1
|
Bellapukonda SM, Singothu S, Singampalli A, Bandela R, Kumar P, Yaddanapudi VM, Bhandari V, Nanduri S, Enneiymy M, AlAjm MF, Oubella A. Exploring spirocyclic isoquinoline-piperidine compounds in tuberculosis therapy: ADMET profiling, docking, DFT, MD simulations, and MMGBSA analysis. Comput Biol Chem 2025; 118:108447. [PMID: 40199053 DOI: 10.1016/j.compbiolchem.2025.108447] [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: 02/07/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Tuberculosis remains a global health challenge due to drug-resistant strains. MmpL3 inhibitors have emerged as promising anti-tubercular agents, and their clinical development has been hindered by poor microsomal stability. This study computationally designed and screened 40 spirocyclic analogs, and compared them with ICA38 and SQ109. In silico analyses, including docking, MD simulations, and DFT calculations, were conducted to assess their potential as anti-tubercular agents, highlighting promising candidates for further development. Docking studies using Glide software identified C21 (3,4- dichloro derivative) and C20 (5-chloro derivative) as promising candidates, exhibiting binding scores of -9.79 kcal/mol and -9.64 kcal/mol, respectively. Both compounds interacted with the active site residue Asp645 via hydrogen bonding and also formed a hydrophobic interaction. DFT results revealed that C21 displayed the balanced chemical reactivity, characterized by high dipole moment (3.63D), an optimal energy gap (0.18752 eV), softness and hardness (η = 0.09376 eV, σ = 10.666 eV⁻¹), high electron affinity (0.02305 eV), high electronegativity (0.11681 eV) and high ionization potential (0.21057 cV). On the other hand, C20 exhibited similar electronic properties with marginal differences than C21. MD simulations showed C21 and C20's stability (RMSD 2.4 Å and 2.2 Å, RMSF <2.5 Å), indicating improved Arg344-Leu354 stability. Additionally, C21 and C20 maintained Asp645 interactions (91 %, 97 %) and showed strong binding with free energy values (MMGBSA: -72.23, -66.50 kcal/mol). These findings highlight the efficiency of the compounds C21 and C20 with strong binding affinity, favorable stability, and optimal electronic properties, making them promising candidates for further development of next-generation MmpL3 inhibitors.
Collapse
Affiliation(s)
- Sri Mounika Bellapukonda
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Siva Singothu
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Anuradha Singampalli
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Rani Bandela
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Pardeep Kumar
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| | - Vasundhra Bhandari
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Mohamed Enneiymy
- Université de Haute-Alsace, CNRS, IS2M UMR 7361, Université de Strasbourg, Mulhouse F-68100, France
| | - Mohamed F AlAjm
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ali Oubella
- Laboratory of Organic and Physical Chemistry, Applied Bioorganic Chemistry Team, Faculty of Sciences, Ibnou Zohr University, Agadir, Morocco
| |
Collapse
|
2
|
Kumar GS, Sahoo AK, Ranjan N, Dwivedi VD, Agrawal S. Suppressing Mycobacterium tuberculosis virulence and drug resistance by targeting Eis protein through computational drug discovery. Mol Divers 2025; 29:1697-1723. [PMID: 39096353 DOI: 10.1007/s11030-024-10946-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024]
Abstract
Tuberculosis (TB) remains a critical health threat, particularly with the emergence of multidrug-resistant strains. This demands attention from scientific communities and healthcare professionals worldwide to develop effective treatments. The enhanced intracellular survival (Eis) protein is an acetyltransferase enzyme of Mycobacterium tuberculosis that functions by adding acetyl groups to aminoglycoside antibiotics, which interferes with their ability to bind to the bacterial ribosome, thereby preventing them from inhibiting protein synthesis and killing the bacterium. Therefore, targeting this protein accelerates the chance of restoring the aminoglycoside drug activity, thereby reducing the emergence of drug-resistant TB. For this, we have screened 406,747 natural compounds from the Coconut database against Eis protein. Based on MM/GBSA rescoring binding energy, the top 5 most prominent natural compounds, viz. CNP0187003 (- 96.14 kcal/mol), CNP0176690 (- 93.79 kcal/mol), CNP0136537 (- 92.31 kcal/mol), CNP0398701 (- 91.96 kcal/mol), and CNP0043390 (- 91.60 kcal/mol) were selected. These compounds exhibited the presence of a substantial number of hydrogen bonds and other significant interactions confirming their strong binding affinity with the Eis protein during the docking process. Subsequently, the MD simulation of these compounds exhibited that the Eis-CNP0043390 complex was the most stable, followed by Eis-CNP0187003 and Eis-CNP0176690 complex, further verified by binding free energy calculation, principal component analysis (PCA), and Free energy landscape analysis. These compounds demonstrated the most favourable results in all parameters utilised for this investigation and may have the potential to inhibit the Eis protein. There these findings will leverage computational techniques to identify and develop a natural compound inhibitor as an alternative for drug-resistant TB.
