cancers (Basel) 2021; 14:cancers14010102. [PMID: 35008266 PMCID: PMC8749886 DOI: 10.3390/cancers14010102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/23/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Germline loss of the CDH1 gene is the primary genetic basis for hereditary diffuse gastric cancer, a disease resulting in elevated risk of both diffuse gastric cancer and lobular breast cancer. Current preventative treatment consists of prophylactic total gastrectomy, a therapy with several associated long-term morbidities. To address the lack of targeted molecular therapies for hereditary diffuse gastric cancer, we have utilized a synthetic lethal approach to identify candidate compounds that can specifically kill CDH1-null cells. Inhibitors of sphingolipid metabolism and vesicle trafficking pathways were identified as promising candidate compounds in a cell line model of CDH1 loss, then further validated in murine-derived organoid models of hereditary diffuse gastric cancer. With further research, these findings may lead to the development of novel chemoprevention strategies for the treatment of hereditary diffuse gastric cancer. Abstract Germline inactivating variants of CDH1 are causative of hereditary diffuse gastric cancer (HDGC), a cancer syndrome characterized by an increased risk of both diffuse gastric cancer and lobular breast cancer. Because loss of function mutations are difficult to target therapeutically, we have taken a synthetic lethal approach to identify targetable vulnerabilities in CDH1-null cells. We have previously observed that CDH1-null MCF10A cells exhibit a reduced rate of endocytosis relative to wildtype MCF10A cells. To determine whether this deficiency is associated with wider vulnerabilities in vesicle trafficking, we screened isogenic MCF10A cell lines with known inhibitors of autophagy, endocytosis, and sphingolipid metabolism. Relative to wildtype MCF10A cells, CDH1−/− MCF10A cells showed significantly greater sensitivity to several drugs targeting these processes, including the autophagy inhibitor chloroquine, the endocytosis inhibitors chlorpromazine and PP1, and the sphingosine kinase 1 inhibitor PF-543. Synthetic lethality was confirmed in both gastric and mammary organoid models of CDH1 loss, derived from CD44-Cre/Cdh1fl/fl/tdTomato mice. Collectively, these results suggest that both sphingolipid metabolism and vesicle trafficking represent previously unrecognised druggable vulnerabilities in CDH1-null cells and may lead to the development of new therapies for HDGC.
Collapse
|
23
|
Zanetti G, Mattarei A, Lista F, Rossetto O, Montecucco C, Pirazzini M. Novel Small Molecule Inhibitors That Prevent the Neuroparalysis of Tetanus Neurotoxin. Pharmaceuticals (Basel) 2021; 14:ph14111134. [PMID: 34832916 PMCID: PMC8618345 DOI: 10.3390/ph14111134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/25/2021] [Accepted: 11/04/2021] [Indexed: 01/22/2023] Open
Abstract
Tetanus neurotoxin (TeNT) is a protein exotoxin produced by Clostridium tetani that causes the deadly spastic neuroparalysis of tetanus. It consists of a metalloprotease light chain and of a heavy chain linked via a disulphide bond. TeNT binds to the neuromuscular junction (NMJ) and it is retro-axonally transported into vesicular compartments to the spinal cord, where it is released and taken up by inhibitory interneuron. Therein, the catalytic subunit is translocated into the cytoplasm where it cleaves its target protein VAMP-1/2 with consequent blockage of the release of inhibitory neurotransmitters. Vaccination with formaldehyde inactivated TeNT prevents the disease, but tetanus is still present in countries where vaccination coverage is partial. Here, we show that small molecule inhibitors interfering with TeNT trafficking or with the reduction of the interchain disulphide bond block the activity of the toxin in neuronal cultures and attenuate tetanus symptoms in vivo. These findings are relevant for the development of therapeutics against tetanus based on the inhibition of toxin molecules that are being retro-transported to or are already within the spinal cord and are, thus, not accessible to anti-TeNT immunoglobulins.
Collapse
Affiliation(s)
- Giulia Zanetti
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (G.Z.); (O.R.)
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Florigio Lista
- Scientific Department, Army Medical Center, Via Santo Stefano Rotondo 4, 00184 Rome, Italy;
| | - Ornella Rossetto
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (G.Z.); (O.R.)
- Italian Research Council, Institute of Neuroscience, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (G.Z.); (O.R.)
- Italian Research Council, Institute of Neuroscience, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy
- Correspondence: (C.M.); (M.P.)
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (G.Z.); (O.R.)
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy
- Correspondence: (C.M.); (M.P.)
| |
Collapse
|
24
|
Majumder P, Singh A, Wang Z, Dutta K, Pahwa R, Liang C, Andrews C, Patel NL, Shi J, de Val N, Walsh STR, Jeon AB, Karim B, Hoang CD, Schneider JP. Surface-fill hydrogel attenuates the oncogenic signature of complex anatomical surface cancer in a single application. NATURE NANOTECHNOLOGY 2021; 16:1251-1259. [PMID: 34556833 PMCID: PMC8595541 DOI: 10.1038/s41565-021-00961-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/19/2021] [Indexed: 05/06/2023]
Abstract
Tumours growing in a sheet-like manner on the surface of organs and tissues with complex topologies represent a difficult-to-treat clinical scenario. Their complete surgical resection is difficult due to the complicated anatomy of the diseased tissue. Residual cancer often responds poorly to systemic therapy and locoregional treatment is hindered by the limited accessibility to microscopic tumour foci. Here we engineered a peptide-based surface-fill hydrogel (SFH) that can be syringe- or spray-delivered to surface cancers during surgery or used as a primary therapy. Once applied, SFH can shape change in response to alterations in tissue morphology that may occur during surgery. Implanted SFH releases nanoparticles composed of microRNA and intrinsically disordered peptides that enter cancer cells attenuating their oncogenic signature. With a single application, SFH shows efficacy in four preclinical models of mesothelioma, demonstrating the therapeutic impact of the local application of tumour-specific microRNA, which might change the treatment paradigm for mesothelioma and possibly other surface cancers.
Collapse
Affiliation(s)
- Poulami Majumder
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Anand Singh
- Thoracic Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ziqiu Wang
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Frederick, MD, USA
| | - Kingshuk Dutta
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, USA
| | - Roma Pahwa
- Urology Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chen Liang
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Caroline Andrews
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Nimit L Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD, USA
| | - Junfeng Shi
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Natalia de Val
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Frederick, MD, USA
- Materials and Structural Analysis Division, Thermo Fisher Scientific, Hillsboro, OR, USA
| | - Scott T R Walsh
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Albert Byungyun Jeon
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Chuong D Hoang
- Thoracic Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Joel P Schneider
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| |
Collapse
|
25
|
Haywood EE, Handy NB, Lopez JW, Ho M, Wilson BA. Insertion-trigger residues differentially modulate endosomal escape by cytotoxic necrotizing factor toxins. J Biol Chem 2021; 297:101347. [PMID: 34715130 PMCID: PMC8592880 DOI: 10.1016/j.jbc.2021.101347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 01/20/2023] Open
Abstract
The cellular specificity, potency, and modular nature of bacterial protein toxins enable their application for targeted cytosolic delivery of therapeutic cargo. Efficient endosomal escape is a critical step in the design of bacterial toxin-inspired drug delivery (BTIDD) vehicles to avoid lysosomal degradation and promote optimal cargo delivery. The cytotoxic necrotizing factor (CNF) family of modular toxins represents a useful model for investigating cargo-delivery mechanisms due to the availability of many homologs with high sequence identity, their flexibility in swapping domains, and their differential activity profiles. Previously, we found that CNFy is more sensitive to endosomal acidification inhibitors than CNF1 and CNF2. Here, we report that CNF3 is even less sensitive than CNF1/2. We identified two amino acid residues within the putative translocation domain (E374 and E412 in CNFy, Q373 and S411 in CNF3) that differentiate between these two toxins. Swapping these corresponding residues in each toxin changed the sensitivity to endosomal acidification and efficiency of cargo-delivery to be more similar to the other toxin. Results suggested that trafficking to the more acidic late endosome is required for cargo delivery by CNFy but not CNF3. This model was supported by results from toxin treatment of cells in the presence of NH4Cl, which blocks endosomal acidification, and of small-molecule inhibitors EGA, which blocks trafficking to late endosomes, and ABMA, which blocks endosomal escape and trafficking to the lysosomal degradative pathway. These findings suggest that it is possible to fine-tune endosomal escape and cytosolic cargo delivery efficiency in designing BTIDD platforms.
