1
|
Ashokcoomar S, Naidoo TJ, Cumming BM, Baig Z, Truebody B, Mackenzie JS, Steyn AJC, Pillay M. Mycobacterium tuberculosis curli pili reduces oxygen consumption rate of THP-1 macrophages during early infection. Int J Biochem Cell Biol 2025; 185:106802. [PMID: 40381906 DOI: 10.1016/j.biocel.2025.106802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/25/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
The development of improved anti-tuberculosis (TB) strategies to address drug-resistance and ineffectual TB treatment regimens should focus on interrupting the initial host-pathogen interaction. This study aimed to elucidate the effect of surface-located adhesin, Mycobacterium tuberculosis (Mtb) curli pili (MTP), on the bioenergetic and metabolomic profiles of THP-1 macrophages during initial stages of infection. Differentiated THP-1 macrophages were infected with wildtype (WT), ∆mtp, or mtp-complemented strains of Mtb. Bioenergetic profiles and metabolic flux were determined and statistical analysis highlighted differences/similarities amongst the THP-1 macrophage groups. The ∆mtp infected THP-1 macrophages mimicked the higher oxygen consumption rate (OCR) for basal respiration, ATP production, maximal respiration and spare respiratory capacity of the uninfected THP-1 macrophages, relative to the WT and mtp-complement infected THP-1 macrophages. The ∆mtp infected THP-1 macrophages displayed the highest compensatory glycolytic rate. Mtb infection caused the redirection of carbon from the tricarboxylic acid cycle to glycolysis, in addition to an increased flux through the pentose phosphate pathway. However, in the ∆mtp infected THP-1 macrophages, the total metabolite abundance was lower, similar to the uninfected THP-1 macrophages. Data indicates that the absence of MTP facilitates prompt clearance of the intracellular pathogen before it establishes a successful infection. This implies that the presence of MTP facilitates the survival of the pathogen during the early stages until infection is established. These findings support the growing evidence that the MTP adhesin is an important virulence factor and interruption of the interaction between pathogen and host, will facilitate swift clearance of the infection by the host.
Collapse
Affiliation(s)
- Shinese Ashokcoomar
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 1st floor Doris Duke Medical Research Institute, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Tarien J Naidoo
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 1st floor Doris Duke Medical Research Institute, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Bridgette M Cumming
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Zainab Baig
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Barry Truebody
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Jared S Mackenzie
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Adrie J C Steyn
- Africa Health Research Institute, Nelson R. Mandela Medical School, 4th floor K-RITH Tower Building, Congella, Private Bag 7, Durban 4013, South Africa.
| | - Manormoney Pillay
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 1st floor Doris Duke Medical Research Institute, Congella, Private Bag 7, Durban 4013, South Africa.
| |
Collapse
|
2
|
Abbasnia S, Hashem Asnaashari AM, Sharebiani H, Soleimanpour S, Mosavat A, Rezaee SA. Mycobacterium tuberculosis and host interactions in the manifestation of tuberculosis. J Clin Tuberc Other Mycobact Dis 2024; 36:100458. [PMID: 38983441 PMCID: PMC11231606 DOI: 10.1016/j.jctube.2024.100458] [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] [Indexed: 07/11/2024] Open
Abstract
The final step of epigenetic processes is changing the gene expression in a new microenvironment in the body, such as neuroendocrine changes, active infections, oncogenes, or chemical agents. The case of tuberculosis (TB) is an outcome of Mycobacterium tuberculosis (M.tb) and host interaction in the manifestation of active and latent TB or clearance. This comprehensive review explains and interprets the epigenetics findings regarding gene expressions on the host-pathogen interactions in the development and progression of tuberculosis. This review introduces novel insights into the complicated host-pathogen interactions, discusses the challengeable results, and shows the gaps in the clear understanding of M.tb behavior. Focusing on the biological phenomena of host-pathogen interactions, the epigenetic changes, and their outcomes provides a promising future for developing effective TB immunotherapies when converting gene expression toward appropriate host immune responses gradually becomes attainable. Overall, this review may shed light on the dark sides of TB pathogenesis as a life-threatening disease. Therefore, it may support effective planning and implementation of epigenetics approaches for introducing proper therapies or effective vaccines.
Collapse
Affiliation(s)
- Shadi Abbasnia
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hiva Sharebiani
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture, and Research (ACECR), Razavi Khorasan, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Zang X, Zhang J, Jiang Y, Feng T, Cui Y, Wang H, Cui Z, Dang G, Liu S. Serine protease Rv2569c facilitates transmission of Mycobacterium tuberculosis via disrupting the epithelial barrier by cleaving E-cadherin. PLoS Pathog 2024; 20:e1012214. [PMID: 38722857 PMCID: PMC11081392 DOI: 10.1371/journal.ppat.1012214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Epithelial cells function as the primary line of defense against invading pathogens. However, bacterial pathogens possess the ability to compromise this barrier and facilitate the transmigration of bacteria. Nonetheless, the specific molecular mechanism employed by Mycobacterium tuberculosis (M.tb) in this process is not fully understood. Here, we investigated the role of Rv2569c in M.tb translocation by assessing its ability to cleave E-cadherin, a crucial component of cell-cell adhesion junctions that are disrupted during bacterial invasion. By utilizing recombinant Rv2569c expressed in Escherichia coli and subsequently purified through affinity chromatography, we demonstrated that Rv2569c exhibited cell wall-associated serine protease activity. Furthermore, Rv2569c was capable of degrading a range of protein substrates, including casein, fibrinogen, fibronectin, and E-cadherin. We also determined that the optimal conditions for the protease activity of Rv2569c occurred at a temperature of 37°C and a pH of 9.0, in the presence of MgCl2. To investigate the function of Rv2569c in M.tb, a deletion mutant of Rv2569c and its complemented strains were generated and used to infect A549 cells and mice. The results of the A549-cell infection experiments revealed that Rv2569c had the ability to cleave E-cadherin and facilitate the transmigration of M.tb through polarized A549 epithelial cell layers. Furthermore, in vivo infection assays demonstrated that Rv2569c could disrupt E-cadherin, enhance the colonization of M.tb, and induce pathological damage in the lungs of C57BL/6 mice. Collectively, these results strongly suggest that M.tb employs the serine protease Rv2569c to disrupt epithelial defenses and facilitate its systemic dissemination by crossing the epithelial barrier.
Collapse
Affiliation(s)
- Xinxin Zang
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Jiajun Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Yanyan Jiang
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Tingting Feng
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Yingying Cui
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Hui Wang
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Ziyin Cui
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Guanghui Dang
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Siguo Liu
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| |
Collapse
|
4
|
Forouharmehr A. Whole proteome screening to develop a potent epitope-based vaccine against Coxiella burnetii: a reverse vaccinology approach. J Biomol Struct Dyn 2024:1-13. [PMID: 38488603 DOI: 10.1080/07391102.2024.2326198] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/27/2024] [Indexed: 03/25/2025]
Abstract
Coxiellosis is known as a threat to human health. This study aimed to develop an epitope-based vaccine against coxiellosis using a whole proteome investigation. In this case, the whole proteome of Coxiella burnetii was collected from the database, then different assessments were performed to select immunogenic proteins. The selected proteins were used for epitopes prediction. The epitope-based vaccine was made using the best-selected epitopes and HBHA protein. The physical and chemical features, as well as secondary and tertiary structures of the developed vaccine were analyzed. The interaction between the developed vaccine and TLR4/MD2 receptor was examined using molecular docking and molecular dynamic simulation. Finally, in silico cloning, codon optimization, and immune response simulation for the developed vaccine were performed. The findings supported a stable, hydrophilic, antigenic and non-allergenic vaccine with a molecular weight equal to 59.261 kDa and 542 amino acid residues in length. The findings showed that the developed vaccine not only could dock to TRL4/MD2 receptor with an affinity of -20.9 kcal/mol and 15 hydrogen bonds, but also the protein-protein complex was stable during molecular dynamic simulation with the binding free energy of -57.9 ± 6.9 kcal/mol. Furthermore, the optimized sequence of the developed vaccine with a CAI value of 0.97, could be cloned into the pET-21a (+) vector. Finally, The results confirmed that the developed vaccine could strongly trigger primary and secondary immune responses. Evidently, the developed vaccine can be an interesting candidate to apply.
Collapse
Affiliation(s)
- Ali Forouharmehr
- Department of Animal Science, Faculty of Agriculture, Lorestan University, Khorramabad, Iran
| |
Collapse
|
5
|
Mishra R, Hannebelle M, Patil VP, Dubois A, Garcia-Mouton C, Kirsch GM, Jan M, Sharma K, Guex N, Sordet-Dessimoz J, Perez-Gil J, Prakash M, Knott GW, Dhar N, McKinney JD, Thacker VV. Mechanopathology of biofilm-like Mycobacterium tuberculosis cords. Cell 2023; 186:5135-5150.e28. [PMID: 37865090 PMCID: PMC10642369 DOI: 10.1016/j.cell.2023.09.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/26/2023] [Accepted: 09/14/2023] [Indexed: 10/23/2023]
Abstract
Mycobacterium tuberculosis (Mtb) cultured axenically without detergent forms biofilm-like cords, a clinical identifier of virulence. In lung-on-chip (LoC) and mouse models, cords in alveolar cells contribute to suppression of innate immune signaling via nuclear compression. Thereafter, extracellular cords cause contact-dependent phagocyte death but grow intercellularly between epithelial cells. The absence of these mechanopathological mechanisms explains the greater proportion of alveolar lesions with increased immune infiltration and dissemination defects in cording-deficient Mtb infections. Compression of Mtb lipid monolayers induces a phase transition that enables mechanical energy storage. Agent-based simulations demonstrate that the increased energy storage capacity is sufficient for the formation of cords that maintain structural integrity despite mechanical perturbation. Bacteria in cords remain translationally active despite antibiotic exposure and regrow rapidly upon cessation of treatment. This study provides a conceptual framework for the biophysics and function in tuberculosis infection and therapy of cord architectures independent of mechanisms ascribed to single bacteria.
Collapse
Affiliation(s)
- Richa Mishra
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Melanie Hannebelle
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Vishal P Patil
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Anaëlle Dubois
- BioElectron Microscopy Facility, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | - Gabriela M Kirsch
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Maxime Jan
- Bioinformatics Competence Centre, University of Lausanne, 1015 Lausanne, Switzerland; Bioinformatics Competence Centre, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Kunal Sharma
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Nicolas Guex
- Bioinformatics Competence Centre, University of Lausanne, 1015 Lausanne, Switzerland; Bioinformatics Competence Centre, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jessica Sordet-Dessimoz
- Histology Core Facility, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jesus Perez-Gil
- Department of Biochemistry, University Complutense Madrid, 28040 Madrid, Spain
| | - Manu Prakash
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Graham W Knott
- BioElectron Microscopy Facility, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Neeraj Dhar
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - John D McKinney
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Vivek V Thacker
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
6
|
Bisht D, Singh R, Sharma D, Sharma D, Gautam S, Gupta MK. Unraveling Major Proteins of Mycobacterium tuberculosis Envelope. CURR PROTEOMICS 2022; 19:372-379. [DOI: 10.2174/1570164619666220908141130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/30/2022] [Accepted: 08/04/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Although treatable, resistant form of tuberculosis (TB) has posed a major impediment to the
effective TB control programme. As the Mycobacterium tuberculosis cell envelope is closely associated
with its virulence and resistance, it is very important to understand the cell envelope for better
treatment of causative pathogens. Cell membrane plays a crucial role in imparting various cell functions.