Collapse
Affiliation(s)
- Geethu S Kumar
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Amaresh Kumar Sahoo
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, India
| | - Nishant Ranjan
- University Centre for Research and Development, Department of Mechanical Engineering, Chandigarh University Gharuan, Mohali, Punjab, India
| | - Vivek Dhar Dwivedi
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India.
- Bioinformatics Research Division, Quanta Calculus, Greater Noida, India.
| | - Sharad Agrawal
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India.
| |
Collapse
|
3
|
Peng X, Pu F, Zhou F, Dai X, Xu F, Wang J, Feng J, Xia P. Has-miR-30c-1-3p inhibits macrophage autophagy and promotes Mycobacterium tuberculosis survival by targeting ATG4B and ATG9B. Sci Rep 2025; 15:10240. [PMID: 40133377 PMCID: PMC11937412 DOI: 10.1038/s41598-025-94452-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Autophagy is a widespread physiological process in the body, which also protects the host by degrading invading pathogens and harmful substances during pathological conditions. Nevertheless, Mycobacterium tuberculosis (MTB), the causative agent of tuberculosis, has evolved strategies to subvert autophagy by modulating microRNA (miRNA) expression, enabling its escape from host defenses. In this study, we established an in vitro model using the human macrophage cell line infected with the highly virulent MTB strain H37Rv. Through RNA sequencing and bioinformatic analysis post H37Rv infection, we screened 14 differentially expressed miRNAs. We predicted and demonstrated that miR-30c-1-3p inhibits autophagy and promotes MTB survival by targeting ATG4B and ATG9B during the infection process. The results showed that miR-30c-1-3p expression was gradually increased before 12 h of H37Rv infection, followed by a decrease. Overexpression of miR-30c-1-3p suppressed autophagic activity. We also identified the targeting of miR-30c-1-3p to ATG4B and ATG9B for the first time, and overexpression of both ATG4B and ATG9B, alone or together, on the basis with upregulation of miR-30c-1-3p reversed the inhibition of autophagy. Autophagy levels were analyzed at different levels by western blot, immunofluorescence, and transmission electron microscopy, all of which showed that upregulation of miR-30c-1-3p inhibited autophagy during H37Rv infection. Additionally, the intervention of miR-30c-1-3p mimics resulted in an increased bacterial load in macrophages, suggesting that MTB achieves immune evasion by upregulating miR-30c-1-3p during infection. In conclusion, our study provides a valuable target for the development of host-directed anti-tuberculosis therapy as well as a new diagnostic marker.
Collapse
Affiliation(s)
- Xianglin Peng
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China
| | - Feifei Pu
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Fangzheng Zhou
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Xiyong Dai
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430022, China
| | - Feng Xu
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430022, China
| | - Junwen Wang
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China
| | - Jing Feng
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China.
| | - Ping Xia
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China.
| |
Collapse
|
4
|
O'Grady JF, McHugo GP, Ward JA, Hall TJ, Faherty O'Donnell SL, Correia CN, Browne JA, McDonald M, Gormley E, Riggio V, Prendergast JGD, Clark EL, Pausch H, Meade KG, Gormley IC, Gordon SV, MacHugh DE. Integrative genomics sheds light on the immunogenetics of tuberculosis in cattle. Commun Biol 2025; 8:479. [PMID: 40128580 PMCID: PMC11933339 DOI: 10.1038/s42003-025-07846-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
Mycobacterium bovis causes bovine tuberculosis (bTB), an infectious disease of cattle that represents a zoonotic threat to humans. Research has shown that the peripheral blood (PB) transcriptome is perturbed during bTB disease but the genomic architecture underpinning this transcriptional response remains poorly understood. Here, we analyse PB transcriptomics data from 63 control and 60 confirmed M. bovis-infected animals and detect 2592 differently expressed genes perturbing multiple immune response pathways. Leveraging imputed genome-wide SNP data, we characterise thousands of cis-expression quantitative trait loci (eQTLs) and show that the PB transcriptome is substantially impacted by intrapopulation genomic variation during M. bovis infection. Integrating our cis-eQTL data with bTB susceptibility GWAS summary statistics, we perform a transcriptome-wide association study and identify 115 functionally relevant genes (including RGS10, GBP4, TREML2, and RELT) and provide important new omics data for understanding the host response to mycobacterial infections that cause tuberculosis in mammals.