Collapse
Affiliation(s)
- Elizabeth E Haywood
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Nicholas B Handy
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - James W Lopez
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Mengfei Ho
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Brenda A Wilson
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.
| |
Collapse
|
26
|
Robb Huhn G, Torres-Mangual N, Clore J, Cilenti L, Frisan T, Teter K. Endocytosis of the CdtA subunit from the Haemophilus ducreyi cytolethal distending toxin. Cell Microbiol 2021; 23:e13380. [PMID: 34292647 DOI: 10.1111/cmi.13380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/30/2022]
Abstract
Many Gram-negative pathogens produce a cytolethal distending toxin (CDT) with two cell-binding subunits (CdtA + CdtC) and a catalytic CdtB subunit. After adhesion to the plasma membrane of a target cell, CDT moves by retrograde transport to endoplasmic reticulum. CdtB then enters the nucleus where it generates DNA breaks that lead to cell cycle arrest and apoptosis or senescence. CdtA anchors the CDT holotoxin to the plasma membrane and is thought to remain on the cell surface after endocytosis of the CdtB/CdtC heterodimer. Here, we re-examined the potential endocytosis and intracellular transport of CdtA from the Haemophilus ducreyi CDT. We recorded the endocytosis of holotoxin-associated CdtA with a cell-based enzyme-linked immunoabsorbent assay (CELISA) and visualised its presence in the early endosomes by confocal microscopy 10 min after CDT binding to the cell surface. Western blot analysis documented the rapid degradation of internalised CdtA. Most of internalised CdtB and CdtC were degraded as well. The rapid rate of CDT internalisation and turnover, which could explain why CdtA endocytosis was not detected in previous studies, suggests only a minor pool of cell-associated CdtB reaches the nucleus. Our work demonstrates that CDT is internalised as an intact holotoxin and identifies the endosomes as the site of CdtA dissociation from CdtB/CdtC. TAKE AWAYS: During the endocytosis of CDT, CdtA is thought to remain at the cell surface. A cell-based ELISA documented the rapid endocytosis of CdtA. CdtA was visualised in the early endosomes by confocal microscopy. Intracellular CdtA was rapidly degraded, along with most of CdtB and CdtC.
Collapse
Affiliation(s)
- G Robb Huhn
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Naly Torres-Mangual
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA.,Colorado State University, Fort Collins, CO, USA
| | - John Clore
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Lucia Cilenti
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Teresa Frisan
- Department of Molecular Biology, Umeå University, Umeå, Sweden.,Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Ken Teter
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
27
|
Yu R, Shen X, Liu M, Liu X, Yin Z, Li X, Feng W, Hu J, Zhang H, Zheng X, Wang P, Zhang Z. The rice blast fungus MoRgs1 functioning in cAMP signaling and pathogenicity is regulated by casein kinase MoCk2 phosphorylation and modulated by membrane protein MoEmc2. PLoS Pathog 2021; 17:e1009657. [PMID: 34133468 PMCID: PMC8208561 DOI: 10.1371/journal.ppat.1009657] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/19/2021] [Indexed: 12/31/2022] Open
Abstract
GTP-binding protein (G-protein) and regulator of G-protein signaling (RGS) mediated signal transduction are critical in the growth and virulence of the rice blast pathogen Magnaporthe oryzae. We have previously reported that there are eight RGS and RGS-like proteins named MoRgs1 to MoRgs8 playing distinct and shared regulatory functions in M. oryzae and that MoRgs1 has a more prominent role compared to others in the fungus. To further explore the unique regulatory mechanism of MoRgs1, we screened a M. oryzae cDNA library for genes encoding MoRgs1-interacting proteins and identified MoCkb2, one of the two regulatory subunits of the casein kinase (CK) 2 MoCk2. We found that MoCkb2 and the sole catalytic subunit MoCka1 are required for the phosphorylation of MoRgs1 at the plasma membrane (PM) and late endosome (LE). We further found that an endoplasmic reticulum (ER) membrane protein complex (EMC) subunit, MoEmc2, modulates the phosphorylation of MoRgs1 by MoCk2. Interestingly, this phosphorylation is also essential for the GTPase-activating protein (GAP) function of MoRgs1. The balance among MoRgs1, MoCk2, and MoEmc2 ensures normal operation of the G-protein MoMagA-cAMP signaling required for appressorium formation and pathogenicity of the fungus. This has been the first report that an EMC subunit is directly linked to G-protein signaling through modulation of an RGS-casein kinase interaction. G-proteins play a significant role in signal perception and transduction during pathogen and host interactions. In the rice blast fungus M. oryzae, previous studies demonstrated that G-protein/cAMP signaling are important for appressorium formation and pathogenicity. One of the eight regulator of G-protein signaling (RGS) and RGS-like proteins, MoRgs1, targets G-protein MoMagA to regulate cAMP levels and growth and virulence of the fungus; however, how MoRgs1 exhibits this function and its own regulation indifferent from other RGS and RGS-like proteins are not clear. We here demonstrated that MoRgs1 is subject to regulation by the casein kinase 2 MoCk2 through protein phosphorylation, and this regulation is also essential for the GTPase-activating protein (GAP) function of MoRgs1. We also showed that the endoplasmic reticulum (ER) membrane complex (EMC) subunit MoEmc2 modulates MoCk2-mediated MoRgs1 phosphorylation. Balanced interactions among MoRgs1, MoEmc2, and MoCk2 ensure normal appressorium formation and pathogenicity of M. oryzae.
Collapse
Affiliation(s)
- Rui Yu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xuetong Shen
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Muxing Liu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xinyu Liu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Ziyi Yin
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xiao Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Wanzhen Feng
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Jiexiong Hu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Haifeng Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xiaobo Zheng
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Ping Wang
- Departments of Microbiology, Immunology, and Parasitology, and Pediatrics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Zhengguang Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China, The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
28
|
Kali S, Jallet C, Azebi S, Cokelaer T, Da Fonseca JP, Wu Y, Barbier J, Cintrat JC, Gillet D, Tordo N. Broad spectrum compounds targeting early stages of rabies virus (RABV) infection. Antiviral Res 2021; 188:105016. [PMID: 33444703 DOI: 10.1016/j.antiviral.2021.105016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 12/16/2022]
Abstract
ABMA and its analogue DABMA are two molecules of the adamantane family known to perturbate the endosomal pathway and to inhibit cell infection of several RNA and DNA viruses. Their activity against Rabies Virus (RABV) infection has already been demonstrated in vitro. (Wu et al., 2017, 2019). Here, we describe in more details their mechanism of action by comparison to Arbidol (umifenovir) and Ribavirin, two broad spectrum antivirals against emerging viruses such as Lassa, Ebola, influenza and Hantaan viruses. ABMA and DABMA, delivered 2 h pre-infection, inhibit RABV infection in vitro with an EC50 of 7.8 μM and 14 μM, respectively. They act at post-entry, by causing RABV accumulation within the endosomal compartment and DABMA specifically diminishes the expression of the GTPase Rab7a controlling the fusion of early endosomes to late endosomes or lysosomes. This may suggest that ABMA and DABMA act at different stages of the late endosomal pathway as supported by their different profile of synergy/antagonism with the fusion inhibitor Arbidol. This difference is further confirmed by the RABV mutants induced by successive passages under increasing selective pressure showing a particular involvement of the viral G protein in the DABMA inhibition while ABMA inhibition induces less mutations dispersed in the M, G and L viral proteins. These results suggest new therapeutic perspectives against rabies.
Collapse
Affiliation(s)
- Sabrina Kali
- Unit Antiviral Strategies, Institut Pasteur, 75724, Paris, France; Institut Pasteur D'Algérie, Dely Ibrahim, Alger, Algeria
| | - Corinne Jallet
- Unit Antiviral Strategies, Institut Pasteur, 75724, Paris, France
| | - Saliha Azebi
- Unit Antiviral Strategies, Institut Pasteur, 75724, Paris, France; Plate-forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75724, Paris, France; Unit Viral Neuroimmunology, Institut Pasteur, 75724, Paris, France
| | - Thomas Cokelaer
- Plate-forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75724, Paris, France; Hub de Bioinformatique et Biostatistique, Institut Pasteur, USR 3756 CNRS, 75724, Paris, France
| | - Juliana Pipoli Da Fonseca
- Plate-forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75724, Paris, France
| | - Yu Wu
- Service D'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, 91191, Gif-sur-Yvette, France
| | - Julien Barbier
- Service D'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, 91191, Gif-sur-Yvette, France
| | - Jean-Christophe Cintrat
- Service de Chimie Bioorganique et Marquage (SCBM), CEA, Université Paris-Saclay, 91191, Gif-sur-Yvette, France
| | - Daniel Gillet
- Service D'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, 91191, Gif-sur-Yvette, France
| | - Noël Tordo
- Unit Antiviral Strategies, Institut Pasteur, 75724, Paris, France; Institut Pasteur de Guinée, BP 4416, Conakry, Guinea.