Proteins being the functional moiety, it is impossible to characterize the functional properties
based on genetic analysis alone. Proteomic based research has indicated mycobacterial envelope as a
good source of antigens/proteins. Envelope/membrane and associated proteins have an anticipated role
in biological processes, which could be of vital importance to the microbe, and hence could qualify as
drug targets. This review provides an overview of the prominent and biologically important cell envelope
and highlights the different functions offered by the proteins associated with it. Selective targeting
of the mycobacterial envelope offers an untapped opportunity to address the problems associated
with the current drug regimen and also will lead to the development of more potent and safer drugs
against all forms of tuberculous infections.
Collapse
Affiliation(s)
- Deepa Bisht
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj,
Agra (UP)-282001, India
| | - Rananjay Singh
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj,
Agra (UP)-282001, India
| | - Devesh Sharma
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj,
Agra (UP)-282001, India
| | - Divakar Sharma
- Department of Microbiology, Maulana Azad Medical College, Bahadur Shah Zafar Marg,
New Delhi-110002, India
| | - Sakshi Gautam
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj,
Agra (UP)-282001, India
| | | |
Collapse
|
7
|
Mycobacterial Adhesion: From Hydrophobic to Receptor-Ligand Interactions. Microorganisms 2022; 10:microorganisms10020454. [PMID: 35208908 PMCID: PMC8875947 DOI: 10.3390/microorganisms10020454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 11/24/2022] Open
Abstract
Adhesion is crucial for the infective lifestyles of bacterial pathogens. Adhesion to non-living surfaces, other microbial cells, and components of the biofilm extracellular matrix are crucial for biofilm formation and integrity, plus adherence to host factors constitutes a first step leading to an infection. Adhesion is, therefore, at the core of pathogens’ ability to contaminate, transmit, establish residency within a host, and cause an infection. Several mycobacterial species cause diseases in humans and animals with diverse clinical manifestations. Mycobacterium tuberculosis, which enters through the respiratory tract, first adheres to alveolar macrophages and epithelial cells leading up to transmigration across the alveolar epithelium and containment within granulomas. Later, when dissemination occurs, the bacilli need to adhere to extracellular matrix components to infect extrapulmonary sites. Mycobacteria causing zoonotic infections and emerging nontuberculous mycobacterial pathogens follow divergent routes of infection that probably require adapted adhesion mechanisms. New evidence also points to the occurrence of mycobacterial biofilms during infection, emphasizing a need to better understand the adhesive factors required for their formation. Herein, we review the literature on tuberculous and nontuberculous mycobacterial adhesion to living and non-living surfaces, to themselves, to host cells, and to components of the extracellular matrix.
Collapse
|
8
|
Abstract
Streptomyces coelicolor is a model organism for the study of Streptomyces, a genus of Gram-positive bacteria that undergoes a complex life cycle and produces a broad repertoire of bioactive metabolites and extracellular enzymes. This study investigated the production and characterization of membrane vesicles (MVs) in liquid cultures of S. coelicolor M145 from a structural and biochemical point of view; this was achieved by combining microscopic, physical and -omics analyses. Two main populations of MVs, with different size and cargo, were isolated and purified. S. coelicolor MV cargo was determined being complex and containing different kinds of proteins and metabolites. In particular, a whole of 166 proteins involved in cell metabolism/differentiation, molecular processing/transport, and stress response was identified in MVs, the latter functional class being also important for bacterial morpho-physiological differentiation. A subset of these proteins was protected from degradation following treatment of MVs with proteinase K, indicating their localization inside the vesicles. Moreover, S. coelicolor MVs contained an array of metabolites, such as antibiotics, vitamins, amino acids and components of carbon metabolism. In conclusion, this analysis provides detailed information on S. coelicolor MVs under basal conditions and corresponding content, which may be useful in a next future to elucidate vesicle biogenesis and functions. Importance Streptomycetes are widely distributed in nature, and they are characterized by a complex life cycle that involves morphological differentiation. They are very relevant in industry because they produce about a half of the antibiotics used clinically and other important pharmaceutical products having natural origin. Streptomyces coelicolor is a model organism for the study of bacterial differentiation and bioactive molecule production. S. coelicolor produces extracellular vesicles carrying many molecules such as proteins and metabolites, including antibiotics. The elucidation of S. coelicolor extracellular vesicle cargo will help to understand different aspects of streptomycete physiology, such as cell communication during differentiation and response to environmental stimuli. Moreover, the capability of carrying different kind of biomolecules opens up new biotechnological possibilities related to drug delivery. Indeed, the decoding of molecular mechanisms involved in cargo selection may lead to the customization of the content of extracellular vesicles.
Collapse
|
9
|
Veyron-Churlet R, Saliou JM, Locht C. Interconnection of the mycobacterial heparin-binding hemagglutinin with cholesterol degradation and heme/iron pathways identified by proximity-dependent biotin identification in Mycobacterium smegmatis. Environ Microbiol 2021; 23:3212-3224. [PMID: 33913567 DOI: 10.1111/1462-2920.15547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/15/2021] [Accepted: 04/26/2021] [Indexed: 11/28/2022]
Abstract
Deciphering protein-protein interactions is a critical step in the identification and the understanding of biological mechanisms deployed by pathogenic bacteria. The development of in vivo technologies to characterize these interactions is still in its infancy, especially for bacteria whose subcellular organization is particularly complex, such as mycobacteria. In this work, we used the proximity-dependent biotin identification (BioID) to define the mycobacterial heparin-binding hemagglutinin (HbhA) interactome in the saprophytic bacterium Mycobacterium smegmatis. M. smegmatis is a commonly used model to study and characterize the physiology of pathogenic mycobacteria, such as Mycobacterium tuberculosis. Here, we adapted the BioID technology to in vivo protein-protein interactions studies in M. smegmatis, which presents several advantages, such as maintaining the complex organization of the mycomembrane, offering the possibility to study membrane or cell wall-associated proteins, including HbhA, in the presence of cofactors and post-translational modifications, such as the complex methylation pattern of HbhA. Using this technology, we found that HbhA is interconnected with cholesterol degradation and heme/iron pathways. These results are in line with previous studies showing the dual localization of HbhA, associated with the cell wall and intracytoplasmic lipid inclusions, and its induction under high iron growth conditions.
Collapse
Affiliation(s)
- Romain Veyron-Churlet
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, F-59000, France
| | - Jean-Michel Saliou
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UMS 2014 - PLBS, Lille, F-59000, France
| | - Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, F-59000, France
| |
Collapse
|
10
|
Abstract
Mycobacteria have unique cell envelopes, surface properties, and growth dynamics, which all play a part in the ability of these important pathogens to infect, evade host immunity, disseminate, and resist antibiotic challenges. Recent atomic force microscopy (AFM) studies have brought new insights into the nanometer-scale ultrastructural, adhesive, and mechanical properties of mycobacteria. The molecular forces with which mycobacterial adhesins bind to host factors, like heparin and fibronectin, and the hydrophobic properties of the mycomembrane have been unraveled by AFM force spectroscopy studies. Real-time correlative AFM and fluorescence imaging have delineated a complex interplay between surface ultrastructure, tensile stresses within the cell envelope, and cellular processes leading to division. The unique capabilities of AFM, which include subdiffraction-limit topographic imaging and piconewton force sensitivity, have great potential to resolve important questions that remain unanswered on the molecular interactions, surface properties, and growth dynamics of this important class of pathogens.
Collapse
|
11
|
Faria-Ramos I, Poças J, Marques C, Santos-Antunes J, Macedo G, Reis CA, Magalhães A. Heparan Sulfate Glycosaminoglycans: (Un)Expected Allies in Cancer Clinical Management. Biomolecules 2021; 11:136. [PMID: 33494442 PMCID: PMC7911160 DOI: 10.3390/biom11020136] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
In an era when cancer glycobiology research is exponentially growing, we are witnessing a progressive translation of the major scientific findings to the clinical practice with the overarching aim of improving cancer patients' management. Many mechanistic cell biology studies have demonstrated that heparan sulfate (HS) glycosaminoglycans are key molecules responsible for several molecular and biochemical processes, impacting extracellular matrix properties and cellular functions. HS can interact with a myriad of different ligands, and therefore, hold a pleiotropic role in regulating the activity of important cellular receptors and downstream signalling pathways. The aberrant expression of HS glycan chains in tumours determines main malignant features, such as cancer cell proliferation, angiogenesis, invasion and metastasis. In this review, we devote particular attention to HS biological activities, its expression profile and modulation in cancer. Moreover, we highlight HS clinical potential to improve both diagnosis and prognosis of cancer, either as HS-based biomarkers or as therapeutic targets.
Collapse
Affiliation(s)
- Isabel Faria-Ramos
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
| | - Juliana Poças
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Molecular Biology Department, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Catarina Marques
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Molecular Biology Department, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - João Santos-Antunes
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Pathology Department, Faculdade de Medicina, University of Porto, 4200-319 Porto, Portugal;
- Gastroenterology Department, Centro Hospitalar S. João, 4200-319 Porto, Portugal
| | - Guilherme Macedo
- Pathology Department, Faculdade de Medicina, University of Porto, 4200-319 Porto, Portugal;
- Gastroenterology Department, Centro Hospitalar S. João, 4200-319 Porto, Portugal
| | - Celso A. Reis
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
- Molecular Biology Department, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Pathology Department, Faculdade de Medicina, University of Porto, 4200-319 Porto, Portugal;
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (I.F.-R.); (J.P.); (C.M.); (J.S.-A.); (C.A.R.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-135 Porto, Portugal
| |
Collapse
|
12
|
Chiu PC, Liou HC, Ling TY, Shen LJ. Development of a Neuroprotective Erythropoietin Modified with a Novel Carrier for the Blood-Brain Barrier. Neurotherapeutics 2020; 17:1184-1196. [PMID: 32144722 PMCID: PMC7609523 DOI: 10.1007/s13311-020-00845-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Extremely high doses of erythropoietin (EPO) has been used for neuroprotection in ischemia-reperfusion brain injury to deliver sufficient amounts of EPO across the blood-brain barrier (BBB); however, harmful outcomes were observed afterward. We aimed to test the ability of HBHAc (heparin-binding haemagglutinin adhesion c), an intracellular delivery peptide for macromolecules, as an EPO carrier across the BBB. The cellular internalization and transcytosis ability of HBHAc-modified EPO (EPO-HBHAc) were evaluated in bEnd.3 cells and in the bEnd.3/CTX TNA2 co-culture BBB model, respectively. Subsequently, the NMDA-induced-toxicity model and ischemia-reperfusion rat model were used to understand the neuronal protective activity of EPO-HBHAc. The biodistribution of EPO-HBHAc was demonstrated in rats by the quantification of EPO-HBHAc in the brain, plasma, and organs by ELISA. Our results demonstrate that EPO-HBHAc exhibited significantly higher cellular internalization in dose- and time-dependent manners and better transcytosis ability than EPO. In addition, the transported EPO-HBHAc in the co-culture transwell system maintained the neuronal protective activity when primary rat cortical neurons underwent NMDA-induced toxicity. The calculated cerebral infarction area of rats treated with EPO-HBHAc was significantly reduced compared to that of rats treated with EPO (29.9 ± 7.0% vs 48.9 ± 7.9%) 24 h after occlusion in 3VO rat experiments. Moreover, the EPO amount in both CSF and damaged cortex from the EPO-HBHAc group was 4.0-fold and 3.0-fold higher than the EPO group, respectively. These results suggest that HBHAc would be a favorable tool for EPO brain delivery and would further extend the clinical applications of EPO in neuroprotection.