Collapse
Affiliation(s)
- John F O'Grady
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland
| | - Gillian P McHugo
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland
| | - James A Ward
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland
| | - Thomas J Hall
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland
| | - Sarah L Faherty O'Donnell
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland
- Irish Blood Transfusion Service, National Blood Centre, James's Street, Dublin, Ireland
| | - Carolina N Correia
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland
- Children's Health Ireland, 32 James's Walk, Rialto, Ireland
| | - John A Browne
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland
| | - Michael McDonald
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland
| | - Eamonn Gormley
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Ireland
- UCD One Health Centre, University College Dublin, Belfield, Ireland
| | - Valentina Riggio
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
- Centre for Tropical Livestock Genetics and Health (CTLGH), Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - James G D Prendergast
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
- Centre for Tropical Livestock Genetics and Health (CTLGH), Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Emily L Clark
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
- Centre for Tropical Livestock Genetics and Health (CTLGH), Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Hubert Pausch
- Animal Genomics, ETH Zurich, Universitaetstrasse 2, Zurich, Switzerland
| | - Kieran G Meade
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland
- UCD One Health Centre, University College Dublin, Belfield, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland
| | - Isobel C Gormley
- UCD School of Mathematics and Statistics, University College Dublin, Belfield, Ireland
| | - Stephen V Gordon
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Ireland
- UCD One Health Centre, University College Dublin, Belfield, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland
| | - David E MacHugh
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Ireland.
- UCD One Health Centre, University College Dublin, Belfield, Ireland.
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland.
| |
Collapse
|
5
|
Luan H, Peng C, Yasin P, Shang Q, Xiang W, Song X. Mannosamine-Engineered Nanoparticles for Precision Rifapentine Delivery to Macrophages: Advancing Targeted Therapy Against Mycobacterium Tuberculosis. Drug Des Devel Ther 2025; 19:2081-2102. [PMID: 40129488 PMCID: PMC11931292 DOI: 10.2147/dddt.s505682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
Background Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), remains one of the leading causes of death among infectious diseases. Enhancing the ability of anti-tuberculosis drugs to eradicate Mycobacterium tuberculosis within host cells remains a significant challenge. Methods A mannosamine-modified nanoparticle delivery system was developed using poly(lactic-co-glycolic acid) (PLGA) copolymers to enhance the targeted delivery of rifapentine (RPT) to macrophages. D-mannosamine was conjugated to PLGA-polyethylene glycol (PLGA-PEG) copolymers through EDC/NHS coupling chemistry, and the resultant RPT-MAN-PLGA-PEG nanoparticles (NPs) were prepared through a combination of phacoemulsification and solvent evaporation methods. The physicochemical properties, toxicity, in vitro drug release profiles, stability, cellular uptake, and anti-TB efficacy of the NPs were systematically evaluated. Results The RPT-MAN-PLGA-PEG NPs had a mean particle size of 108.2 ± 7.2 nm, with encapsulation efficiency and drug loading rates of 81.2 ± 6.3% and 13.7 ± 0.7%, respectively. RPT release from the NPs was sustained for over 60 hours. Notably, the phagocytic uptake of the MAN-PLGA NPs by macrophages was significantly higher compared to PLGA-PEG NPs. Both NPs improved pharmacokinetic parameters without inducing significant organ toxicity. The minimum inhibitory concentration for the NPs was 0.047 μg/mL, compared to 0.2 μg/mL for free RPT. Conclusion The engineered RPT-MAN-PLGA-PEG NPs effectively enhanced macrophage uptake in vitro and facilitated the intracellular clearance of Mtb. This nanoparticle-based delivery system offers a promising approach for improving the precision of anti-TB therapy, extending drug release, optimizing pharmacokinetic profiles, augmenting antimicrobial efficacy, and mitigating drug-related toxicities.