| |
Collapse
|
29
|
Exosome mimicry by a HAVCR1-NPC1 pathway of endosomal fusion mediates hepatitis A virus infection. Nat Microbiol 2020; 5:1096-1106. [PMID: 32541946 PMCID: PMC7483988 DOI: 10.1038/s41564-020-0740-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 05/12/2020] [Indexed: 12/11/2022]
Abstract
Cell-to-cell communication by exosomes controls normal and pathogenic processes1,2. Viruses can spread in exosomes and thereby avoid immune recognition3. While biogenesis, binding and uptake of exosomes are well characterized4,5, delivery of exosome cargo into the cytoplasm is poorly understood3. We report that the phosphatidylserine receptor HAVCR1 (refs. 6,7) and the cholesterol transporter NPC1 (ref. 8) participate in cargo delivery from exosomes of hepatitis A virus (HAV)-infected cells (exo-HAV) by clathrin-mediated endocytosis. Using CRISPR-Cas9 knockout technology, we show that these two lipid receptors, which interact in the late endosome9, are necessary for the membrane fusion and delivery of RNA from exo-HAV into the cytoplasm. The HAVCR1-NPC1 pathway, which Ebola virus exploits to infect cells9, mediates HAV infection by exo-HAV, which indicates that viral infection via this exosome mimicry mechanism does not require an envelope glycoprotein. The capsid-free viral RNA in the exosome lumen, but not the endosomal uncoating of HAV particles contained in the exosomes, is mainly responsible for exo-HAV infectivity as assessed by methylene blue inactivation of non-encapsidated RNA. In contrast to exo-HAV, infectivity of HAV particles is pH-independent and requires HAVCR1 or another as yet unidentified receptor(s) but not NPC1. Our findings show that envelope-glycoprotein-independent fusion mechanisms are shared by exosomes and viruses, and call for a reassessment of the role of envelope glycoproteins in infection.
Collapse
|
30
|
Pandit S, Roy S, Pillai J, Banerjee S. Formulation and Intracellular Trafficking of Lipid-Drug Conjugate Nanoparticles Containing a Hydrophilic Antitubercular Drug for Improved Intracellular Delivery to Human Macrophages. ACS OMEGA 2020; 5:4433-4448. [PMID: 32175491 PMCID: PMC7066568 DOI: 10.1021/acsomega.9b03523] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/20/2020] [Indexed: 06/10/2023]
Abstract
Isoniazid is an important first-line antitubercular drug used in the treatment of all major clinical manifestations of tuberculosis, including both pulmonary and cerebral diseases. However, it is associated with significant drawbacks due to its inherent hydrophilic nature, including poor gut permeability and an inability to cross the lipophilic blood-brain barrier, which, in turn, limit its clinical efficacy. We hypothesized that the addition of a hydrophobic moiety to this molecule would help overcome these limitations and improve its bioavailability in the bloodstream. Therefore, we designed a stable, covalently linked lipid-drug conjugate of isoniazid with a short lipid chain of stearoyl chloride. Further, lipid-drug conjugate nanoparticles were synthesized from the bulk lipid-drug conjugate by a cold high-pressure homogenization method enabled by the optimized use of aqueous surfactants. The nanoparticle formulation was characterized systematically using in vitro physicochemical analytical methods, including atomic force microscopy, transmission electron microscopy, differential scanning calorimetry, X-ray diffraction, attenuated total reflectance, particle size, ζ-potential, and drug release studies, and the mechanism of drug release kinetics. These investigations revealed that the lipid-drug conjugate nanoparticles were loaded with an appreciable amount of isoniazid conjugate (92.73 ± 6.31% w/w). The prepared lipid-drug conjugate nanoparticles displayed a uniform shape with a smooth surface having a particle size of 124.60 ± 5.56 nm. In vitro drug release studies showed sustained release up to 72 h in a phosphate-buffered solution at pH 7.4. The release profile fitted to various known models of release kinetics revealed that the Higuchi model of diffusion kinetics was the best-fitting model (R 2 = 0.9929). In addition, confocal studies showed efficient uptake of lipid-drug conjugate nanoparticles by THP-1 macrophages presumably because of increased lipophilicity and anionic surface charge. This was followed by progressive intracellular trafficking into endosomal and lysosomal vesicles and colocalization with intravesicular compartmental proteins associated with mycobacterium tuberculosis pathogenesis, including CD63, LAMP-2, EEA1, and Rab11. The developed lipid-drug conjugate nanoparticles, therefore, displayed significant ability to improve the intracellular delivery of a highly water-soluble drug such as isoniazid.
Collapse
Affiliation(s)
- Sayantan Pandit
- Implants,
Devices & Drug Delivery Systems (ID3S) Laboratory,
Centre for Biodesign & Diagnostics (CBD), Translational Health Science & Technology Institute (THSTI), Faridabad, Haryana 121001, India
| | - Subhadeep Roy
- Implants,
Devices & Drug Delivery Systems (ID3S) Laboratory,
Centre for Biodesign & Diagnostics (CBD), Translational Health Science & Technology Institute (THSTI), Faridabad, Haryana 121001, India
- Department
of Pharmaceutical Sciences, School of Bio-Sciences & Bio-Technology, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh 226025, India
| | - Jonathan Pillai
- Implants,
Devices & Drug Delivery Systems (ID3S) Laboratory,
Centre for Biodesign & Diagnostics (CBD), Translational Health Science & Technology Institute (THSTI), Faridabad, Haryana 121001, India
| | - Subham Banerjee
- Implants,
Devices & Drug Delivery Systems (ID3S) Laboratory,
Centre for Biodesign & Diagnostics (CBD), Translational Health Science & Technology Institute (THSTI), Faridabad, Haryana 121001, India
- Department
of Pharmaceutics, National Institute of
Pharmaceutical Education & Research (NIPER), Guwahati, Assam 781125, India
| |
Collapse
|
31
|
Wu Y, Boulogne C, Carle S, Podinovskaia M, Barth H, Spang A, Cintrat J, Gillet D, Barbier J. Regulation of endo‐lysosomal pathway and autophagic flux by broad‐spectrum antipathogen inhibitor ABMA. FEBS J 2020; 287:3184-3199. [DOI: 10.1111/febs.15201] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/10/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Yu Wu
- Université Paris‐Saclay CEAINRAE Médicaments et Technologies pour la Santé (MTS) SIMoS Gif‐sur‐Yvette91191France
| | - Claire Boulogne
- IMAGERIE‐GIF Institute for Integrative Biology of the Cell (I2BC) CEA CNRS Université Paris‐Sud Université Paris‐Saclay Gif‐sur‐Yvette France
| | - Stefan Carle
- Institute of Pharmacology and Toxicology University of Ulm Medical Center Germany
| | | | - Holger Barth
- Institute of Pharmacology and Toxicology University of Ulm Medical Center Germany
| | - Anne Spang
- Growth and Development Biozentrum University of Basel Switzerland
| | - Jean‐Christophe Cintrat
- Université Paris‐Saclay CEA INRAE Médicaments et Technologies pour la Santé (MTS) SCBM Gif‐sur‐Yvette91191France
| | - Daniel Gillet
- Université Paris‐Saclay CEAINRAE Médicaments et Technologies pour la Santé (MTS) SIMoS Gif‐sur‐Yvette91191France
| | - Julien Barbier
- Université Paris‐Saclay CEAINRAE Médicaments et Technologies pour la Santé (MTS) SIMoS Gif‐sur‐Yvette91191France
| |
Collapse
|
32
|
Martinez-Jaramillo C, Trujillo-Vargas CM. Dissecting the localization of lipopolysaccharide-responsive and beige-like anchor protein (LRBA) in the endomembrane system. Allergol Immunopathol (Madr) 2020; 48:8-17. [PMID: 31883622 DOI: 10.1016/j.aller.2019.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION AND OBJECTIVES LRBA deficiency is caused by loss of LRBA protein expression, due to either homozygous or compounds heterozygous mutations in LRBA. LRBA deficiency has been shown to affect vesicular trafficking and autophagy. To date, LRBA has been observed in the cytosol, Golgi apparatus and some lysosomes in LPS-stimulated murine macrophages. The objectives of the present study were to study the LRBA localization in organelles involved in vesicular traffic, phagocytosis, and autophagy in mononuclear phagocytes (MP). MATERIALS AND METHODS We analyzed LRBA colocalization with different endosomes markets using confocal microscopy in MP. We used the autophagy inhibitors to determine the role of LRBA in formation, maturation or degradation of the autophagosome. RESULTS LRBA intracellular trafficking depends on the activity of the GTPase ADP ribosylation factor-1 (ARF) in MP. LRBA was identified in early, late endosomes but did not colocalize strongly with lysosomal markers. Although LRBA appears not to be recruited during the phagocytic cargo uptake, it greatly colocalized with the microtubule-associated protein 1A/1B-light chain 3 (LC3) under a steady state and this decreased after the induction of autophagy flux. Although the use of inhibitors of lysosome fusion did not restore the LRBA/LC3 colocalization, inhibitors of either early to late endosomes trafficking or PI3K pathway did. CONCLUSIONS Taken together, our results show that LRBA is located in endomembrane system vesicles, mainly in the early and late endosomes. Although LRBA appears not to be involved in the phagocytic uptake, it is recruited in the early steps of the autophagy flux.