Collapse
Affiliation(s)
- Po-Chuan Chiu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Houng-Chi Liou
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Jiuan Shen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
13
|
Moule MG, Cirillo JD. Mycobacterium tuberculosis Dissemination Plays a Critical Role in Pathogenesis. Front Cell Infect Microbiol 2020; 10:65. [PMID: 32161724 PMCID: PMC7053427 DOI: 10.3389/fcimb.2020.00065] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/07/2020] [Indexed: 12/27/2022] Open
Abstract
Mycobacterium tuberculosis is primarily a respiratory pathogen. However, 15% of infections worldwide occur at extrapulmonary sites causing additional complications for diagnosis and treatment of the disease. In addition, dissemination of M. tuberculosis out of the lungs is thought to be more than just a rare event leading to extrapulmonary tuberculosis, but rather a prerequisite step that occurs during all infections, producing secondary lesions that can become latent or productive. In this review we will cover the clinical range of extrapulmonary infections and the process of dissemination including evidence from both historical medical literature and animal experiments for dissemination and subsequent reseeding of the lungs through the lymphatic and circulatory systems. While the mechanisms of M. tuberculosis dissemination are not fully understood, we will discuss the various models that have been proposed to address how this process may occur and summarize the bacterial virulence factors that facilitate M. tuberculosis dissemination.
Collapse
Affiliation(s)
- Madeleine G. Moule
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Jeffrey D. Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX, United States
| |
Collapse
|
14
|
Vinod V, Vijayrajratnam S, Vasudevan AK, Biswas R. The cell surface adhesins of Mycobacterium tuberculosis. Microbiol Res 2019; 232:126392. [PMID: 31841935 DOI: 10.1016/j.micres.2019.126392] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/11/2019] [Accepted: 12/07/2019] [Indexed: 02/07/2023]
Abstract
Bacterial cell surface adhesins play a major role in facilitating host colonization and subsequent establishment of infection. The surface of Mycobacterium tuberculosis, owing to the complex architecture of its cell envelope, expresses numerous adhesins with varied chemical nature, including proteins, lipids, lipoproteins, glycoproteins and glycopolymers. Studies on mycobacterial adhesins show that they bind with multifarious host receptors and extracellular matrix (ECM) components. In this review we have highlighted the adhesins that are abundantly present on the mycobacterial surface and their interactions with host receptors. M. tuberculosis interacts with various host cell surface receptors such as toll like receptors, C-type lectin receptors, scavenger receptors, and Fc and complement receptors. Apart from these, ECM components like fibronectin, collagen, elastin, laminin, fibrillin and vitronectin also provide binding sites for surface adhesins of the tubercle bacilli. M. tuberculosis adhesins include proteins with and without signal peptide sequence and transmembrane proteins. Other surface adhesin macromolecules of M. tuberculosis comprises of lipids, glycolipids and glycopolymers. The interaction between the mycobacterial adhesins and their host receptors result in adhesion of the microbe to the host cells, induction of immune response and aid in the pathogenesis of the disease. A thorough understanding of the different M. tuberculosis surface adhesins and host receptors will provide a better picture of interaction between them at molecular level. The information gained on adhesins and host receptors will prove beneficial in developing novel therapeutic strategies such as the use of anti-adhesin molecules to hinder the adhesion of bacteria to the host cells, thereby preventing establishment of infection. The surface molecules discussed in this review will also benefit in identification of new drug targets, diagnostic markers or vaccine candidates against the deadly pathogen.
Collapse
Affiliation(s)
- Vivek Vinod
- Center for Nanosciences and Molecular Medicine, Amrita School of Medical Sciences and Research Center (AIMS), Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Sukhithasri Vijayrajratnam
- Center for Nanosciences and Molecular Medicine, Amrita School of Medical Sciences and Research Center (AIMS), Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Anil Kumar Vasudevan
- Department of Microbiology, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Raja Biswas
- Center for Nanosciences and Molecular Medicine, Amrita School of Medical Sciences and Research Center (AIMS), Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India.
| |
Collapse
|
15
|
Chiu PC, Hsieh PY, Kang JW, Chang PH, Shen LJ. Study of the intracellular delivery mechanism of a pH-sensitive peptide modified with enhanced green fluorescent protein. J Drug Target 2019; 28:408-418. [PMID: 31524004 DOI: 10.1080/1061186x.2019.1669041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The targeted delivery of therapeutic agents is a promising approach to enhance the efficacy and reduce the toxicity of cancer treatments. Understanding the intracellular endocytic mechanisms of a cell penetrating peptide (CPP) in an acidic environment is important for targeted delivery of macromolecules to tumours. In this study, we constructed a pH-sensitive CPP-based delivery system for the intracellular delivery of macromolecules. A pH-sensitive CPP, HBHAc, was fused with a model protein, enhanced green fluorescent protein (EGFP), through recombinant DNA technology. We found that is essential that negatively charged proteoglycans on the cell surface interact with HBHAc-EGFP prior to the cellular uptake of HBHAc-EGFP. The uptake was significantly restricted at 4 °C under pH conditions of both 6.5 and 7.5. The increased positive charge of HBHAc-EGFP under the acidic condition leads to a pH-dependent cellular uptake, and we observed that the internalisation of HBHAc-EGFP was significantly higher at pH 6.5 than at pH 7.5 (p < .05). Thus, with pH-sensitive activity, HBHAc is expected to improve tumour-targeted intracellular protein delivery. Moreover, our findings provide a new insight that the endocytic pathway may change under different pH conditions and suggest that this unique phenomenon benefits pH-sensitive drug delivery for tumour therapy.
Collapse
Affiliation(s)
- Po-Chuan Chiu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Yu Hsieh
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jyun-Wei Kang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Hsun Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Jiuan Shen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
16
|
De Maio F, Squeglia F, Goletti D, Delogu G. The Mycobacterial HBHA Protein: A Promising Biomarker for Tuberculosis. Curr Med Chem 2019; 26:2051-2060. [PMID: 30378481 DOI: 10.2174/0929867325666181029165805] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/01/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
Abstract
A major goal in tuberculosis (TB) research is the identification, among the subjects infected with Mycobacterium tuberculosis (Mtb), of those with active TB, or at higher risk of developing active disease, from the latently infected subjects. The classical heterogeneity of Mtb infection and TB disease is a major obstacle toward the identification of reliable biomarkers that can stratify Mtb infected subjects based on disease risk. The heparin-binding haemagglutinin (HBHA) is a mycobacterial surface antigen that is implicated in tuberculosis (TB) pathogenesis. The host immune response against HBHA varies depending on the TB status and several studies are supporting the role of HBHA as a useful biomarker of TB.
Collapse
Affiliation(s)
- Flavio De Maio
- Institute of Microbiology, Università Cattolica del Sacro Cuore, Milano, Italy.,Fondazione Policlinico Universitario A. Gemelli- IRCCS, Rome, Italy
| | - Flavia Squeglia
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16. I-80134 Napoli, Italy
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI) IRCCCS, Rome, Italy
| | - Giovanni Delogu
- Institute of Microbiology, Università Cattolica del Sacro Cuore, Milano, Italy.,Fondazione Policlinico Universitario A. Gemelli- IRCCS, Rome, Italy
| |
Collapse
|
17
|
Kolbe K, Veleti SK, Reiling N, Lindhorst TK. Lectins of Mycobacterium tuberculosis - rarely studied proteins. Beilstein J Org Chem 2019; 15:1-15. [PMID: 30680034 PMCID: PMC6334816 DOI: 10.3762/bjoc.15.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/29/2018] [Indexed: 12/19/2022] Open
Abstract
The importance of bacterial lectins for adhesion, pathogenicity, and biofilm formation is well established for many Gram-positive and Gram-negative bacteria. However, there is very little information available about lectins of the tuberculosis-causing bacterium, Mycobacterium tuberculosis (Mtb). In this paper we review previous studies on the carbohydrate-binding characteristics of mycobacteria and related Mtb proteins, discussing their potential relevance to Mtb infection and pathogenesis.
Collapse
Affiliation(s)
- Katharina Kolbe
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, 33 North Drive, Bethesda, 20892, MD, United States
| | - Sri Kumar Veleti
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, 33 North Drive, Bethesda, 20892, MD, United States
| | - Norbert Reiling
- Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, Parkallee 22, 23845 Borstel, Germany.,German Center for Infection Research (DZIF), Borstel Site, 23845 Borstel, Germany
| | - Thisbe K Lindhorst
- Otto Diels Institute of Organic Chemistry, Christiana Albertina University of Kiel, Otto-Hahn-Platz 3-4, 24118 Kiel, Germany
| |
Collapse
|
18
|
Marimani M, Ahmad A, Duse A. The role of epigenetics, bacterial and host factors in progression of Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 2018; 113:200-214. [PMID: 30514504 DOI: 10.1016/j.tube.2018.10.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/21/2018] [Accepted: 10/23/2018] [Indexed: 12/29/2022]
Abstract
Tuberculosis (TB) infection caused by Mycobacterium tuberculosis (Mtb) is still a persistent global health problem, particularly in developing countries. The World Health Organization (WHO) reported a mortality rate of about 1.8 million worldwide due to TB complications in 2015. The Bacillus Calmette-Guérin (BCG) vaccine was introduced in 1921 and is still widely used to prevent TB development. This vaccine offers up to 80% protection against various forms of TB; however its efficacy against lung infection varies among different geographical settings. Devastatingly, the development of various forms of drug-resistant TB strains has significantly impaired the discovery of effective and safe anti-bacterial agents. Consequently, this necessitated discovery of new drug targets and novel anti-TB therapeutics to counter infection caused by various Mtb strains. Importantly, various factors that contribute to TB development have been identified and include bacterial resuscitation factors, host factors, environmental factors and genetics. Furthermore, Mtb-induced epigenetic changes also play a crucial role in evading the host immune response and leads to bacterial persistence and dissemination. Recently, the application of GeneXpert MTB/RIF® to rapidly diagnose and identify drug-resistant strains and discovery of different molecular markers that distinguish between latent and active TB infection has motivated and energised TB research. Therefore, this review article will briefly discuss the current TB state, highlight various mechanisms employed by Mtb to evade the host immune response as well as to discuss some modern molecular techniques that may potentially target and inhibit Mtb replication.
Collapse
Affiliation(s)
- Musa Marimani
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Aijaz Ahmad
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, South Africa.
| | - Adriano Duse
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, South Africa
| |
Collapse
|
19
|
Girardin RC, Bai G, He J, Sui H, McDonough KA. AbmR (Rv1265) is a novel transcription factor of Mycobacterium tuberculosis that regulates host cell association and expression of the non-coding small RNA Mcr11. Mol Microbiol 2018; 110:811-830. [PMID: 30207611 PMCID: PMC6282994 DOI: 10.1111/mmi.14126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/07/2018] [Accepted: 09/09/2018] [Indexed: 12/11/2022]
Abstract
Gene regulatory networks used by Mycobacterium tuberculosis (Mtb) during infection include many genes of unknown function, confounding efforts to determine their roles in Mtb biology. Rv1265 encodes a conserved hypothetical protein that is expressed during infection and in response to elevated levels of cyclic AMP. Here, we report that Rv1265 is a novel auto‐inhibitory ATP‐binding transcription factor that upregulates expression of the small non‐coding RNA Mcr11, and propose that Rv1265 be named ATP‐binding mcr11regulator (AbmR). AbmR directly and specifically bound DNA, as determined by electrophoretic mobility shift assays, and this DNA‐binding activity was enhanced by AbmR’s interaction with ATP. Genetic knockout of abmR in Mtb increased abmR promoter activity and eliminated growth phase‐dependent increases in mcr11 expression during hypoxia. Mutagenesis identified arginine residues in the carboxy terminus that are critical for AbmR’s DNA‐binding activity and gene regulatory function. Limited similarity to other DNA‐ or ATP‐binding domains suggests that AbmR belongs to a novel class of DNA‐ and ATP‐binding proteins. AbmR was also found to form large organized structures in solution and facilitate the serum‐dependent association of Mtb with human lung epithelial cells. These results indicate a potentially complex role for AbmR in Mtb biology.