Collapse
Affiliation(s)
- Haopeng Luan
- Department of Spine Surgery, the Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830002, People’s Republic of China
| | - Cong Peng
- Department of Spine Surgery, the Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830002, People’s Republic of China
| | - Parhat Yasin
- Department of Spine Surgery, the Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830002, People’s Republic of China
| | - Qisong Shang
- Department of Spine Surgery, the Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830002, People’s Republic of China
| | - Wei Xiang
- Department of Spine Surgery, the Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830002, People’s Republic of China
| | - Xinghua Song
- Department of Spine Surgery, the Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830002, People’s Republic of China
| |
Collapse
|
6
|
Kotlyarov S, Oskin D. The Role of Inflammation in the Pathogenesis of Comorbidity of Chronic Obstructive Pulmonary Disease and Pulmonary Tuberculosis. Int J Mol Sci 2025; 26:2378. [PMID: 40141021 PMCID: PMC11942565 DOI: 10.3390/ijms26062378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/23/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
The comorbid course of chronic obstructive pulmonary disease (COPD) and pulmonary tuberculosis is an important medical and social problem. Both diseases, although having different etiologies, have many overlapping relationships that mutually influence their course and prognosis. The aim of the current review is to discuss the role of different immune mechanisms underlying inflammation in COPD and pulmonary tuberculosis. These mechanisms are known to involve both the innate and adaptive immune system, including various cellular and intercellular interactions. There is growing evidence that immune mechanisms involved in the pathogenesis of both COPD and tuberculosis may jointly contribute to the tuberculosis-associated obstructive pulmonary disease (TOPD) phenotype. Several studies have reported prior tuberculosis as a risk factor for COPD. Therefore, the study of the mechanisms that link COPD and tuberculosis is of considerable clinical interest.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Dmitry Oskin
- Department of Infectious Diseases and Phthisiology, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
7
|
Riaz SM, Hanevik K, Sviland L, Mustafa T. Characterization of Early Lesions of Human Post-Primary Tuberculosis and Its Progression to Necrosis Using Archival Material of the Pre-Antibiotic Era. Pathogens 2025; 14:224. [PMID: 40137709 PMCID: PMC11944378 DOI: 10.3390/pathogens14030224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Primary and post-primary TB are distinct entities. Primary TB occurs when the patient is infected with Mycobacterium tuberculosis (MTB) for the first time without prior immunity, and post-primary TB occurs when the patient has developed immunity against the primary infection. Post-primary TB occurs only in humans. It accounts for 80% of all clinical cases and nearly 100% of transmissions of infection. Early lesions of post-primary TB are reversible, and studying it using modern immunological tools holds the key to developing preventive or treatment strategies. Human lung tissue from untreated TB patients was acquired from pathology archives stored at the Gades Institute of Pathology, Haukeland University Hospital, Bergen, Norway, from 1931 to 1947. Manual immunohistochemistry was performed for macrophage (CD68, CD64 and CD163), T cells (CD3 and CD8), matrix metalloproteinases (MMP-9), and markers for programmed death-pathway PD/PDL-1. Digital quantification was performed using Qupath software. In early lesions of post-primary TB, macrophages showed mixed-phenotype M1 and M2, expressed PDL-1, and were compartmentalized in the alveolar space. T-cells expressed PD-1 and were compartmentalized in the interstitial wall surrounding early lesions. MTB antigens and MMP-9 were also found in early lesions. As the lesion progressed towards necrosis, macrophages showed predominant M1 morphology, and expressions of PDL-1, PD-1, CD8+ cells, and MTB antigens increased. In the early lesions of post-primary TB, the compartmentalization of macrophages in the alveoli and T cells in the interstitium was shown. The PDL-PD1 pathway probably facilitated the mycobacterial growth by evading host immunity.