Collapse
|
33
|
Takano T, Wakayama Y, Doki T. Endocytic Pathway of Feline Coronavirus for Cell Entry: Differences in Serotype-Dependent Viral Entry Pathway. Pathogens 2019; 8:pathogens8040300. [PMID: 31888266 PMCID: PMC6963708 DOI: 10.3390/pathogens8040300] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/15/2019] [Accepted: 12/15/2019] [Indexed: 12/30/2022] Open
Abstract
Feline coronavirus (FCoV) is a pathogen causing a lethal infectious disease in cats, feline infectious peritonitis. It has two serotypes (type I FCoV and type II FCoV). According to our previous study, type I FCoV infection is inhibited by compounds inducing intracellular cholesterol accumulation, whereas type II FCoV infection is not inhibited. Intracellular cholesterol accumulation was reported to disrupt late endosome function. Based on these findings, types I and II FCoV are considered to enter the cytosol through late and early endosomes, respectively. We investigated whether the antiviral activities of a late endosome trafficking inhibitor and cholesterol-accumulating agents are different between the FCoV serotypes. The late endosome trafficking inhibitor did not inhibit type II FCoV infection, but it inhibited type I FCoV infection. Type I FCoV infection was inhibited by cholesterol-accumulating triazoles, but not by non-cholesterol-accumulating triazoles. These phenomena were observed in both feline cell lines and feline primary macrophages. This study provides additional information on the differences in intracellular reproductive cycle between type I and type II FCoV.
Collapse
|
34
|
Wu Y, Pons V, Noël R, Kali S, Shtanko O, Davey RA, Popoff MR, Tordo N, Gillet D, Cintrat JC, Barbier J. DABMA: A Derivative of ABMA with Improved Broad-Spectrum Inhibitory Activity of Toxins and Viruses. ACS Med Chem Lett 2019; 10:1140-1147. [PMID: 31413797 DOI: 10.1021/acsmedchemlett.9b00155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023] Open
Abstract
The small molecule ABMA has been previously shown to protect cells against multiple toxins and pathogens including virus, intracellular bacteria, and parasite. Its mechanism of action is directly associated with host endolysosomal pathway rather than targeting toxin or pathogen itself. However, the relationship of its broad-spectrum anti-infection activity and chemical structure is not yet resolved. Here, we synthesized a series of derivatives and compared their activities against diphtheria toxin (DT). Dimethyl-ABMA (DABMA), one of the most potent analogs with about 20-fold improvement in protection efficacy against DT, was identified with a similar mechanism of action to ABMA. Moreover, DABMA exhibited enhanced efficacy against Clostridium difficile toxin B (TcdB), Clostridium sordellii lethal toxin (TcsL), Pseudomonas Exotoxin A (PE) as well as Rabies and Ebola viruses. The results revealed a structure-activity relationship of ABMA, which is a starting point for its clinical development as broad-spectrum drug against existing and emerging infectious diseases.
Collapse
Affiliation(s)
- Yu Wu
- Service d’Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université
Paris-Saclay, LabEx LERMIT, 91191 Gif-sur-Yvette, France
| | - Valérie Pons
- Service de Chimie Bio-organique et de Marquage (SCBM), CEA, Université Paris-Saclay, LabEx LERMIT, 91191 Gif-sur-Yvette, France
| | - Romain Noël
- Service de Chimie Bio-organique et de Marquage (SCBM), CEA, Université Paris-Saclay, LabEx LERMIT, 91191 Gif-sur-Yvette, France
| | - Sabrina Kali
- Antiviral Strategies Unit, Virology Department, Institut Pasteur, 75015 Paris, France
| | - Olena Shtanko
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas 78227, United States
| | - Robert A. Davey
- Department of Microbiology, NEIDL, Boston University, Boston, Massachusetts 02118, United States
| | - Michel R. Popoff
- Bactéries anaérobies et Toxines, Institut Pasteur, 75015 Paris, France
| | - Noël Tordo
- Antiviral Strategies Unit, Virology Department, Institut Pasteur, 75015 Paris, France
| | - Daniel Gillet
- Service d’Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université
Paris-Saclay, LabEx LERMIT, 91191 Gif-sur-Yvette, France
| | - Jean-Christophe Cintrat
- Service de Chimie Bio-organique et de Marquage (SCBM), CEA, Université Paris-Saclay, LabEx LERMIT, 91191 Gif-sur-Yvette, France
| | - Julien Barbier
- Service d’Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université
Paris-Saclay, LabEx LERMIT, 91191 Gif-sur-Yvette, France
| |
Collapse
|
35
|
Juliano RL, Wang L, Tavares F, Brown EG, James L, Ariyarathna Y, Ming X, Mao C, Suto M. Structure-activity relationships and cellular mechanism of action of small molecules that enhance the delivery of oligonucleotides. Nucleic Acids Res 2019; 46:1601-1613. [PMID: 29361039 PMCID: PMC5829638 DOI: 10.1093/nar/gkx1320] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/15/2018] [Indexed: 02/06/2023] Open
Abstract
The pharmacological effects of antisense and siRNA oligonucleotides are hindered by the tendency of these molecules to become entrapped in endomembrane compartments thus failing to reach their targets in the cytosol or nucleus. We have previously used high throughput screening to identify small molecules that enhance the escape of oligonucleotides from intracellular membrane compartments and have termed such molecules OECs (oligonucleotide enhancing compounds). Here, we report on the structure-activity relationships of a family of OECs that are analogs of a hit that emerged from our original screen. These studies demonstrate key roles for the lipophilic aromatic groups, the tertiary nitrogen, and the carbamate moiety of the parent compound. We have also investigated the intracellular site of action of the OECs and have shown that activity is due to the release of oligonucleotides from intermediate endosomal compartments rather than from early endosomes or from highly acidic downstream compartments. At high concentrations of OECs toxicity occurs in a manner that is independent of caspases or of lysosomal cathepsins but instead involves increased plasma membrane permeability. Thus, in addition to describing specific characteristics of this family of OECs, the current study provides insights into basic mechanisms of oligonucleotide trafficking and their implications for oligonucleotide delivery.
Collapse
Affiliation(s)
- Rudolph L Juliano
- Initos Pharmaceuticals LLC, Chapel Hill, NC 27599, USA.,UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Ling Wang
- Initos Pharmaceuticals LLC, Chapel Hill, NC 27599, USA
| | - Francis Tavares
- Chemogenics Biopharma, Research Triangle Park, NC 27709, USA
| | - Edward G Brown
- Chemogenics Biopharma, Research Triangle Park, NC 27709, USA
| | - Lindsey James
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | | | - Xin Ming
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Chengqiong Mao
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Mark Suto
- Southern Research Institute, Birmingham, AL 35205, USA
| |
Collapse
|
36
|
Banerjee S, Roy S, Bhaumik KN, Pillai J. Mechanisms of the effectiveness of lipid nanoparticle formulations loaded with anti-tubercular drugs combinations toward overcoming drug bioavailability in tuberculosis. J Drug Target 2019; 28:55-69. [DOI: 10.1080/1061186x.2019.1613409] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Subham Banerjee
- Centre for Biodesign & Diagnostics (CBD), Translational Health Science & Technology Institute (THSTI), Faridabad, India
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, India
| | - Subhadeep Roy
- Centre for Biodesign & Diagnostics (CBD), Translational Health Science & Technology Institute (THSTI), Faridabad, India
| | - Kaushik Nath Bhaumik
- Centre for Biodesign & Diagnostics (CBD), Translational Health Science & Technology Institute (THSTI), Faridabad, India
| | - Jonathan Pillai
- Centre for Biodesign & Diagnostics (CBD), Translational Health Science & Technology Institute (THSTI), Faridabad, India
| |
Collapse
|
37
|
Ozog S, Chen CX, Simpson E, Garijo O, Timberlake ND, Minder P, Verhoeyen E, Torbett BE. CD46 Null Packaging Cell Line Improves Measles Lentiviral Vector Production and Gene Delivery to Hematopoietic Stem and Progenitor Cells. Mol Ther Methods Clin Dev 2019; 13:27-39. [PMID: 30603655 PMCID: PMC6310745 DOI: 10.1016/j.omtm.2018.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022]
Abstract
Lentiviral vectors (LVs) pseudotyped with the measles virus hemagglutinin (H) and fusion (F) glycoproteins have been reported to more efficiently transduce hematopoietic stem and progenitor cells (HSPCs) compared with vesicular stomatitis virus glycoprotein (VSV-G) pseudotyped LVs. However, a limit to H/F LV use is the low titer of produced vector. Here we show that measles receptor (CD46) expression on H/F transfected HEK293T vector-producing cells caused adjacent cell membrane fusion, resulting in multinucleate syncytia formation and death prior to peak vector production, leading to contaminating cell membranes that co-purified with LV. H/F LVs produced in CD46 null HEK293T cells, generated by CRISPR/Cas9-mediated knockout of CD46, produced 2-fold higher titer vector compared with LVs produced in CD46+ HEK293T cells. This resulted in approximately 2- to 3-fold higher transduction of HSPCs while significantly reducing target cell cytotoxicity caused by producer cell contaminates. Improved H/F LV entry into HSPCs and distinct entry mechanisms compared with VSV-G LV were also observed by confocal microscopy. Given that vector production is a major source of cost and variability in clinical trials of gene therapy, we propose that the use of CD46 null packaging cells may help to address these challenges.