Collapse
Affiliation(s)
- Roxie C Girardin
- Department of Biomedical Sciences, School of Public Health, University at Albany, PO Box 22002, Albany, NY, 12201-2002, USA
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Jie He
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Haixin Sui
- Department of Biomedical Sciences, School of Public Health, University at Albany, PO Box 22002, Albany, NY, 12201-2002, USA.,Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Kathleen A McDonough
- Department of Biomedical Sciences, School of Public Health, University at Albany, PO Box 22002, Albany, NY, 12201-2002, USA.,Wadsworth Center, New York State Department of Health, Albany, NY, USA
| |
Collapse
|
20
|
van Leeuwen LM, Boot M, Kuijl C, Picavet DI, van Stempvoort G, van der Pol SM, de Vries HE, van der Wel NN, van der Kuip M, van Furth AM, van der Sar AM, Bitter W. Mycobacteria employ two different mechanisms to cross the blood-brain barrier. Cell Microbiol 2018; 20:e12858. [PMID: 29749044 PMCID: PMC6175424 DOI: 10.1111/cmi.12858] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/27/2018] [Accepted: 04/23/2018] [Indexed: 12/16/2022]
Abstract
Central nervous system (CNS) infection by Mycobacterium tuberculosis is one of the most devastating complications of tuberculosis, in particular in early childhood. In order to induce CNS infection, M. tuberculosis needs to cross specialised barriers protecting the brain. How M. tuberculosis crosses the blood-brain barrier (BBB) and enters the CNS is not well understood. Here, we use transparent zebrafish larvae and the closely related pathogen Mycobacterium marinum to answer this question. We show that in the early stages of development, mycobacteria rapidly infect brain tissue, either as free mycobacteria or within circulating macrophages. After the formation of a functionally intact BBB, the infiltration of brain tissue by infected macrophages is delayed, but not blocked, suggesting that crossing the BBB via phagocytic cells is one of the mechanisms used by mycobacteria to invade the CNS. Interestingly, depletion of phagocytic cells did not prevent M. marinum from infecting the brain tissue, indicating that free mycobacteria can independently cause brain infection. Detailed analysis showed that mycobacteria are able to cause vasculitis by extracellular outgrowth in the smaller blood vessels and by infecting endothelial cells. Importantly, we could show that this second mechanism is an active process that depends on an intact ESX-1 secretion system, which extends the role of ESX-1 secretion beyond the macrophage infection cycle.
Collapse
Affiliation(s)
- Lisanne M. van Leeuwen
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
- Paediatric Infectious Diseases and ImmunologyVU Medical CenterAmsterdamThe Netherlands
| | - Maikel Boot
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
| | - Coen Kuijl
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
| | - Daisy I. Picavet
- Cell Biology and Histology, Electron Microscopy Centre AmsterdamAcademic Medical CentreAmsterdamThe Netherlands
| | - Gunny van Stempvoort
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
| | - Susanne M.A. van der Pol
- Molecular Cell Biology and Immunology, Amsterdam NeuroscienceVU Medical CenterAmsterdamThe Netherlands
| | - Helga E. de Vries
- Molecular Cell Biology and Immunology, Amsterdam NeuroscienceVU Medical CenterAmsterdamThe Netherlands
| | - Nicole N. van der Wel
- Cell Biology and Histology, Electron Microscopy Centre AmsterdamAcademic Medical CentreAmsterdamThe Netherlands
| | - Martijn van der Kuip
- Paediatric Infectious Diseases and ImmunologyVU Medical CenterAmsterdamThe Netherlands
| | | | | | - Wilbert Bitter
- Medical Microbiology and Infection ControlVU Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
21
|
Abstract
Syndecan-1 (Sdc1) is a major cell surface heparan sulfate (HS) proteoglycan of epithelial cells, a cell type targeted by many bacterial pathogens early in their pathogenesis. Loss of Sdc1 in mice is a gain-of-function mutation that significantly decreases the susceptibility to several bacterial infections, suggesting that subversion of Sdc1 is an important virulence strategy. HS glycosaminoglycan (GAG) chains of cell surface Sdc1 promote bacterial pathogenesis by facilitating the attachment of bacteria to host cells. Engagement of cell surface Sdc1 HS chains by bacterial adhesins transmits signal through the highly conserved Sdc1 cytoplasmic domain, which can lead to uptake of intracellular bacterial pathogens. On the other hand, several bacteria that do not require Sdc1 for their attachment and invasion stimulate Sdc1 shedding and exploit the capacity of Sdc1 ectodomain HS GAGs to disarm innate defense mechanisms to evade immune clearance. Recent data suggest that select HS sulfate motifs, and not the overall charge of HS, are important in the inhibition of innate immune mechanisms. Here, we discuss several examples of Sdc1 subversion in bacterial infections.
Collapse
|
22
|
Squeglia F, Ruggiero A, De Simone A, Berisio R. A structural overview of mycobacterial adhesins: Key biomarkers for diagnostics and therapeutics. Protein Sci 2017; 27:369-380. [PMID: 29139177 DOI: 10.1002/pro.3346] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/09/2017] [Accepted: 11/09/2017] [Indexed: 01/14/2023]
Abstract
Adherence, colonization, and survival of mycobacteria in host cells require surface adhesins, which are attractive pharmacotherapeutic targets. A large arsenal of pilus and non-pilus adhesins have been identified in mycobacteria. These adhesins are capable of interacting with host cells, including macrophages and epithelial cells and are essential to microbial pathogenesis. In the last decade, several structures of mycobacterial adhesins responsible for adhesion to either macrophages or extra cellular matrix proteins have been elucidated. In addition, key structural and functional information have emerged for the process of mycobacterial adhesion to epithelial cells, mediated by the Heparin-binding hemagglutinin (HBHA). In this review, we provide an overview of the structural and functional features of mycobacterial adhesins and discuss their role as important biomarkers for diagnostics and therapeutics. Based on the reported data, it appears clear that adhesins are endowed with a variety of different structures and functions. Most adhesins play important roles in the cell life of mycobacteria and are key virulence factors. However, they have adapted to an extracellular life to exert a role in host-pathogen interaction. The type of interactions they form with the host and the adhesin regions involved in binding is partly known and is described in this review.
Collapse
Affiliation(s)
- Flavia Squeglia
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, Napoli, I-80134, Italy
| | - Alessia Ruggiero
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, Napoli, I-80134, Italy
| | - Alfonso De Simone
- Division of Molecular Biosciences, Imperial College London, SW7 2AZ, UK
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, Napoli, I-80134, Italy
| |
Collapse
|
23
|
Huang TY, Irene D, Zulueta MML, Tai TJ, Lain SH, Cheng CP, Tsai PX, Lin SY, Chen ZG, Ku CC, Hsiao CD, Chyan CL, Hung SC. Structure of the Complex between a Heparan Sulfate Octasaccharide and Mycobacterial Heparin-Binding Hemagglutinin. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201612518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Teng-Yi Huang
- Genomics Research Center; Academia Sinica; No. 128, Section 2, Academia Road Taipei 115 Taiwan
| | - Deli Irene
- Department of Chemistry; National Dong Hwa University; No. 1, Section 2, Da Hsueh Road, Shoufeng Hualien 974 Taiwan
| | - Medel Manuel L. Zulueta
- Genomics Research Center; Academia Sinica; No. 128, Section 2, Academia Road Taipei 115 Taiwan
| | - Tzu-Jui Tai
- Department of Chemistry; National Dong Hwa University; No. 1, Section 2, Da Hsueh Road, Shoufeng Hualien 974 Taiwan
| | - Shih-Han Lain
- Department of Chemistry; National Dong Hwa University; No. 1, Section 2, Da Hsueh Road, Shoufeng Hualien 974 Taiwan
| | - Cheng-Po Cheng
- Genomics Research Center; Academia Sinica; No. 128, Section 2, Academia Road Taipei 115 Taiwan
| | - Ping-Xi Tsai
- Genomics Research Center; Academia Sinica; No. 128, Section 2, Academia Road Taipei 115 Taiwan
| | - Shu-Yi Lin
- Genomics Research Center; Academia Sinica; No. 128, Section 2, Academia Road Taipei 115 Taiwan
| | - Zhi-Geng Chen
- Genomics Research Center; Academia Sinica; No. 128, Section 2, Academia Road Taipei 115 Taiwan
| | - Chiao-Chu Ku
- Institute of Molecular Biology; Academia Sinica; No. 128, Section 2, Academia Road Taipei 115 Taiwan
| | - Chwan-Deng Hsiao
- Institute of Molecular Biology; Academia Sinica; No. 128, Section 2, Academia Road Taipei 115 Taiwan
| | - Chia-Lin Chyan
- Department of Chemistry; National Dong Hwa University; No. 1, Section 2, Da Hsueh Road, Shoufeng Hualien 974 Taiwan
| | - Shang-Cheng Hung
- Genomics Research Center; Academia Sinica; No. 128, Section 2, Academia Road Taipei 115 Taiwan
| |
Collapse
|
24
|
Huang TY, Irene D, Zulueta MML, Tai TJ, Lain SH, Cheng CP, Tsai PX, Lin SY, Chen ZG, Ku CC, Hsiao CD, Chyan CL, Hung SC. Structure of the Complex between a Heparan Sulfate Octasaccharide and Mycobacterial Heparin-Binding Hemagglutinin. Angew Chem Int Ed Engl 2017; 56:4192-4196. [PMID: 28294485 DOI: 10.1002/anie.201612518] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/11/2016] [Indexed: 11/06/2022]
Abstract
Heparin-binding hemagglutinin (HBHA) is a 199 amino acid virulence factor at the envelope of Mycobacterium tuberculosis that contributes to latent tuberculosis. The binding of HBHA to respiratory epithelial cells, which leads to extrapulmonary dissemination of the pathogen, is mediated by cell-surface heparan sulfate (HS). We report the structural characterization of the HBHA/HS complex by NMR spectroscopy. To develop a model for the molecular recognition, the first chemically synthesized uniformly 13 C- and 15 N-labeled HS octasaccharide and a uniformly 13 C- and 15 N-labeled form of HBHA were prepared. Residues 180-195 at the C-terminal region of HBHA show large chemical shift perturbation upon association with the octasaccharide. Molecular dynamics simulations conforming to the multidimensional NMR data revealed key electrostatic and even hydrophobic interactions between the binding partners that may aid in the development of agents targeting the binding event.