Collapse
Affiliation(s)
- Syeda Mariam Riaz
- Centre for International Health, Department of Global Public Health and Primary Care, Faculty of Medicine and Dentistry, University of Bergen, 5020 Bergen, Norway;
| | - Kurt Hanevik
- Department of Clinical Science, Faculty of Medicine and Dentistry, University of Bergen, 5020 Bergen, Norway;
- National Center for Tropical Infectious Diseases, Medical Department, Haukeland University Hospital, 5020 Bergen, Norway
| | - Lisbet Sviland
- Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, 5020 Bergen, Norway;
- Department of Pathology, Haukeland University Hospital, 5020 Bergen, Norway
| | - Tehmina Mustafa
- Centre for International Health, Department of Global Public Health and Primary Care, Faculty of Medicine and Dentistry, University of Bergen, 5020 Bergen, Norway;
- Department of Thoracic Medicine, Haukeland University Hospital, 5020 Bergen, Norway
| |
Collapse
|
8
|
Liu D, Abdiriyim A, Zhang L, Ruzitohti B. Functional and mechanistic insights into the stealth protein full-length CpsY is conducive to understanding immune evasion mechanisms by Mycobacterium tuberculosis. Tuberculosis (Edinb) 2025; 152:102616. [PMID: 39985825 DOI: 10.1016/j.tube.2025.102616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/08/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Mycobacterium tuberculosis (Mtb) is a crucial and destructive intracellular pathogen responsible for causing tuberculosis (TB), a disease of substantial morbidity and mortality. Mtb capsular polysaccharides can misdirect the host's immune response pathways, resulting in additional challenges in TB treatment. These capsule polysaccharides are biosynthesized by a series of stealth proteins including CpsY. Our prior investigations elucidated the structural and functional information of the central domain (aa 201-520) of CpsY within Mtb. However, within the host milieu, it is the full-length iteration of CpsY, rather than its truncated form CpsY201-520, that assumes pivotal roles in immune evasion. Consequently, investigating the functional mechanism of full-length CpsY is extremely important. Here, we found that the indispensable role of four conserved regions (CR1-CR4) in governing the phosphotransferase activity of full-length CpsY. Notably, the deletion of S2 (ΔS2) dramatically increased the activity compared to the wild-type (WT) full-length CpsY, thereby revealing S2 in the regulatory dynamics governing the inactivation and activation of full-length CpsY. The gene cpsY helps Mtb to survive in macrophages. Our findings were useful for the development of vaccines and immunotherapies targeting Mtb.
Collapse
Affiliation(s)
- Dafeng Liu
- Xinjiang Key Laboratory of Lavender Conservation and Utilization, College of Biological Sciences and Technology, Yili Normal University, Yining, 835000, Xinjiang, China; School of Life Sciences, Xiamen University, Xiamen, 361102, Fujian, China.
| | - Ablikim Abdiriyim
- Xinjiang Key Laboratory of Lavender Conservation and Utilization, College of Biological Sciences and Technology, Yili Normal University, Yining, 835000, Xinjiang, China
| | - Lvxia Zhang
- Xinjiang Key Laboratory of Lavender Conservation and Utilization, College of Biological Sciences and Technology, Yili Normal University, Yining, 835000, Xinjiang, China
| | - Buayxam Ruzitohti
- Xinjiang Key Laboratory of Lavender Conservation and Utilization, College of Biological Sciences and Technology, Yili Normal University, Yining, 835000, Xinjiang, China
| |
Collapse
|
9
|
Ceccato J, Gualtiero G, Piazza M, Carraro S, Buso H, Felice C, Rattazzi M, Scarpa R, Vianello F, Cinetto F. Shaping Rare Granulomatous Diseases in the Lab: How New Models Are Changing the Game. Cells 2025; 14:293. [PMID: 39996765 PMCID: PMC11853845 DOI: 10.3390/cells14040293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
In vitro models serve as valuable tools for understanding the complex cellular and molecular interactions involved in granuloma formation, providing a controlled environment to explore the underlying mechanisms of their development and function. Various models have been developed to replicate granulomatous diseases, even though they may lack the sophistication needed to fully capture the variability present in clinical spectra and environmental influences. Traditional cultures of PBMCs have been widely used to generate granuloma models, enabling the study of aggregation responses to various stimuli. However, growing cells on a two-dimensional (2D) plastic surface as a monolayer can lead to altered cellular responses and the modulation of signaling pathways, which may not accurately represent in vivo conditions. In response to these limitations, the past decade has seen significant advancements in the development of three-dimensional (3D) in vitro models, which more effectively mimic in vivo conditions and provide better insights into cell-cell and cell-microenvironment interactions. Meanwhile, the use of in vivo animal models in biomedical research must adhere to the principle of the three Rs (replacement, reduction, and refinement) while ensuring that the models faithfully replicate human-specific processes. This review summarizes and compares the main models developed to investigate granulomas, focusing on their contribution to advancing our understanding of granuloma biology. We also discuss the strengths and limitations of each model, offering insights into their biological relevance and practical applications.