Collapse
Affiliation(s)
- Stosh Ozog
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Craig X. Chen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Bishops School, La Jolla, CA 92037, USA
| | - Elizabeth Simpson
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Olivia Garijo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nina D. Timberlake
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Petra Minder
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Els Verhoeyen
- CIRI–International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France
- Inserm, U1111; Ecole Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1; CNRS, UMR5308, Lyon, France
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | - Bruce E. Torbett
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
38
|
Glucosylceramidase Maintains Influenza Virus Infection by Regulating Endocytosis. J Virol 2019; 93:JVI.00017-19. [PMID: 30918081 PMCID: PMC6613767 DOI: 10.1128/jvi.00017-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/14/2019] [Indexed: 01/07/2023] Open
Abstract
Influenza virus is the pathogen responsible for the second largest pandemic in human history. A better understanding of how influenza virus enters host cells may lead to the development of more-efficacious therapies against emerging strains of the virus. Here we show that the glycosphingolipid metabolizing enzyme glucosylceramidase is required for optimal influenza virus trafficking to late endosomes and for consequent fusion, entry, and infection. We also provide evidence that promotion of influenza virus entry by glucosylceramidase extends to other endosome-entering viruses and is due to a general requirement for this enzyme, and hence for optimal levels of glucosylceramide, for efficient trafficking of endogenous cargos, such as the epidermal growth factor (EGF) receptor, along the endocytic pathway. This work therefore has implications for the basic process of endocytosis as well as for pathogenic processes, including virus entry and Gaucher disease. Influenza virus is an RNA virus encapsulated in a lipid bilayer derived from the host cell plasma membrane. Previous studies showed that influenza virus infection depends on cellular lipids, including the sphingolipids sphingomyelin and sphingosine. Here we examined the role of a third sphingolipid, glucosylceramide, in influenza virus infection following clustered regularly interspaced short palindromic repeats with Cas9 (CRISPR-Cas9)-mediated knockout (KO) of its metabolizing enzyme glucosylceramidase (GBA). After confirming GBA knockout of HEK 293 and A549 cells by both Western blotting and lipid mass spectrometry, we observed diminished infection in both KO cell lines by a PR8 (H1N1) green fluorescent protein (GFP) reporter virus. We further showed that the reduction in infection correlated with impaired influenza virus trafficking to late endosomes and hence with fusion and entry. To examine whether GBA is required for other enveloped viruses, we compared the results seen with entry mediated by the glycoproteins of Ebola virus, influenza virus, vesicular stomatitis virus (VSV), and measles virus in GBA knockout cells. Entry inhibition was relatively robust for Ebola virus and influenza virus, modest for VSV, and mild for measles virus, suggesting a greater role for viruses that enter cells by fusing with late endosomes. As the virus studies suggested a general role for GBA along the endocytic pathway, we tested that hypothesis and found that trafficking of epidermal growth factor (EGF) to late endosomes and degradation of its receptor were impaired in GBA knockout cells. Collectively, our findings suggest that GBA is critically important for endocytic trafficking of viruses as well as of cellular cargos, including growth factor receptors. Modulation of glucosylceramide levels may therefore represent a novel accompaniment to strategies to antagonize “late-penetrating” viruses, including influenza virus. IMPORTANCE Influenza virus is the pathogen responsible for the second largest pandemic in human history. A better understanding of how influenza virus enters host cells may lead to the development of more-efficacious therapies against emerging strains of the virus. Here we show that the glycosphingolipid metabolizing enzyme glucosylceramidase is required for optimal influenza virus trafficking to late endosomes and for consequent fusion, entry, and infection. We also provide evidence that promotion of influenza virus entry by glucosylceramidase extends to other endosome-entering viruses and is due to a general requirement for this enzyme, and hence for optimal levels of glucosylceramide, for efficient trafficking of endogenous cargos, such as the epidermal growth factor (EGF) receptor, along the endocytic pathway. This work therefore has implications for the basic process of endocytosis as well as for pathogenic processes, including virus entry and Gaucher disease.
Collapse
|
39
|
Li X, Zhong K, Yin Z, Hu J, Wang W, Li L, Zhang H, Zheng X, Wang P, Zhang Z. The seven transmembrane domain protein MoRgs7 functions in surface perception and undergoes coronin MoCrn1-dependent endocytosis in complex with Gα subunit MoMagA to promote cAMP signaling and appressorium formation in Magnaporthe oryzae. PLoS Pathog 2019; 15:e1007382. [PMID: 30802274 PMCID: PMC6405168 DOI: 10.1371/journal.ppat.1007382] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/07/2019] [Accepted: 01/29/2019] [Indexed: 01/09/2023] Open
Abstract
Regulator of G-protein signaling (RGS) proteins primarily function as GTPase-accelerating proteins (GAPs) to promote GTP hydrolysis of Gα subunits, thereby regulating G-protein mediated signal transduction. RGS proteins could also contain additional domains such as GoLoco to inhibit GDP dissociation. The rice blast fungus Magnaporthe oryzae encodes eight RGS and RGS-like proteins (MoRgs1 to MoRgs8) that have shared and distinct functions in growth, appressorium formation and pathogenicity. Interestingly, MoRgs7 and MoRgs8 contain a C-terminal seven-transmembrane domain (7-TM) motif typical of G-protein coupled receptor (GPCR) proteins, in addition to the conserved RGS domain. We found that MoRgs7, but not MoRgs8, couples with Gα MoMagA to undergo endocytic transport from the plasma membrane to the endosome upon sensing of surface hydrophobicity. We also found that MoRgs7 can interact with hydrophobic surfaces via a hydrophobic interaction, leading to the perception of environmental hydrophobiccues. Moreover, we found that MoRgs7-MoMagA endocytosis is regulated by actin patch-associated protein MoCrn1, linking it to cAMP signaling. Our studies provided evidence suggesting that MoRgs7 could also function in a GPCR-like manner to sense environmental signals and it, together with additional proteins of diverse functions, promotes cAMP signaling required for developmental processes underlying appressorium function and pathogenicity.