Collapse
Affiliation(s)
- Teng-Yi Huang
- Genomics Research Center, Academia Sinica, No. 128, Section 2, Academia Road, Taipei, 115, Taiwan
| | - Deli Irene
- Department of Chemistry, National Dong Hwa University, No. 1, Section 2, Da Hsueh Road, Shoufeng, Hualien, 974, Taiwan
| | - Medel Manuel L Zulueta
- Genomics Research Center, Academia Sinica, No. 128, Section 2, Academia Road, Taipei, 115, Taiwan
| | - Tzu-Jui Tai
- Department of Chemistry, National Dong Hwa University, No. 1, Section 2, Da Hsueh Road, Shoufeng, Hualien, 974, Taiwan
| | - Shih-Han Lain
- Department of Chemistry, National Dong Hwa University, No. 1, Section 2, Da Hsueh Road, Shoufeng, Hualien, 974, Taiwan
| | - Cheng-Po Cheng
- Genomics Research Center, Academia Sinica, No. 128, Section 2, Academia Road, Taipei, 115, Taiwan
| | - Ping-Xi Tsai
- Genomics Research Center, Academia Sinica, No. 128, Section 2, Academia Road, Taipei, 115, Taiwan
| | - Shu-Yi Lin
- Genomics Research Center, Academia Sinica, No. 128, Section 2, Academia Road, Taipei, 115, Taiwan
| | - Zhi-Geng Chen
- Genomics Research Center, Academia Sinica, No. 128, Section 2, Academia Road, Taipei, 115, Taiwan
| | - Chiao-Chu Ku
- Institute of Molecular Biology, Academia Sinica, No. 128, Section 2, Academia Road, Taipei, 115, Taiwan
| | - Chwan-Deng Hsiao
- Institute of Molecular Biology, Academia Sinica, No. 128, Section 2, Academia Road, Taipei, 115, Taiwan
| | - Chia-Lin Chyan
- Department of Chemistry, National Dong Hwa University, No. 1, Section 2, Da Hsueh Road, Shoufeng, Hualien, 974, Taiwan
| | - Shang-Cheng Hung
- Genomics Research Center, Academia Sinica, No. 128, Section 2, Academia Road, Taipei, 115, Taiwan
| |
Collapse
|
25
|
Zimmermann N, Thormann V, Hu B, Köhler AB, Imai-Matsushima A, Locht C, Arnett E, Schlesinger LS, Zoller T, Schürmann M, Kaufmann SH, Wardemann H. Human isotype-dependent inhibitory antibody responses against Mycobacterium tuberculosis. EMBO Mol Med 2016; 8:1325-1339. [PMID: 27729388 PMCID: PMC5090662 DOI: 10.15252/emmm.201606330] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Accumulating evidence from experimental animal models suggests that antibodies play a protective role against tuberculosis (TB). However, little is known about the antibodies generated upon Mycobacterium tuberculosis (MTB) exposure in humans. Here, we performed a molecular and functional characterization of the human B‐cell response to MTB by generating recombinant monoclonal antibodies from single isolated B cells of untreated adult patients with acute pulmonary TB and from MTB‐exposed healthcare workers. The data suggest that the acute plasmablast response to MTB originates from reactivated memory B cells and indicates a mucosal origin. Through functional analyses, we identified MTB inhibitory antibodies against mycobacterial antigens including virulence factors that play important roles in host cell infection. The inhibitory activity of anti‐MTB antibodies was directly linked to their isotype. Monoclonal as well as purified serum IgA antibodies showed MTB blocking activity independently of Fc alpha receptor expression, whereas IgG antibodies promoted the host cell infection. Together, the data provide molecular insights into the human antibody response to MTB and may thereby facilitate the design of protective vaccination strategies.
Collapse
Affiliation(s)
- Natalie Zimmermann
- Research Group Molecular Immunology, Max Planck Institute for Infection Biology, Berlin, Germany.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany.,B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Verena Thormann
- Research Group Molecular Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Bo Hu
- Research Group Molecular Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Anne-Britta Köhler
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Aki Imai-Matsushima
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Camille Locht
- U1019 - UMR 8204 - CIIL - Centre for Infection and Immunity of Lille, University of Lille, Lille, France.,CNRS, UMR 8204, Lille, France.,Inserm, U1019, Lille, France.,CHU Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Eusondia Arnett
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Thomas Zoller
- Department of Infectious Diseases and Respiratory Medicine, Charité University Medical Center, Berlin, Germany
| | - Mariana Schürmann
- Department of Infectious Diseases and Respiratory Medicine, Charité University Medical Center, Berlin, Germany
| | - Stefan He Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hedda Wardemann
- Research Group Molecular Immunology, Max Planck Institute for Infection Biology, Berlin, Germany .,B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
26
|
Zimmermann N, Saiga H, Houthuys E, Moura-Alves P, Koehler A, Bandermann S, Dorhoi A, Kaufmann SHE. Syndecans promote mycobacterial internalization by lung epithelial cells. Cell Microbiol 2016; 18:1846-1856. [PMID: 27279134 DOI: 10.1111/cmi.12627] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 05/11/2016] [Accepted: 06/05/2016] [Indexed: 01/16/2023]
Abstract
Pulmonary tuberculosis (TB) is an airborne disease caused by the intracellular bacterial pathogen Mycobacterium tuberculosis (Mtb). Alveolar epithelial cells and macrophages are the first point of contact for Mtb in the respiratory tract. However, the mechanisms of mycobacterial attachment to, and internalization by, nonprofessional phagocytes, such as epithelial cells, remain incompletely understood. We identified syndecan 4 (Sdc4) as mycobacterial attachment receptor on alveolar epithelial cells. Sdc4 mRNA expression was increased in human and mouse alveolar epithelial cells after mycobacterial infection. Sdc4 knockdown in alveolar epithelial cells or blocking with anti-Sdc4 antibody reduced mycobacterial attachment and internalization. At the molecular level, interactions between epithelial cells and mycobacteria involved host Sdc and the mycobacterial heparin-binding hemagglutinin adhesin. In vivo, Sdc1/Sdc4 double-knockout mice were more resistant to Mtb colonization of the lung. Our work reveals a role for distinct Sdcs in promoting mycobacterial entry into alveolar epithelial cells with impact on outcome of TB disease.
Collapse
Affiliation(s)
- Natalie Zimmermann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany.,Research Group of Molecular Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hiroyuki Saiga
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Erica Houthuys
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Pedro Moura-Alves
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Anne Koehler
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Silke Bandermann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Anca Dorhoi
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
27
|
García B, Merayo-Lloves J, Martin C, Alcalde I, Quirós LM, Vazquez F. Surface Proteoglycans as Mediators in Bacterial Pathogens Infections. Front Microbiol 2016; 7:220. [PMID: 26941735 PMCID: PMC4764700 DOI: 10.3389/fmicb.2016.00220] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/10/2016] [Indexed: 11/18/2022] Open
Abstract
Infectious diseases remain an important global health problem. The interaction of a wide range of pathogen bacteria with host cells from many different tissues is frequently mediated by proteoglycans. These compounds are ubiquitous complex molecules which are not only involved in adherence and colonization, but can also participate in other steps of pathogenesis. To overcome the problem of microbial resistance to antibiotics new therapeutic agents could be developed based on the characteristics of the interaction of pathogens with proteoglycans.
Collapse
Affiliation(s)
- Beatriz García
- Department of Functional Biology, Microbiology, Faculty of Medicine, University of OviedoOviedo, Spain; Instituto Oftalmológico Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de OviedoOviedo, Spain
| | - Jesús Merayo-Lloves
- Instituto Oftalmológico Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de OviedoOviedo, Spain; Department of Surgery, University of OviedoOviedo, Spain
| | - Carla Martin
- Department of Functional Biology, Microbiology, Faculty of Medicine, University of OviedoOviedo, Spain; Instituto Oftalmológico Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de OviedoOviedo, Spain
| | - Ignacio Alcalde
- Instituto Oftalmológico Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de Oviedo Oviedo, Spain
| | - Luis M Quirós
- Department of Functional Biology, Microbiology, Faculty of Medicine, University of OviedoOviedo, Spain; Instituto Oftalmológico Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de OviedoOviedo, Spain
| | - Fernando Vazquez
- Department of Functional Biology, Microbiology, Faculty of Medicine, University of OviedoOviedo, Spain; Instituto Oftalmológico Fernández-Vega, Fundación de Investigación Oftalmológica, Universidad de OviedoOviedo, Spain; Service of Microbiology, Central University Hospital of AsturiasOviedo, Spain
| |
Collapse
|
28
|
Pavlicek RL, Fine-Coulson K, Gupta T, Quinn FD, Posey JE, Willby M, Castro-Garza J, Karls RK. Rv3351c, a Mycobacterium tuberculosis gene that affects bacterial growth and alveolar epithelial cell viability. Can J Microbiol 2015; 61:938-47. [PMID: 26492080 DOI: 10.1139/cjm-2015-0528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Despite the interactions known to occur between various lower respiratory tract pathogens and alveolar epithelial cells (AECs), few reports examine factors influencing the interplay between Mycobacterium tuberculosis bacilli and AECs during infection. Importantly, in vitro studies have demonstrated that the M. tuberculosis hbha and esxA gene products HBHA and ESAT6 directly or indirectly influence AEC survival. In this report, we identify Rv3351c as another M. tuberculosis gene that impacts the fate of both the pathogen and AEC host. Intracellular replication of an Rv3351c mutant in the human AEC type II pneumocyte cell line A549 was markedly reduced relative to the complemented mutant and parent strain. Deletion of Rv3351c diminished the release of lactate dehydrogenase and decreased uptake of trypan blue vital stain by host cells infected with M. tuberculosis bacilli, suggesting attenuated cytotoxic effects. Interestingly, an isogenic hbha mutant displayed reductions in AEC killing similar to those observed for the Rv3351c mutant. This opens the possibility that multiple M. tuberculosis gene products interact with AECs. We also observed that Rv3351c aids intracellular replication and survival of M. tuberculosis in macrophages. This places Rv3351c in the same standing as HBHA and ESAT6, which are important factors in AECs and macrophages. Defining the mechanism(s) by which Rv3351c functions to aid pathogen survival within the host may lead to new drug or vaccine targets.
Collapse
Affiliation(s)
- Rebecca L Pavlicek
- a Department of Infectious Diseases, University of Georgia, 220 Riverbend Road, Athens, GA 30602, USA
| | - Kari Fine-Coulson
- a Department of Infectious Diseases, University of Georgia, 220 Riverbend Road, Athens, GA 30602, USA
| | - Tuhina Gupta
- a Department of Infectious Diseases, University of Georgia, 220 Riverbend Road, Athens, GA 30602, USA
| | - Frederick D Quinn
- a Department of Infectious Diseases, University of Georgia, 220 Riverbend Road, Athens, GA 30602, USA
| | - James E Posey
- b Mycobacteriology Laboratory Branch, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Melisa Willby
- b Mycobacteriology Laboratory Branch, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Jorge Castro-Garza
- c Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey, N.L. 64720, Mexico
| | - Russell K Karls
- a Department of Infectious Diseases, University of Georgia, 220 Riverbend Road, Athens, GA 30602, USA
| |
Collapse
|
29
|
Dando SJ, Mackay-Sim A, Norton R, Currie BJ, St John JA, Ekberg JAK, Batzloff M, Ulett GC, Beacham IR. Pathogens penetrating the central nervous system: infection pathways and the cellular and molecular mechanisms of invasion. Clin Microbiol Rev 2014; 27:691-726. [PMID: 25278572 PMCID: PMC4187632 DOI: 10.1128/cmr.00118-13] [Citation(s) in RCA: 300] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The brain is well protected against microbial invasion by cellular barriers, such as the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB). In addition, cells within the central nervous system (CNS) are capable of producing an immune response against invading pathogens. Nonetheless, a range of pathogenic microbes make their way to the CNS, and the resulting infections can cause significant morbidity and mortality. Bacteria, amoebae, fungi, and viruses are capable of CNS invasion, with the latter using axonal transport as a common route of infection. In this review, we compare the mechanisms by which bacterial pathogens reach the CNS and infect the brain. In particular, we focus on recent data regarding mechanisms of bacterial translocation from the nasal mucosa to the brain, which represents a little explored pathway of bacterial invasion but has been proposed as being particularly important in explaining how infection with Burkholderia pseudomallei can result in melioidosis encephalomyelitis.