Collapse
Affiliation(s)
- Jessica Ceccato
- Hematology and Clinical Immunology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (J.C.); (G.G.)
- Veneto Institute of Molecular Medicine (VIMM), 35128 Padua, Italy
| | - Giulia Gualtiero
- Hematology and Clinical Immunology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (J.C.); (G.G.)
- Veneto Institute of Molecular Medicine (VIMM), 35128 Padua, Italy
| | - Maria Piazza
- Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.P.); (H.B.); (C.F.); (M.R.); (R.S.); (F.C.)
| | - Samuela Carraro
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Helena Buso
- Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.P.); (H.B.); (C.F.); (M.R.); (R.S.); (F.C.)
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Carla Felice
- Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.P.); (H.B.); (C.F.); (M.R.); (R.S.); (F.C.)
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Marcello Rattazzi
- Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.P.); (H.B.); (C.F.); (M.R.); (R.S.); (F.C.)
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Riccardo Scarpa
- Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.P.); (H.B.); (C.F.); (M.R.); (R.S.); (F.C.)
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Fabrizio Vianello
- Hematology and Clinical Immunology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (J.C.); (G.G.)
- Veneto Institute of Molecular Medicine (VIMM), 35128 Padua, Italy
| | - Francesco Cinetto
- Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.P.); (H.B.); (C.F.); (M.R.); (R.S.); (F.C.)
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, 31100 Treviso, Italy;
| |
Collapse
|
10
|
Ma R, Yang W, Guo W, Zhang H, Wang Z, Ge Z. Single-cell transcriptome analysis reveals the dysregulated monocyte state associated with tuberculosis progression. BMC Infect Dis 2025; 25:210. [PMID: 39939918 PMCID: PMC11823163 DOI: 10.1186/s12879-025-10612-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 02/06/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND In tuberculosis (TB) infection, monocytes play a crucial role in regulating the balance between immune tolerance and immune response through various mechanisms. A deeper understanding of the roles of monocyte subsets in TB immune responses may facilitate the development of novel immunotherapeutic strategies and improve TB prevention and treatment. METHODS We retrieved and processed raw single-cell RNA-seq data from SRP247583. Single-cell RNA-seq combined with bioinformatics analysis was employed to investigate the roles of monocytes in TB progression. RESULTS Our findings revealed that classical monocytes expressing inflammatory mediators increased as the disease progressed, whereas non-classical monocytes expressing molecules associated with anti-pathogen infection were progressively depleted. Pseudotime analysis delineated the differentiation trajectory of monocytes from classical to intermediate to non-classical subsets. An abnormal differentiation trajectory to non-classical monocytes may represent a key mechanism underlying TB pathogenesis, with CEBPB and CORO1A identified as genes potentially related to TB development. Analysis of key transcription factors in non-classical monocytes indicated that IRF9 was the only downregulated transcription factor with high AUC activity in this subset. The expression of IRF9 exhibited a decreasing trend in both latent TB infection (LTBI) and active TB groups. Furthermore, dysregulation of transcription factor regulatory networks appeared to impair ferroptosis, with ferroptosis-associated genes MEF2C, MICU1, and PRR5 identified as potential targets of IRF9. Through cell communication analysis, we found that interactions between non-classical monocytes and other subpopulations may mediate TB progression, with MIF and LGALS9 highlighted as potential signaling pathways. CONCLUSION This study employs bioinformatics analysis in conjunction with single-cell sequencing technology to uncover the crucial role of monocyte subsets in tuberculosis infection.
Collapse
Affiliation(s)
- Rong Ma
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wanzhong Yang
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wei Guo
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
| | - Honglai Zhang
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
| | - Zemin Wang
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China
| | - Zhaohui Ge
- The First Clinical Medical School of Ningxia Medical University, Yinchuan, China.