Collapse
Affiliation(s)
- Xiao Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China
| | - Kaili Zhong
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China
| | - Ziyi Yin
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China
| | - Jiexiong Hu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China
| | - Wenhao Wang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China
| | - Lianwei Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China
| | - Haifeng Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China
| | - Xiaobo Zheng
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China
| | - Ping Wang
- Departments of Pediatrics, and Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Zhengguang Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China
| |
Collapse
|
40
|
Hartmann S, Nusbaum DJ, Kim K, Alameh S, Ho CLC, Cruz RL, Levitin A, Bradley KA, Martchenko M. Role of a Small Molecule in the Modulation of Cell Death Signal Transduction Pathways. ACS Infect Dis 2018; 4:1746-1754. [PMID: 30354048 DOI: 10.1021/acsinfecdis.8b00231] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inflammasomes activate caspase-1 in response to molecular signals from pathogens and other dangerous stimuli as a part of the innate immune response. A previous study discovered a small-molecule, 4-fluoro- N'-[1-(2-pyridinyl)ethylidene]benzohydrazide, which we named DN1, that reduces the cytotoxicity of anthrax lethal toxin (LT). We determined that DN1 protected cells irrespectively of LT concentration and reduced the pathogenicity of an additional bacterial exotoxin and several viruses. Using the LT cytotoxicity pathway, we show that DN1 does not prevent LT internalization and catalytic activity or caspase-1 activation. Moreover, DN1 does not affect the proteolytic activity of host cathepsin B, which facilitates the cytoplasmic entry of toxins. PubChem Bioactivities lists two G protein-coupled receptors (GPCR), type-1 angiotensin II receptor and apelin receptor, as targets of DN1. The inhibition of phosphatidylinositol 3-kinase, phospholipase C, and protein kinase B, which are downstream of GPCR signaling, synergized with DN1 in protecting cells from LT. We hypothesize that DN1-mediated antagonism of GPCRs modulates signal transduction pathways to induce a cellular state that reduces LT-induced pyroptosis downstream of caspase-1 activation. DN1 also reduced the susceptibility of Drosophila melanogaster to toxin-associated bacterial infections. Future experiments will aim to further characterize how DN1 modulates signal transduction pathways to inhibit pyroptotic cell death in LT-sensitive macrophages. DN1 represents a novel chemical probe to investigate host cellular mechanisms that mediate cell death in response to pathogenic agents.
Collapse
Affiliation(s)
- Stella Hartmann
- School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Drive, Claremont, California 91711, United States
| | - David J. Nusbaum
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, 609 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Kevin Kim
- School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Drive, Claremont, California 91711, United States
| | - Saleem Alameh
- School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Drive, Claremont, California 91711, United States
| | - Chi-Lee C. Ho
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, 609 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Renae L. Cruz
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, 609 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Anastasia Levitin
- School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Drive, Claremont, California 91711, United States
| | - Kenneth A. Bradley
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, 609 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Mikhail Martchenko
- School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Drive, Claremont, California 91711, United States
| |
Collapse
|
41
|
Storm L, Bikker FJ, Nazmi K, Hulst AG, der Riet-Van Oeveren DV, Veerman ECI, Hays JP, Kaman WE. Anthrax protective antigen is a calcium-dependent serine protease. Virulence 2018; 9:1085-1091. [PMID: 30052476 PMCID: PMC6086315 DOI: 10.1080/21505594.2018.1486139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Bacillus anthracis secretes a three component exotoxin-complex, which contributes to anthrax pathogenesis. Formation of this complex starts with the binding of protective antigen (PA) to its cellular receptor. In this study, we report that PA is a calcium-dependent serine protease and that the protein potentially uses this proteolytic activity for receptor binding. Additionally our findings shed new light on previous research describing the inhibition of anthrax toxins and exotoxin formation. Importantly, inhibition of the proteolytic activity of protective antigen could be a novel therapeutic strategy in fighting B. anthracis-related infections.
Collapse
Affiliation(s)
- Lisanne Storm
- a Department of Medical Microbiology and Infectious Diseases , Erasmus University Medical Centre , Rotterdam , The Netherlands
| | - Floris J Bikker
- b Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam , University of Amsterdam and VU University Amsterdam , Amsterdam , The Netherlands
| | - Kamran Nazmi
- b Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam , University of Amsterdam and VU University Amsterdam , Amsterdam , The Netherlands
| | - Albert G Hulst
- c Department of CBRN Protection , Netherlands Organization for Applied Scientific Research TNO , Rijswijk , The Netherlands
| | - Debora V der Riet-Van Oeveren
- c Department of CBRN Protection , Netherlands Organization for Applied Scientific Research TNO , Rijswijk , The Netherlands
| | - Enno C I Veerman
- b Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam , University of Amsterdam and VU University Amsterdam , Amsterdam , The Netherlands
| | - John P Hays
- a Department of Medical Microbiology and Infectious Diseases , Erasmus University Medical Centre , Rotterdam , The Netherlands
| | - Wendy E Kaman
- a Department of Medical Microbiology and Infectious Diseases , Erasmus University Medical Centre , Rotterdam , The Netherlands.,b Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam , University of Amsterdam and VU University Amsterdam , Amsterdam , The Netherlands
| |
Collapse
|
42
|
Acquaye-Seedah E, Huang Y, Sutherland JN, DiVenere AM, Maynard JA. Humanised monoclonal antibodies neutralise pertussis toxin by receptor blockade and reduced retrograde trafficking. Cell Microbiol 2018; 20:e12948. [PMID: 30152075 PMCID: PMC6519169 DOI: 10.1111/cmi.12948] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 08/02/2018] [Accepted: 08/20/2018] [Indexed: 12/12/2022]
Abstract
Pertussis toxin (PTx) is a major protective antigen produced by Bordetella pertussis that is included in all current acellular vaccines. Of several well‐characterized monoclonal antibodies binding this toxin, the humanised hu1B7 and hu11E6 antibodies are highly protective in multiple in vitro and in vivo assays. In this study, we determine the molecular mechanisms of protection mediated by these antibodies. Neither antibody directly binds the B. pertussis bacterium nor supports antibody‐dependent complement cytotoxicity. Both antibodies, either individually or as a cocktail, form multivalent complexes with soluble PTx that bind the FcγRIIb receptor more tightly than antibody alone, suggesting that the antibodies may accelerate PTx clearance via immune complex formation. However, a receptor binding assay and cellular imaging indicate that the main mechanism used by hu11E6 is competitive inhibition of PTx binding to its cellular receptor. In contrast, the main hu1B7 neutralising mechanism appears to be inhibition of PTx internalisation and retrograde trafficking. We assessed the effects of hu1B7 on PTx retrograde trafficking in CHO‐K1 cells using quantitative immunofluorescence microscopy. In the absence of hu1B7 or after incubation with an isotype control antibody, PTx colocalizes to organelles in a manner consistent with retrograde transport. However, after preincubation with hu1B7, PTx appears restricted to the membrane surface with colocalization to organelles associated with retrograde transport significantly reduced. Together, these data support a model whereby hu11E6 and hu1B7 interfere with PTx receptor binding and PTx retrograde trafficking, respectively.
Collapse
Affiliation(s)
- Edith Acquaye-Seedah
- Department of Biochemistry, The University of Texas at Austin, Austin, Texas.,Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas
| | - Yimin Huang
- Department of Cell and Molecular Biology, The University of Texas at Austin, Austin, Texas.,Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas
| | - Jamie N Sutherland
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas
| | - Andrea M DiVenere
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas
| | - Jennifer A Maynard
- Department of Biochemistry, The University of Texas at Austin, Austin, Texas.,Department of Cell and Molecular Biology, The University of Texas at Austin, Austin, Texas.,Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
43
|
Hartmann S, Lopez Cruz R, Alameh S, Ho CLC, Rabideau A, Pentelute BL, Bradley KA, Martchenko M. Characterization of Novel Piperidine-Based Inhibitor of Cathepsin B-Dependent Bacterial Toxins and Viruses. ACS Infect Dis 2018; 4:1235-1245. [PMID: 29749721 DOI: 10.1021/acsinfecdis.8b00053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Exploiting the host endocytic trafficking pathway is a common mechanism by which bacterial exotoxins gain entry to exert virulent effects upon the host cells. A previous study identified a small-molecule, 1-(2,6-dimethyl-1-piperidinyl)-3-[(2-isopropyl-5-methylcyclohexyl)oxy]-2-propanol, that blocks the process of anthrax lethal toxin (LT) cytotoxicity. Here, we report the characterization of the bioactivity of this compound, which we named RC1. We found that RC1 protected host cells independently of LT concentration and also blocked intoxication by other bacterial exotoxins, suggesting that the target of the compound is a host factor. Using the anthrax LT intoxication pathway as a reference, we show that while anthrax toxin is able to bind to cells and establish an endosomal pore in the presence of the drug, the toxin is unable to translocate into the cytosol. We demonstrate that RC1 does not inhibit the toxin directly but rather reduces the enzymatic activity of host cathepsin B that mediates the escape of toxins into the cytoplasm from late endosomes. We demonstrate that the pathogenicity of Human cytomegalovirus and Herpes simplex virus 1, which relies on cathepsin B protease activity, is reduced by RC1. This study reveals the potential of RC1 as a broad-spectrum host-oriented therapy against several aggressive and deadly pathogens.