Collapse
Affiliation(s)
- Samantha J Dando
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Alan Mackay-Sim
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Robert Norton
- Townsville Hospital, Townsville, Queensland, Australia
| | - Bart J Currie
- Menzies School of Health Research and Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - James A St John
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Jenny A K Ekberg
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Michael Batzloff
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Glen C Ulett
- School of Medical Science and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia
| | - Ifor R Beacham
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
30
|
Govender VS, Ramsugit S, Pillay M. Mycobacterium tuberculosis adhesins: potential biomarkers as anti-tuberculosis therapeutic and diagnostic targets. Microbiology (Reading) 2014; 160:1821-1831. [DOI: 10.1099/mic.0.082206-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Adhesion to host cells is a precursor to host colonization and evasion of the host immune response. Conversely, it triggers the induction of the immune response, a process vital to the host’s defence against infection. Adhesins are microbial cell surface molecules or structures that mediate the attachment of the microbe to host cells and thus the host–pathogen interaction. They also play a crucial role in bacterial aggregation and biofilm formation. In this review, we discuss the role of adhesins in the pathogenesis of the aetiological agent of tuberculosis, Mycobacterium tuberculosis. We also provide insight into the structure and characteristics of some of the characterized and putative M. tuberculosis adhesins. Finally, we examine the potential of adhesins as targets for the development of tuberculosis control strategies.
Collapse
Affiliation(s)
- Viveshree S. Govender
- Medical Microbiology and Infection Control, University of KwaZulu-Natal, Durban, South Africa
| | - Saiyur Ramsugit
- Medical Microbiology and Infection Control, University of KwaZulu-Natal, Durban, South Africa
| | - Manormoney Pillay
- Medical Microbiology and Infection Control, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
31
|
Vir P, Gupta D, Agarwal R, Verma I. Interaction of alveolar epithelial cells with CFP21, a mycobacterial cutinase-like enzyme. Mol Cell Biochem 2014; 396:187-99. [PMID: 25091806 DOI: 10.1007/s11010-014-2154-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 07/14/2014] [Indexed: 12/17/2022]
Abstract
Mycobacterium tuberculosis (M. tb), an intracellular pathogen, has the ability to infect alveolar epithelial cells (AEC) also in addition to alveolar macrophages. The virulence of M. tb is attributed to proteins encoded by genomic regions of deletion (RD) and till date 16 such regions (RD1-RD16) have been identified. Culture filtrate protein 21 (CFP21), a RD2 secretory protein, is a cutinase-like enzyme that possesses esterase and lipolytic activity. It is hypothesized that CFP21 could be playing a role in M. tb virulence by disrupting the host cell integrity. In this study, recombinant CFP21 was expressed and purified. The in vitro exposure of type I (WI26) and type II (A549) AEC to CFP21 resulted in a significant decline in their cellular viability by inducing cell apoptosis. However, the cytotoxic effects were more pronounced in WI26 cells than in A549 cells. The analysis of immune responses in CFP21-treated AEC exhibited significant production of reactive oxygen species and anti-inflammatory cytokine TGF-β which indicated oxidative stress-mediated cell death. These results show that CFP21 could play an important role in M. tb pathogenesis by disrupting the host alveolar barrier and thereby facilitating mycobacterial dissemination.
Collapse
Affiliation(s)
- Pooja Vir
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Sector-12, Chandigarh, 160012, India
| | | | | | | |
Collapse
|
32
|
Wu FLL, Yeh TH, Chen YL, Chiu YC, Cheng JC, Wei MF, Shen LJ. Intracellular delivery of recombinant arginine deiminase (rADI) by heparin-binding hemagglutinin adhesion peptide restores sensitivity in rADI-resistant cancer cells. Mol Pharm 2014; 11:2777-86. [PMID: 24950134 DOI: 10.1021/mp5001372] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recombinant arginine deiminase (rADI) has been used in clinical trials for arginine-auxotrophic cancers. However, the emergence of rADI resistance, due to the overexpression of argininosuccinate synthetase (AS), has introduced an obstacle in its clinical application. Here, we have proposed a strategy for the intracellular delivery of rADI, which depletes both extracellular and intracellular arginine, to restore the sensitivity of rADI-resistant cancer cells. In this study, the C terminus of heparin-binding hemagglutinin adhesion protein from Mycobacterium tuberculosis (HBHAc), which contains 23 amino acids, was used to deliver rADI into rADI-resistant human breast adenocarcinoma cells (MCF-7). Chemical conjugates (l- and d-HBHAc-SPDP-rADI) and a recombinant fusion protein (rHBHAc-ADI) were produced. l- and d-HBHAc-SPDP-rADI showed a significantly higher cellular uptake of rADI by MCF-7 cells compared to that of rADI alone. Cell viability was significantly decreased in a dose-dependent manner in response to l- and d-HBHAc-SPDP-rADI treatments. In addition, the ratio of intracellular concentration of citrulline to arginine in cells treated with l- and d-HBHAc-SPDP-rADI was significantly increased by 1.4- and 1.7-fold, respectively, compared with that obtained in cells treated with rADI alone (p < 0.001). Similar results were obtained with the recombinant fusion protein rHBHAc-ADI. Our study demonstrates that the increased cellular uptake of rADI by HBHAc modification can restore the sensitivity of rADI treatment in MCF-7 cells. rHBHAc-ADI may represent a novel class of antitumor enzyme with an intracellular mechanism that is independent of AS expression.
Collapse
Affiliation(s)
- Fe-Lin Lin Wu
- School of Pharmacy and ‡Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University , Taipei 10050, Taiwan
| | | | | | | | | | | | | |
Collapse
|
33
|
Lutay N, Håkansson G, Alaridah N, Hallgren O, Westergren-Thorsson G, Godaly G. Mycobacteria bypass mucosal NF-kB signalling to induce an epithelial anti-inflammatory IL-22 and IL-10 response. PLoS One 2014; 9:e86466. [PMID: 24489729 PMCID: PMC3904915 DOI: 10.1371/journal.pone.0086466] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 12/13/2013] [Indexed: 01/20/2023] Open
Abstract
The mechanisms by which mycobacteria subvert the inflammatory defence to establish chronic infection remain an unresolved question in the pathogenesis of tuberculosis. Using primary epithelial cells, we have analysed mycobacteria induced epithelial signalling pathways from activation of TLRs to cytokine secretion. Mycobacterium bovis bacilli Calmette-Guerin induced phosphorylation of glycogen synthase kinase (GSK)3 by PI3K-Akt in the signalling pathway downstream of TLR2 and TLR4. Mycobacteria did not suppress NF-κB by activating the peroxisome proliferator-activated receptor γ. Instead the pro-inflammatory NF-κB was bypassed by mycobacteria induced GSK3 inhibition that promoted the anti-inflammatory transcription factor CREB. Mycobacterial infection did not thus induce mucosal pro-inflammatory response as measured by TNFα and IFNγ secretion, but led to an anti-inflammatory IL-10 and IL-22 production. Apart from CREB, MAP3Ks p38 and ERK1/2 activated the transcription factor AP-1 leading to IL-6 production. Interestingly, blocking of TLR4 before infection decreased epithelial IL-6 secretion, but increased the CREB-activated IL-10 production. Our data indicate that mycobacteria suppress epithelial pro-inflammatory production by suppressing NF-κB activation thereby shifting the infection towards an anti-inflammatory state. This balance between the host immune response and the pathogen could determine the outcome of infection.
Collapse
Affiliation(s)
- Nataliya Lutay
- Division of Laboratory Medicine, Department of MIG, Lund University, Lund, Sweden
| | - Gisela Håkansson
- Division of Laboratory Medicine, Department of MIG, Lund University, Lund, Sweden
| | - Nader Alaridah
- Division of Laboratory Medicine, Department of MIG, Lund University, Lund, Sweden
| | - Oskar Hallgren
- Division of Clinical Sciences, Department of Respiratory Medicine and Allergology, Lund University, Lund, Sweden
| | - Gunilla Westergren-Thorsson
- Division of Vascular- and Respiratory Research Unit of Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Gabriela Godaly
- Division of Laboratory Medicine, Department of MIG, Lund University, Lund, Sweden
| |
Collapse
|
34
|
Stylianou E, Diogo GR, Pepponi I, Dolleweerd C, Arias MA, Locht C, Rider CC, Sibley L, Cutting SM, Loxley A, Ma JK, Reljic R. Mucosal delivery of antigen‐coated nanoparticles to lungs confers protective immunity against tuberculosis infection in mice. Eur J Immunol 2013; 44:440-9. [DOI: 10.1002/eji.201343887] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/25/2013] [Accepted: 11/05/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Elena Stylianou
- Infection and Immunity Research CentreSt George's University of London London UK
- Jenner InstituteJohn Radcliffe HospitalUniversity of Oxford Oxford UK
| | - Gil R. Diogo
- Infection and Immunity Research CentreSt George's University of London London UK
| | - Ilaria Pepponi
- Infection and Immunity Research CentreSt George's University of London London UK
- Jenner InstituteJohn Radcliffe HospitalUniversity of Oxford Oxford UK
| | - Craig Dolleweerd
- Infection and Immunity Research CentreSt George's University of London London UK
| | - Mauricio A. Arias
- Infection and Immunity Research CentreSt George's University of London London UK
| | - Camille Locht
- Institute Pasteur de Lille Lille France
- Inserm U1019 Lille France
- CNRS UMR8204 Lille France
- Université Lille Nord de France Lille France
| | | | - Laura Sibley
- School of Biological SciencesRoyal Holloway University of London Egham UK
| | - Simon M. Cutting
- School of Biological SciencesRoyal Holloway University of London Egham UK
| | - Andrew Loxley
- Particle Sciences, Inc Pennsylvania, Bethlehem PA USA
| | - Julian K.C. Ma
- Infection and Immunity Research CentreSt George's University of London London UK
| | - Rajko Reljic
- Infection and Immunity Research CentreSt George's University of London London UK
| |
Collapse
|
35
|
Dias AA, Raze D, de Lima CS, Marques MADM, Drobecq H, Debrie AS, Ribeiro-Guimarães ML, Biet F, Pessolani MCV. Mycobacterial laminin-binding histone-like protein mediates collagen-dependent cytoadherence. Mem Inst Oswaldo Cruz 2013; 107 Suppl 1:174-82. [PMID: 23283469 DOI: 10.1590/s0074-02762012000900025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Accepted: 07/17/2012] [Indexed: 11/22/2022] Open
Abstract
When grown in the presence of exogenous collagen I, Mycobacterium bovis BCG was shown to form clumps. Scanning electron microscopy examination of these clumps revealed the presence of collagen fibres cross-linking the bacilli. Since collagen is a major constituent of the eukaryotic extracellular matrices, we assayed BCG cytoadherence in the presence of exogenous collagen I. Collagen increased the interaction of the bacilli with A549 type II pneumocytes or U937 macrophages, suggesting that BCG is able to recruit collagen to facilitate its attachment to host cells. Using an affinity chromatography approach, we have isolated a BCG collagen-binding protein corresponding to the previously described mycobacterial laminin-binding histone-like protein (LBP/Hlp), a highly conserved protein associated with the mycobacterial cell wall. Moreover, Mycobacterium leprae LBP/Hlp, a well-characterized adhesin, was also able to bind collagen I. Finally, using recombinant fragments of M. leprae LBP/Hlp, we mapped the collagen-binding activity within the C-terminal domain of the adhesin. Since this protein was already shown to be involved in the recognition of laminin and heparan sulphate-containing proteoglycans, the present observations reinforce the adhesive activities of LBP/Hlp, which can be therefore considered as a multifaceted mycobacterial adhesin, playing an important role in both leprosy and tuberculosis pathogenesis.