- General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
11
|
Zhu Y, Zhang L, Wang Z, Li T, Chen Y, Lu L, Liu H, Kong D, Peng Y, Chen X, Hu C, Chen H, Guo A. Circular RNA ZNF277 Sponges miR-378d to Inhibit the Intracellular Survival of Mycobacterium tuberculosis by Upregulating Rab10. Cells 2025; 14:262. [PMID: 39996735 PMCID: PMC11853707 DOI: 10.3390/cells14040262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Circular RNAs (circRNAs) are covalently closed non-coding RNAs formed by back-splicing, lacking a 5' cap and poly-A tail. They could act as important regulatory factors in the host's anti-tuberculosis immune process, but only a few have been identified, and their molecular mechanisms remain largely unclear. Here, we identified a novel circRNA, circ-ZNF277, which responds to Mycobacterium tuberculosis (Mtb) infection in THP-1 cells. Circ-ZNF277 binds microRNA-378d (miR-378d) in vivo. The expression level of circ-ZNF277 affects the clearance of the intracellular Mtb in THP-1 cells. Mechanistically, more circ-ZNF277 molecules could absorb more miR-378d, thereby competitively activating the NF-κB signaling pathway, promoting the release of pro-inflammatory cytokines including interleukins IL-1β and IL-6, and tumor necrosis factor-α (TNF-α), and inhibiting the survival of intracellular Mtb. Expressing miR-378d or si-Rab10 targeting the transcription of Rab10 could antagonize the effects of overexpression of circ-ZNF277, resulting in the reduced intracellular survival of Mtb. In summary, circ-ZNF277 inhibits the intracellular survival of Mtb via the miR-378d/Rab10 axis. This finding represents a novel mechanism of circular RNA in regulating host immune responses during Mtb infection.
Collapse
Affiliation(s)
- Yifan Zhu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Lei Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zijian Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Ting Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yingyu Chen
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Lu Lu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Han Liu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Delai Kong
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yongchong Peng
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xi Chen
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Changmin Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Aizhen Guo
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (Y.Z.); (L.Z.); (X.C.)
- National Professional Laboratory for Animal Tuberculosis (Wuhan), Ministry of Agriculture and Rural Affairs, International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
12
|
Singh A, Singh A, Saraswati SSK, Rana AK, Singh A, Verma C, Sinha V, Kalra K, Natarajan K. Suppressive effects of toll-like receptor 2, toll-like receptor 4, and toll-like receptor 7 on protective responses to Mycobacterium bovis BCG from epithelial cells. Microbes Infect 2025; 27:105428. [PMID: 39368609 DOI: 10.1016/j.micinf.2024.105428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Mycobacteria have several mechanisms for evasion of protective responses mounted by the host. In this study, we unravel yet another mechanism that is mediated by Toll-Like Receptors TLR2, TLR4, and TLR7 in epithelial cells. We show that mycobacterial infection of epithelial cells increases the expression of TLR2, TLR4, and TLR7. Stimulation of either TLR along with mycobacterial infection results in an inhibition of oxidative burst resulting in increased survival of mycobacteria inside epithelial cells. TLR stimulation along with mycobacterial infection also inhibits activation of epithelial cells for T cell responses by differentially regulating the activation of ERK-MAPK and p38-MAPK along with inhibition of co-stimulatory molecule CD86 expression. Furthermore, stimulation of either TLR inhibits the induction of apoptosis and autophagy. Knockdown of either TLR by specific siRNAs reverses the inhibition by ROS and apoptosis by mycobacteria and results in reduced intracellular survival of mycobacteria in a MyD88-dependent manner. These results point towards a negative role for TLR2, TLR4, and TLR7 in regulating protective responses to M. bovis BCG infection in epithelial cells.
Collapse
Affiliation(s)
- Aarti Singh
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India.