Collapse
Affiliation(s)
- Stella Hartmann
- School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Drive, Claremont, California 91711, United States
| | - Renae Lopez Cruz
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, 609 Charles E Young Drive East, Los Angeles, California 90095, United States
| | - Saleem Alameh
- School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Drive, Claremont, California 91711, United States
| | - Chi-Lee C. Ho
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, 609 Charles E Young Drive East, Los Angeles, California 90095, United States
| | - Amy Rabideau
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Bradley L. Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Kenneth A. Bradley
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, 609 Charles E Young Drive East, Los Angeles, California 90095, United States
| | - Mikhail Martchenko
- School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Drive, Claremont, California 91711, United States
| |
Collapse
|
44
|
Sampayo RG, Toscani AM, Rubashkin MG, Thi K, Masullo LA, Violi IL, Lakins JN, Cáceres A, Hines WC, Coluccio Leskow F, Stefani FD, Chialvo DR, Bissell MJ, Weaver VM, Simian M. Fibronectin rescues estrogen receptor α from lysosomal degradation in breast cancer cells. J Cell Biol 2018; 217:2777-2798. [PMID: 29980625 PMCID: PMC6080927 DOI: 10.1083/jcb.201703037] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 12/20/2017] [Accepted: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
Estrogen receptor α (ERα) is expressed in tissues as diverse as brains and mammary glands. In breast cancer, ERα is a key regulator of tumor progression. Therefore, understanding what activates ERα is critical for cancer treatment in particular and cell biology in general. Using biochemical approaches and superresolution microscopy, we show that estrogen drives membrane ERα into endosomes in breast cancer cells and that its fate is determined by the presence of fibronectin (FN) in the extracellular matrix; it is trafficked to lysosomes in the absence of FN and avoids the lysosomal compartment in its presence. In this context, FN prolongs ERα half-life and strengthens its transcriptional activity. We show that ERα is associated with β1-integrin at the membrane, and this integrin follows the same endocytosis and subcellular trafficking pathway triggered by estrogen. Moreover, ERα+ vesicles are present within human breast tissues, and colocalization with β1-integrin is detected primarily in tumors. Our work unravels a key, clinically relevant mechanism of microenvironmental regulation of ERα signaling.
Collapse
Affiliation(s)
- Rocío G Sampayo
- Universidad de Buenos Aires, Instituto de Oncología "Ángel H. Roffo", Área Investigación, Buenos Aires, Argentina .,Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Ciudad Universitaria, Buenos Aires, Argentina.,Universidad Nacional de San Martín, Instituto de Nanosistemas, Campus Miguelete, San Martín, Argentina
| | - Andrés M Toscani
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, IQUIBICEN UBA-CONICET y Universidad Nacional de Luján, Departamento de Ciencias Básicas, Buenos Aires, Argentina
| | - Matthew G Rubashkin
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Kate Thi
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Luciano A Masullo
- Centro de Investigaciones en Bionanociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ianina L Violi
- Centro de Investigaciones en Bionanociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Jonathon N Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Alfredo Cáceres
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, Córdoba, Argentina
| | - William C Hines
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Federico Coluccio Leskow
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, IQUIBICEN UBA-CONICET y Universidad Nacional de Luján, Departamento de Ciencias Básicas, Buenos Aires, Argentina
| | - Fernando D Stefani
- Centro de Investigaciones en Bionanociencias, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dante R Chialvo
- Center for Complex Systems and Brain Sciences, Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Tecnológicas, San Martín, Argentina
| | - Mina J Bissell
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Marina Simian
- Universidad de Buenos Aires, Instituto de Oncología "Ángel H. Roffo", Área Investigación, Buenos Aires, Argentina .,Universidad Nacional de San Martín, Instituto de Nanosistemas, Campus Miguelete, San Martín, Argentina
| |
Collapse
|
45
|
Maisel S, Broka D, Schroeder J. Intravesicular epidermal growth factor receptor subject to retrograde trafficking drives epidermal growth factor-dependent migration. Oncotarget 2017; 9:6463-6477. [PMID: 29464085 PMCID: PMC5814225 DOI: 10.18632/oncotarget.23766] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/23/2017] [Indexed: 12/31/2022] Open
Abstract
The Epidermal Growth Factor Receptor (EGFR) is frequently mutated and overexpressed in metastatic cancer. Although EGFR is a transmembrane tyrosine kinase localized to the basolateral membrane in normal epithelium, it is frequently found intracellularly localized in transformed cells. We have previously demonstrated the epithelial adaptor protein mucin 1 (MUC1) alters trafficking of EGFR, inhibiting its degradation and promoting its translocation to the nucleus, where it can directly modulate gene transcription. Here, we demonstrate that MUC1 promotes the retention of EGF-bound EGFR in Early Endosome Antigen1 (EEA1)-positive vesicles while preventing its trafficking to the lysosome. These events result in the accumulation of endosomal vesicles harboring active receptor throughout the cell and a reorganization of the actin cytoskeleton. EGF-dependent cell migration and filopodia formation is reliant upon this altered trafficking, and can be prevented by blocking retrograde trafficking. Together, these results indicate that intracellular EGFR may play an essential role in cancer metastasis and a potential mechanism for the failure of therapeutic antibodies in EGFR-driven metastatic breast cancer.
Collapse
Affiliation(s)
- Sabrina Maisel
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA.,Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Derrick Broka
- Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Joyce Schroeder
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA.,Arizona Cancer Center, University of Arizona, Tucson, AZ, USA.,Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA.,BIO5 Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
46
|
de la Puente P, Luderer MJ, Federico C, Jin A, Gilson RC, Egbulefu C, Alhallak K, Shah S, Muz B, Sun J, King J, Kohnen D, Salama NN, Achilefu S, Vij R, Azab AK. Enhancing proteasome-inhibitory activity and specificity of bortezomib by CD38 targeted nanoparticles in multiple myeloma. J Control Release 2017; 270:158-176. [PMID: 29196043 DOI: 10.1016/j.jconrel.2017.11.045] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/29/2017] [Accepted: 11/27/2017] [Indexed: 01/03/2023]
Abstract
The establishment of more effective treatments that can circumvent chemoresistance in Multiple Myeloma (MM) is a priority. Although bortezomib (BTZ) is one of the most potent proteasome inhibitors available, still possesses limitations related to dose limiting side effects. Several strategies have been developed to improve the delivery of chemotherapies to MM by targeting different moieties expressed on MM cells to nanoparticle delivery systems (NPs), which have failed mainly due to their heterogeneous expression on these cells. Our goal was to test CD38 targeted chitosan NPs as novel targeting moiety for MM to improve the potency and efficacy of BTZ in MM cells and reduce the side effects in healthy tissue. We have showed preferential BTZ release in tumor-microenvironment, specific binding to MM cells, and an improved drug cellular uptake through BTZ diffusion from the surface and endocytosed NPs, which translated in enhanced proteasome inhibition and robust cytotoxic effect on MM cells when BTZ was administered through anti-CD38 chitosan NPs. Furthermore, the anti-CD38 chitosan NPs specifically delivered therapeutic agents to MM cells improving therapeutic efficacy and reducing side effects in vivo. The anti-CD38 chitosan NPs showed low toxicity profile allowing enhancement of proteasome-inhibitory activity and specificity of BTZ by endocytosis-mediated uptake of CD38 representing a promising therapy in MM.
Collapse
Affiliation(s)
- Pilar de la Puente
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Micah J Luderer
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Cinzia Federico
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Abbey Jin
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, MO, USA
| | - Rebecca C Gilson
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, MO, USA
| | - Christopher Egbulefu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, MO, USA
| | - Kinan Alhallak
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Shruti Shah
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Barbara Muz
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Jennifer Sun
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Justin King
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, USA
| | - Daniel Kohnen
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, USA
| | - Noha Nabil Salama
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, MO, USA; Department of Pharmaceutics and Industrial Pharmacy, Cairo University Faculty of Pharmacy, Cairo, Egypt
| | - Samuel Achilefu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, MO, USA
| | - Ravi Vij
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA.
| |
Collapse
|
47
|
ABMA, a small molecule that inhibits intracellular toxins and pathogens by interfering with late endosomal compartments. Sci Rep 2017; 7:15567. [PMID: 29138439 PMCID: PMC5686106 DOI: 10.1038/s41598-017-15466-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/24/2017] [Indexed: 12/30/2022] Open
Abstract
Intracellular pathogenic microorganisms and toxins exploit host cell mechanisms to enter, exert their deleterious effects as well as hijack host nutrition for their development. A potential approach to treat multiple pathogen infections and that should not induce drug resistance is the use of small molecules that target host components. We identified the compound 1-adamantyl (5-bromo-2-methoxybenzyl) amine (ABMA) from a cell-based high throughput screening for its capacity to protect human cells and mice against ricin toxin without toxicity. This compound efficiently protects cells against various toxins and pathogens including viruses, intracellular bacteria and parasite. ABMA provokes Rab7-positive late endosomal compartment accumulation in mammalian cells without affecting other organelles (early endosomes, lysosomes, the Golgi apparatus, the endoplasmic reticulum or the nucleus). As the mechanism of action of ABMA is restricted to host-endosomal compartments, it reduces cell infection by pathogens that depend on this pathway to invade cells. ABMA may represent a novel class of broad-spectrum compounds with therapeutic potential against diverse severe infectious diseases.