Collapse
Affiliation(s)
- André Alves Dias
- Laboratório de Microbiologia Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Vir P, Gupta D, Agarwal R, Verma I. Immunomodulation of alveolar epithelial cells by Mycobacterium tuberculosis phosphatidylinositol mannosides results in apoptosis. APMIS 2013; 122:268-82. [PMID: 23919648 DOI: 10.1111/apm.12141] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/22/2013] [Indexed: 11/27/2022]
Abstract
During intracellular residence in macrophages, mycobacterial lipids, namely phosphatidylinositol mannosides (PIM) and lipoarabinomannans, are expelled in the lung milieu to interact with host cells. PIM include a group of essential lipid components of Mycobacterium tuberculosis (M. tb) cell wall. Given that PIM function as mycobacterial adhesins for binding to host cells, the present study explored the consequences of interaction of M. tb PIM with alveolar epithelial cells (AEC). A 24-h PIM exposure at a concentration of 10 μg/mL to AEC conferred cytolysis to AEC via induction of apoptosis, suggesting their potential to alter alveolar epithelium integrity. The results also reflected that type I like AEC are more sensitive to cytolysis than type II AEC. PIM-treated AEC exhibited significant production of reactive oxygen species (ROS) and an immunosuppressive cytokine transforming growth factor-β (TGF-β) in the culture supernatants. Although AEC displayed constitutive mRNA transcripts for toll-like receptors (TLR2 and 4) as well as chemokines (IL-8 and MCP-1), no significant change in their expression was observed upon PIM treatment. Collectively, these results offer insights into PIM potential as M. tb virulence factor that might promote mycobacterial dissemination by causing cytolysis of AEC via increased production of ROS and TGF-β.
Collapse
Affiliation(s)
- Pooja Vir
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | | | | |
Collapse
|
37
|
Verdurmen WPR, Wallbrecher R, Schmidt S, Eilander J, Bovee-Geurts P, Fanghänel S, Bürck J, Wadhwani P, Ulrich AS, Brock R. Cell surface clustering of heparan sulfate proteoglycans by amphipathic cell-penetrating peptides does not contribute to uptake. J Control Release 2013; 170:83-91. [PMID: 23669260 DOI: 10.1016/j.jconrel.2013.05.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/27/2013] [Accepted: 05/02/2013] [Indexed: 10/26/2022]
Abstract
For arginine-rich cell-penetrating peptides (CPPs), an association with heparan sulfate (HS) chains is considered the first step in the stimulation of uptake for many cells. Much less is known about the role of HS chains in the cell-association and internalization of arginine-free amphipathic CPP such as transportan-10 (TP10). Here, we report that various TP10 analogs differ in their capacity to accumulate on HS-rich plasma membranes in an HS-dependent manner. No accumulation was observed on HS-poor plasma membranes or when HS was removed by enzymatic cleavage. The TP10 analog that strongly clustered on the cell surface, also showed a pronounced capacity to form clusters with HS chains in solution. However, aggregation occurred in a thermodynamically different way compared to the interaction of arginine-rich CPP with HS. To monitor the impact of the peptide on the aggregation of the glycocalyx by time-lapse microscopy, sialic acids were visualized by metabolic labeling using copper-free click chemistry to attach fluorophores to metabolically incorporated azido sugars. Strikingly, a highly enhanced HS-mediated accumulation on the plasma membrane of a particular TP10 analog did not correlate with a better uptake. These findings illustrate that the mode of interaction between cell-penetrating peptides and HS chains has important functional consequences regarding peptide internalization and that there is no direct coupling of interaction, accumulation and uptake.
Collapse
Affiliation(s)
- Wouter P R Verdurmen
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Post 286, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Syndecan-1 and heparanase: potential markers for activity evaluation and differential diagnosis of Crohn's disease. Inflamm Bowel Dis 2013; 19:1025-33. [PMID: 23511033 DOI: 10.1097/mib.0b013e318280298f] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Syndecan-1 (SDC1) and its endo-beta-D-glucuronidase heparanase (HPA) are implicated in the maintenance of intestinal barrier function, but their detailed functions in Crohn's disease (CD) are not fully investigated. The aim of this study was to determine alteration patterns of SDC1 and HPA and their potential roles in evaluating disease activity and differentiating CD from intestinal tuberculosis (ITB). METHODS Tissue and serum specimens were obtained from 89 patients, including 15 patients with functional bowel disorders, 18 active patients with ITB, and 56 patients with CD (remission = 19, active = 37). Basic clinical data were collected and routine blood tests were analyzed. SDC1 and HPA were measured by immunohistochemistry, enzyme-linked immunosorbent assay, reverse transcriptase polymerase chain reaction, and western blot. Colonic epithelial cells were incubated with recombinant HPA, tumor necrosis factor alpha (TNF-α), and mycobacterium tuberculosis culture filtrate protein to detect the alterations of SDC1 and HPA. RESULTS In the CD group, SDC1 was significantly decreased in mucosa and increased in serum, whereas HPA level in both were elevated. Such alterations were associated with clinicopathological features representing disease activity and injury severity and were not available in functional bowel disorder and ITB groups. Recombinant HPA incubation increased soluble SDC1 in culture supernatants (P = 2 × 10(-4)), and low-dose TNF-α effectively enhanced HPA's activity (P = 3 × 10(-6)). Exogenous TNF-α destroyed cellular SDC1 and raised HPA expressions dose dependently, whereas mycobacterium tuberculosis culture filtrate protein showed no effects. CONCLUSIONS Unique alterations of SDC1 and HPA are shown in both patients with CD and in vitro model. The results indicate SDC1 and HPA are potential markers for CD in evaluating its disease activity and differentiating it from ITB.
Collapse
|
39
|
How cationic lipids transfer nucleic acids into cells and across cellular membranes: Recent advances. J Control Release 2013; 166:46-56. [DOI: 10.1016/j.jconrel.2012.12.014] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 12/06/2012] [Accepted: 12/10/2012] [Indexed: 12/16/2022]
|
40
|
Adlakha N, Vir P, Verma I. Effect of mycobacterial secretory proteins on the cellular integrity and cytokine profile of type II alveolar epithelial cells. Lung India 2012; 29:313-8. [PMID: 23243342 PMCID: PMC3519014 DOI: 10.4103/0970-2113.102796] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Pulmonary tuberculosis (TB) is caused by Mycobacterium tuberculosis (M. tb). In lungs, alveolar macrophages and type II alveolar epithelial cells serve as a replicative niche for this pathogen. Secretory proteins released by actively replicating tubercle bacilli are known to interact with host cells at the initial stages of infection. To understand the role of these cells in TB pathogenesis, it is important to identify the mycobacterial components involved in interaction with alveolar epithelial cells. Materials and Methods: We fractionated the whole secretory proteome of M. tb H37Rv into 10 narrow molecular mass fractions (A1-A10; <20 kDa to >90 kDa) that were studied for their binding potential with A549; type II alveolar epithelial cell line. We also studied the consequences of this interaction in terms of change in epithelial cell viability by MTT assay and cytokine release by ELISA. Results: Our results show that several mycobacterial proteins bind and confer cytolysis in epithelial cells. Amongst all the fractions, proteins ranging from 35-45 kDa (A5) exhibited highest binding to A549 cells with a consequence of cytolysis of these cells. This fraction (A5) also led to release of various cytokines important in anti-mycobacterial immunity. Conclusion: Fraction A5 (35-45 kDa) of mycobacterial secretory proteome play an important role in mediating M. tb interaction with type II alveolar epithelial cells with the consequences detrimental for the TB pathogenesis. Further studies are being carried out to identify the candidate proteins from this region.
Collapse
Affiliation(s)
- Nidhi Adlakha
- Department of Biochemistry, TB Immunology and Cell Biology Lab, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | | |
Collapse
|
41
|
Håkansson G, Lutay N, Andersson M, Hallgren O, Westergren-Thorsson G, Svensson M, Godaly G. Epithelial G protein-coupled receptor kinases regulate the initial inflammatory response during mycobacterial infection. Immunobiology 2012; 218:984-94. [PMID: 23312955 DOI: 10.1016/j.imbio.2012.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Revised: 11/12/2012] [Accepted: 11/14/2012] [Indexed: 01/04/2023]
Abstract
The interaction between mycobacteria and epithelium is unexplored, but may determine the outcome of the infection. We have analyzed the role of two G protein-coupled receptors, CXCR1 and CXCR2 that are important regulators of many pulmonary diseases. We found that mycobacteria significantly increased the expression of both CXCR1 and CXCR2 on alveolar epithelial cells and both receptors were found to be important for neutrophil diapedesis across primary endothelial cells towards infected mucosa. Mycobacteria, lipoarabinomannan or 19-kDa glycolipoprotein up-regulated the inhibitory G protein-coupled receptor kinase (GRK)2, while GRK3 was less affected. Mycobacteria-induced GRK2 up-regulation decreased chemokine transcription and secretion thereby affecting the neutrophil recruitment to infected mucosa. These events were completely abolished by blocking these receptors prior to infection as the blocking increased epithelial immune responses. We have identified novel interactions occurring in the initial phase of mycobacterial infections by which mycobacterial manipulate epithelial inflammatory responses.
Collapse
Affiliation(s)
- Gisela Håkansson
- Department of MIG, Division of Laboratory Medicine, Lund University, Lund, Sweden
| | | | | | | | | | | | | |
Collapse
|
42
|
Zaman A. Docking studies and network analyses reveal capacity of compounds from Kandelia rheedii to strengthen cellular immunity by interacting with host proteins during tuberculosis infection. Bioinformation 2012; 8:1012-20. [PMID: 23275699 PMCID: PMC3524883 DOI: 10.6026/97320630081012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Accepted: 10/15/2012] [Indexed: 01/09/2023] Open
Abstract
Kandelia rheedii (locally known as Guria or Rasunia), widely found and used in Indian subcontinent, is a well-known herbal cure to tuberculosis. However, neither the mechanism nor the active components of the plant extract responsible for mediating this action has yet been confirmed. Here in this study, molecular interactions of three compounds (emodin, fusaric acid and skyrin) from the plant extract with the host protein targets (casein kinase (CSNK), estrogen receptor (ERBB), dopamine β-hydroxylase (DBH) and glucagon receptor (Gcgr)) has been found. These protein targets are known to be responsible for strengthening cellular immunity against Mycobacteria tuberculosis. The specific interactions of these three compounds with the respective protein targets have been discussed here. The insights from study should further help us designing molecular medicines against tuberculosis.
Collapse
Affiliation(s)
- Aubhishek Zaman
- Molecular Biology Laboratory, Department of Biochemistry and Molecular Biology and Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka-1000, Bangladesh
| |
Collapse
|
43
|
ur Rehman Z, Sjollema KA, Kuipers J, Hoekstra D, Zuhorn IS. Nonviral gene delivery vectors use syndecan-dependent transport mechanisms in filopodia to reach the cell surface. ACS NANO 2012; 6:7521-7532. [PMID: 22857607 DOI: 10.1021/nn3028562] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Lipoplexes and polyplexes, that is, assemblies of cationic lipids and polymers with nucleic acids, respectively, are popular nanocarriers for delivery of genes or siRNA into cells for therapeutic or cell biological purposes. Although endocytosis represents a major mechanism for their cellular entry, very little is known about parameters that govern early events in the initial interaction of such delivery devices with the cell surface. Here, we demonstrate that prior to entry, poly- and lipoplexes are captured by thin, actin-rich filopodial extensions, protruding from the cell surface. Subsequent additional recruitment and local clustering of filopodia-localized syndecans, presumably driven by multivalent interactions with the polycationic nanocarriers, appear instrumental in their processing to the cell body. Detailed microscopic analyses reveal that the latter relies on either directional surfing along or retraction of the filopodia. By interfering with actin polymerization or inhibiting the motor protein myosin II, localized at the base of filopodia, our data reveal that the binding of the nanocarriers to and subsequent clustering of syndecans initiates actin retrograde flow, which moves the syndecan-bound nanocarriers to the cell body. At the present experimental conditions, inhibition of this process inhibits nanocarrier-mediated transfection by 50-90%. The present findings add novel insight to our understanding of the mechanism of nanocarrier-cell surface interaction, which may be instrumental in further improving delivery efficiency. In addition, the current experimental approach may also be of relevance to improving our understanding of cellular infection by viruses and pathogenic bacteria, given a striking parallel in filopodia-mediated processing of these infectious particles and nanocarriers.
Collapse
Affiliation(s)
- Zia ur Rehman
- University Medical Center Groningen, University of Groningen, Department of Cell Biology, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
44
|
Risk stratification of latent tuberculosis defined by combined interferon gamma release assays. PLoS One 2012; 7:e43285. [PMID: 22912846 PMCID: PMC3422279 DOI: 10.1371/journal.pone.0043285] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 07/23/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Most individuals infected with Mycobacterium tuberculosis develop latent tuberculosis infection (LTBI). Some may progress to active disease and would benefit from preventive treatment yet no means currently exists to predict who will reactivate. Here, we provide an approach to stratify LTBI based on IFN-γ responses to two antigens, the recombinant Early-Secreted Antigen Target-6 (rESAT-6) and the latency antigen Heparin-Binding Haemagglutinin (HBHA). METHODS We retrospectively analyzed results from in-house IFN-γ-release assays with HBHA (HBHA-IGRA) and rESAT-6 (rESAT-6-IGRA) performed during a 12-year period on serial blood samples (3 to 9) collected from 23 LTBI subjects in a low-TB incidence country. Both the kinetics of the absolute IFN-γ concentrations secreted in response to each antigen and the dynamics of HBHA/rESAT-6-induced IFN-γ concentrations ratios were examined. RESULTS This analysis allowed the identification among the LTBI subjects of three major groups. Group A featured stable HBHA and rESAT-6-IGRA profiles with an HBHA/rESAT-6 ratio persistently higher than 1, and with high HBHA- and usually negative rESAT-6-IGRA responses throughout the study. Group B had changing HBHA/rESAT-6 ratios fluctuating from 0.0001 to 10,000, with both HBHA and rESAT-6 responses varying over time at least once during the follow-up. Group C was characterized by a progressive disappearance of all responses. CONCLUSIONS By combining the measures of IFN-γ concentrations secreted in response to an early and a latency antigens, LTBI subjects can be stratified into different risk groups. We propose that disappearing responses indicate cure, that persistent responses to HBHA with HBHA/rESAT-6 ratios ≥ 1 represent stable LTBI subjects, whereas subjects with ratios varying from >1 to <1 should be closely monitored as they may represent the highest-risk group, as illustrated by a case report, and should therefore be prioritized for preventive treatment.
Collapse
|
45
|
Castro-Garza J, Swords WE, Karls RK, Quinn FD. Dual mechanism for Mycobacterium tuberculosis cytotoxicity on lung epithelial cells. Can J Microbiol 2012; 58:909-16. [PMID: 22720783 DOI: 10.1139/w2012-067] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mycobacterium tuberculosis strains CDC1551 and Erdman were used to assess cytotoxicity in infected A549 human alveolar epithelial cell monolayers. Strain CDC1551 was found to induce qualitatively greater disruption of A549 monolayers than was strain Erdman, although total intracellular and cell-associated bacterial growth rates over the course of the infections were not significantly different. Cell-free culture supernatants from human monocytic cells infected with either of the 2 M. tuberculosis strains produced a cytotoxic effect on A549 cells, correlating with the amount of tumor necrosis factor alpha (TNF-α) released by the infected monocytes. The addition of TNF-α-neutralizing antibodies to the supernatants from infected monocyte cultures did prevent the induction of a cytotoxic effect on A549 cells overlaid with this mixture but did not prevent the death of epithelial cells when added prior to infection with M. tuberculosis bacilli. Thus, these data agree with previous observations that lung epithelial cells infected with M. tuberculosis bacilli are rapidly killed in vitro. In addition, the data indicate that some of the observed epithelial cell killing may be collateral damage; the result of TNF-α released from M. tuberculosis-infected monocytes.
Collapse
Affiliation(s)
- Jorge Castro-Garza
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey, Mexico.
| | | | | | | |
Collapse
|
46
|
Lebrun P, Raze D, Fritzinger B, Wieruszeski JM, Biet F, Dose A, Carpentier M, Schwarzer D, Allain F, Lippens G, Locht C. Differential contribution of the repeats to heparin binding of HBHA, a major adhesin of Mycobacterium tuberculosis. PLoS One 2012; 7:e32421. [PMID: 22403657 PMCID: PMC3293801 DOI: 10.1371/journal.pone.0032421] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 01/26/2012] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Tuberculosis remains one of the most important causes of global mortality and morbidity, and the molecular mechanisms of the pathogenesis are still incompletely understood. Only few virulence factors of the causative agent Mycobacterium tuberculosis are known. One of them is the heparin-binding haemagglutinin (HBHA), an important adhesin for epithelial cells and an extrapulmonary dissemination factor. HBHA mediates mycobacterial adherence to epithelial cells via the interactions of its C-terminal, lysine rich repeat domain with sulfated glycoconjugates on the surface of epithelial cells. METHODOLOGY/PRINCIPAL FINDINGS Using defined heparin sulfate (HS) analogs, we determined the minimal heparin fragment length for HBHA binding and structural adaptations of the HBHA heparin-binding domain (HBD) upon binding to heparin. The NMR studies show significant shifts of all residues in the HBD upon interaction with heparin, with stronger shifts in the last repeats compared to the upstream repeats, and indicated that the HS fragments with 14 sugar units cover the entire C-terminal lysine-rich domain of HBHA. The differential implication of the repeats is determined by the relative position of prolines and lysines within each repeat, and may contribute to binding specificity. GAG binding induces a non-homogeneous structural rearrangement in the HBD, with stabilization of a nascent α-helix only in the last penta-repeats. CONCLUSION/SIGNIFICANCE Mycobacterial HBHA undergoes structural adaptation upon interaction with GAGs, which is likely involved in binding specificities of the adhesin, and mycobacterial pathogens may use HBD polymorphisms for host or organ specificity. Further studies will aim at decoding the complementarity between HBD repeats and HS sequence.
Collapse
Affiliation(s)
- Pierre Lebrun
- INSERM U1019, Lille, France
- CNRS UMR 8204, Lille, France
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
- Univ Lille Nord de France, Lille, France
| | - Dominique Raze
- INSERM U1019, Lille, France
- CNRS UMR 8204, Lille, France
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
- Univ Lille Nord de France, Lille, France
| | - Bernd Fritzinger
- Univ Lille Nord de France, Lille, France
- CNRS UMR8576 Structural and Functional Glycobiology, Villeneuve d'Ascq, France
| | - Jean-Michel Wieruszeski
- Univ Lille Nord de France, Lille, France
- CNRS UMR8576 Structural and Functional Glycobiology, Villeneuve d'Ascq, France
| | - Franck Biet
- UR1282, Infectiologie Animale, Sante' Publique (IASP-311), INRA, Nouzilly, France
| | - Alexander Dose
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Mathieu Carpentier
- Univ Lille Nord de France, Lille, France
- CNRS UMR8576 Structural and Functional Glycobiology, Villeneuve d'Ascq, France
| | - Dirk Schwarzer
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Fabrice Allain
- Univ Lille Nord de France, Lille, France
- CNRS UMR8576 Structural and Functional Glycobiology, Villeneuve d'Ascq, France
| | - Guy Lippens
- Univ Lille Nord de France, Lille, France
- CNRS UMR8576 Structural and Functional Glycobiology, Villeneuve d'Ascq, France
| | - Camille Locht
- INSERM U1019, Lille, France
- CNRS UMR 8204, Lille, France
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
- Univ Lille Nord de France, Lille, France
| |
Collapse
|
47
|
Esposito C, Cantisani M, D'Auria G, Falcigno L, Pedone E, Galdiero S, Berisio R. Mapping key interactions in the dimerization process of HBHA fromMycobacterium tuberculosis, insights into bacterial agglutination. FEBS Lett 2012; 586:659-67. [DOI: 10.1016/j.febslet.2012.01.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 12/28/2011] [Accepted: 01/18/2012] [Indexed: 11/16/2022]
|
48
|
Heparin-binding hemagglutinin HBHA from Mycobacterium tuberculosis affects actin polymerisation. Biochem Biophys Res Commun 2011; 410:339-44. [DOI: 10.1016/j.bbrc.2011.05.159] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 05/31/2011] [Indexed: 11/23/2022]
|
49
|
Triggered Mycobacterium tuberculosis heparin-binding hemagglutinin adhesin folding and dimerization. J Bacteriol 2011; 193:2089-96. [PMID: 21398546 DOI: 10.1128/jb.01231-10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The heparin-binding hemagglutinin adhesin (HBHA) is a surface adhesin on the human pathogen Mycobacterium tuberculosis. Previously, it has been shown that HBHA exists as a dimer in solution. We investigated the detailed nature of this dimer using circular dichroism spectroscopy and analytical ultracentrifugation techniques. We demonstrate that the heparan sulfate (HS) binding region does not play a role in dimerization in solution, while the linker region between the predicted N-terminal coiled-coil and the C-terminal HS binding region does affect dimer stability. The majority of contacts responsible for dimerization, folding, and stability lie within the predicted coiled-coil region of HBHA, while the N-terminal helix preceding the coiled-coil appears to trigger the folding and dimerization of HBHA. Constructs lacking this initial helix or containing site-specific mutations produce nonhelical monomers in solution. Thus, we show that HBHA dimerization and folding are linked and that the N-terminal region of this cell surface adhesin triggers the formation of an HBHA coiled-coil dimer.
Collapse
|
50
|
Lefrançois LH, Pujol C, Bodier CC, Teixeira-Gomez AP, Drobecq H, Rosso ML, Raze D, Dias AA, Hugot JP, Chacon O, Barletta RG, Locht C, Vidal Pessolani MC, Biet F. Characterization of the Mycobacterium avium subsp. paratuberculosis laminin-binding/histone-like protein (Lbp/Hlp) which reacts with sera from patients with Crohn's disease. Microbes Infect 2011; 13:585-94. [PMID: 21334452 DOI: 10.1016/j.micinf.2011.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 01/18/2011] [Accepted: 02/02/2011] [Indexed: 10/18/2022]
Abstract
Mycobacterium avium subsp. paratuberculosis (Map) causes a chronic enteric disease in ruminants, called paratuberculosis or Johne's disease. The current model proposes that after ingestion by the host, Map crosses the intestinal barrier via internalization by the M cells. Experimental observations suggest, however, that Map may also transcytose the intestinal wall via the enterocytes, but the mechanisms involved in this process remain poorly understood. Cytoadherence assays performed on epithelial cells with Map revealed that the addition of laminin to the cell culture increases adhesion. A Map protein was isolated by heparin-Sepharose chromatography and identified as a laminin-binding protein like. The gene encoding this protein named Lbp/Hlp was identified in the Map genome sequence at locus MAP3024 (annotated Hup B). The deduced Map Lbp/Hlp amino acid sequence reveals 80% identity with that reported for other mycobacteria. The C-terminal domain involved in adhesion is mainly composed of arginine and lysine residues modified by methylation. In vitro tests demonstrated that recombinant Lbp/Hlp binds laminin, heparin, collagen and epithelial cells. Interestingly, we found that this adhesin corresponds to the antigen described as the target of pANCA and serum antibodies of patients with Crohn's disease.
Collapse
Affiliation(s)
- Louise H Lefrançois
- INRA, UR1282, Infectiologie Animale, Santé Publique (IASP-311), F-37380 Nouzilly, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|