Collapse
|
48
|
Pirazzini M, Azarnia Tehran D, Zanetti G, Rossetto O, Montecucco C. Hsp90 and Thioredoxin-Thioredoxin Reductase enable the catalytic activity of Clostridial neurotoxins inside nerve terminals. Toxicon 2017; 147:32-37. [PMID: 29111118 DOI: 10.1016/j.toxicon.2017.10.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/21/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022]
Abstract
Botulinum (BoNTs) and tetanus (TeNT) neurotoxins are the most toxic substances known and form the growing family of Clostridial neurotoxins (CNT), the etiologic agents of botulism and tetanus. CNT are composed of a metalloprotease light chain (L), linked via a disulfide bond to a heavy chain (H). H mediates the binding to nerve terminals and the membrane translocation of L into the cytosol, where its substrates, the three SNARE proteins, are localized. L translocation is accompanied by unfolding and, once delivered on the cytosolic side of the endosome membrane, it has to be reduced and reacquire the native fold to be active. The Thioredoxin-Thioredoxin Reductase system (Trx-TrxR) specifically reduces the interchain disulfide bond while the cytosolic chaperone protein Hsp90 mediates L refolding. Both steps are essential for CNT activity and their inhibition efficiently blocks the neurotoxicity in cultured neurons and mice. Trx and its reductase physically interact with Hsp90 and are loosely bound to the cytosolic side of synaptic vesicles, the organelle exploited by CNT to enter nerve terminals and wherefrom L is translocated into the cytosol. Therefore, Trx, TrxR and Hsp90 orchestrate a chaperone-redox molecular machinery that enables the catalytic activity of the L inside nerve terminals. Given the fundamental role of L reduction and refolding, this machinery represents a rational target for the development of mechanism-based antitoxins.
Collapse
Affiliation(s)
- Marco Pirazzini
- Dipartimento di Scienze Biomediche, Università di Padova, Via U. Bassi 58/B, 35121 Padova, Italy.
| | - Domenico Azarnia Tehran
- Dipartimento di Scienze Biomediche, Università di Padova, Via U. Bassi 58/B, 35121 Padova, Italy
| | - Giulia Zanetti
- Dipartimento di Scienze Biomediche, Università di Padova, Via U. Bassi 58/B, 35121 Padova, Italy
| | - Ornella Rossetto
- Dipartimento di Scienze Biomediche, Università di Padova, Via U. Bassi 58/B, 35121 Padova, Italy
| | - Cesare Montecucco
- Dipartimento di Scienze Biomediche, Università di Padova, Via U. Bassi 58/B, 35121 Padova, Italy; Istituto CNR di Neuroscienze, Università di Padova, Via U. Bassi 58/B, 35121 Padova, Italy
| |
Collapse
|
49
|
Upadhyay A, Arsi K, Wagle BR, Upadhyaya I, Shrestha S, Donoghue AM, Donoghue DJ. Trans-Cinnamaldehyde, Carvacrol, and Eugenol Reduce Campylobacter jejuni Colonization Factors and Expression of Virulence Genes in Vitro. Front Microbiol 2017; 8:713. [PMID: 28487683 PMCID: PMC5403884 DOI: 10.3389/fmicb.2017.00713] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 04/06/2017] [Indexed: 11/13/2022] Open
Abstract
Campylobacter jejuni is a major foodborne pathogen that causes severe gastroenteritis in humans characterized by fever, diarrhea, and abdominal cramps. In the human gut, Campylobacter adheres and invades the intestinal epithelium followed by cytolethal distending toxin mediated cell death, and enteritis. Reducing the attachment and invasion of Campylobacter to intestinal epithelium and expression of its virulence factors such as motility and cytolethal distending toxin (CDT) production could potentially reduce infection in humans. This study investigated the efficacy of sub-inhibitory concentrations (SICs, concentration not inhibiting bacterial growth) of three GRAS (generally recognized as safe) status phytochemicals namely trans-cinnamaldehyde (TC; 0.005, 0.01%), carvacrol (CR; 0.001, 0.002%), and eugenol (EG; 0.005, 0.01%) in reducing the attachment, invasion, and translocation of C. jejuni on human intestinal epithelial cells (Caco-2). Additionally, the effect of these phytochemicals on Campylobacter motility and CDT production was studied using standard bioassays and gene expression analysis. All experiments had duplicate samples and were replicated three times on three strains (wild type S-8, NCTC 11168, 81-176) of C. jejuni. Data were analyzed using ANOVA with GraphPad ver. 6. Differences between the means were considered significantly different at P < 0.05. The majority of phytochemical treatments reduced C. jejuni adhesion, invasion, and translocation of Caco-2 cells (P < 0.05). In addition, the phytochemicals reduced pathogen motility and production of CDT in S-8 and NCTC 11168 (P < 0.05). Real-time quantitative PCR revealed that phytochemicals reduced the transcription of select C. jejuni genes critical for infection in humans (P < 0.05). Results suggest that TC, CR, and EG could potentially be used to control C. jejuni infection in humans.
Collapse
Affiliation(s)
- Abhinav Upadhyay
- Department of Poultry Science, University of ArkansasFayetteville, AR, USA
| | - Komala Arsi
- Department of Poultry Science, University of ArkansasFayetteville, AR, USA
| | - Basanta R Wagle
- Department of Poultry Science, University of ArkansasFayetteville, AR, USA
| | - Indu Upadhyaya
- Department of Poultry Science, University of ArkansasFayetteville, AR, USA
| | - Sandip Shrestha
- Department of Poultry Science, University of ArkansasFayetteville, AR, USA
| | - Ann M Donoghue
- Poultry Production and Product Safety Research Unit, Agricultural Research Service (USDA)Fayetteville, AR, USA
| | - Dan J Donoghue
- Department of Poultry Science, University of ArkansasFayetteville, AR, USA
| |
Collapse
|
50
|
Grimm C, Butz E, Chen CC, Wahl-Schott C, Biel M. From mucolipidosis type IV to Ebola: TRPML and two-pore channels at the crossroads of endo-lysosomal trafficking and disease. Cell Calcium 2017; 67:148-155. [PMID: 28457591 DOI: 10.1016/j.ceca.2017.04.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/05/2017] [Accepted: 04/05/2017] [Indexed: 01/05/2023]
Abstract
What do lysosomal storage disorders such as mucolipidosis type IV have in common with Ebola, cancer cell migration, or LDL-cholesterol trafficking? LDL-cholesterol, certain bacterial toxins and viruses, growth factors, receptors, integrins, macromolecules destined for degradation or secretion are all sorted and transported via the endolysosomal system (ES). There are several pathways known in the ES, e.g. the degradation, the recycling, or the retrograde trafficking pathway. The ES comprises early and late endosomes, lysosomes and recycling endosomes as well as autophagosomes and lysosome related organelles. Contact sites between the ES and the endoplasmic reticulum or the Golgi apparatus may also be considered part of it. Dysfunction of this complex intracellular machinery can cause or contribute to the development of a number of diseases ranging from neurodegenerative, infectious, or metabolic diseases to retinal and pigmentation disorders as well as cancer and autophagy-related diseases. Endolysosomal ion channels such as mucolipins (TRPMLs) and two-pore channels (TPCs) play an important role in intracellular cation/calcium signaling and homeostasis and appear to critically contribute to the proper function of the endolysosomal trafficking network.
Collapse
Affiliation(s)
- Christian Grimm
- Munich Center for Integrated Protein Science CIPSM, Center for Drug Research, Ludwig-Maximilians-Universität, München, Germany; Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Germany.
| | - Elisabeth Butz
- Munich Center for Integrated Protein Science CIPSM, Center for Drug Research, Ludwig-Maximilians-Universität, München, Germany; Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Germany
| | - Cheng-Chang Chen
- Munich Center for Integrated Protein Science CIPSM, Center for Drug Research, Ludwig-Maximilians-Universität, München, Germany; Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Germany
| | - Christian Wahl-Schott
- Munich Center for Integrated Protein Science CIPSM, Center for Drug Research, Ludwig-Maximilians-Universität, München, Germany; Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Germany
| | - Martin Biel
- Munich Center for Integrated Protein Science CIPSM, Center for Drug Research, Ludwig-Maximilians-Universität, München, Germany; Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Germany.
| |
Collapse
|