| | - Akshita Singh
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | | | - Ankush Kumar Rana
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Aayushi Singh
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Chaitenya Verma
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Vishal Sinha
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Kanika Kalra
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Krishnamurthy Natarajan
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
13
|
Jain M, Vyas R. Unveiling the silent defenders: mycobacterial stress sensors at the forefront to combat tuberculosis. Crit Rev Biotechnol 2025:1-19. [PMID: 39880585 DOI: 10.1080/07388551.2024.2449367] [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: 01/24/2024] [Revised: 07/12/2024] [Accepted: 09/14/2024] [Indexed: 01/31/2025]
Abstract
The global escalation in tuberculosis (TB) cases accompanied by the emergence of multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains of Mycobacterium tuberculosis (M.tb) emphasizes the critical requirement for novel potent drugs. The M.tb demonstrates extraordinary adaptability, thriving in diverse conditions, and always finds itself in win-win situations regardless of whether the environment is favorable or unfavorable; no matter the magnitude of the challenge, it can endure and survive. This review aims to uncover the role of multiple stress sensors of M.tb that assist bacteria in remaining viable within the host for years against various physiological stresses offered by the host. M.tb is an exceptionally triumphant pathogen, primarily due to its adeptness in developing defense mechanisms against stressful situations. The recent advances emphasize the significance of M.tb stress sensors, including chaperones, proteases, transcription factors, riboswitches, inteins, etc., employed in responding to a spectrum of physiological stresses imposed by the host, encompassing surface stress, host immune responses, osmotic stress, oxidative and nitrosative stresses, cell envelope stress, environmental stress, reductive stress, and drug pressure. These sensors act as silent defenders orchestrating adaptive strategies, with limited comprehensive information in current literature, necessitating a focused review. The M.tb strategies utilizing these stress sensors to mitigate the impact of traumatic conditions demand attention to neutralize this pathogen effectively. Moreover, the intricacies of these stress sensors provide potential targets to design an effective TB drug using structure-based drug design against this formidable global health threat.
Collapse
Affiliation(s)
- Manya Jain
- Department of Life Sciences, Shiv Nadar Institution of Eminence (Deemed to be University), Gautam Buddha Nagar, Uttar Pradesh, India
| | - Rajan Vyas
- Department of Life Sciences, Shiv Nadar Institution of Eminence (Deemed to be University), Gautam Buddha Nagar, Uttar Pradesh, India
| |
Collapse
|
14
|
Sasaninia K, Mohan AS, Badaoui A, Glassman I, Yoon S, Karapetyan A, Kolloli A, Kumar R, Ramasamy S, Subbian S, Venketaraman V. Glutathione Depletion Exacerbates Hepatic Mycobacterium tuberculosis Infection. BIOLOGY 2025; 14:131. [PMID: 40001899 PMCID: PMC11852144 DOI: 10.3390/biology14020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/12/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025]
Abstract
Extrapulmonary tuberculosis (EPTB) accounts for approximately 17% of all Mycobacterium tuberculosis (M.tb) infections globally. Immunocompromised individuals, such as those with HIV infection or type 2 diabetes mellitus (T2DM), are at an increased risk for EPTB. Previous studies have demonstrated that patients with HIV and T2DM exhibit diminished synthesis of glutathione (GSH) synthesizing enzymes. In a murine model, we showed that the diethyl maleate (DEM)-induced depletion of GSH in the lungs led to increased M.tb burden and an impaired pulmonary granulomatous response to M.tb infection. However, the effects of GSH depletion during active EPTB in the liver and spleen have yet to be elucidated. In this study, we evaluated hepatic GSH and malondialdehyde (MDA) levels, as well as cytokine profiles, in untreated and DEM-treated M.tb-infected wild-type (WT) C57BL/6 mice. Additionally, we assessed hepatic and splenic M.tb burdens and tissue pathologies. DEM treatment resulted in a significant decrease in the levels of the reduced form of GSH and an increase in MDA, oxidized GSH, and interleukin (IL)-6 levels. Furthermore, DEM-induced GSH decrease was associated with decreased production of IL-12 and IL-17 and elevated production of interferon-gamma (IFN-γ), tumor necrosis factor (TNF)-α, and transforming growth factor (TGF)-β. A significant increase in M.tb growth was detected in the liver and spleen in DEM-treated M.tb-infected mice. Large, disorganized lymphocyte infiltrates were detected in the hepatic tissues of DEM-treated mice. Overall, GSH diminishment impaired the granulomatous response to M.tb in the liver and exacerbated M.tb growth in both the liver and spleen. These findings provide critical insights into the immunomodulatory role of GSH in TB pathogenesis and suggest potential therapeutic avenues for the treatment of extrapulmonary M.tb infections.
Collapse
Affiliation(s)
- Kayvan Sasaninia
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.S.); (A.S.M.); (A.B.); (I.G.); (S.Y.); (A.K.)
| | - Aishvaryaa Shree Mohan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.S.); (A.S.M.); (A.B.); (I.G.); (S.Y.); (A.K.)
| | - Ali Badaoui
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.S.); (A.S.M.); (A.B.); (I.G.); (S.Y.); (A.K.)
| | - Ira Glassman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.S.); (A.S.M.); (A.B.); (I.G.); (S.Y.); (A.K.)
| | - Sonyeol Yoon
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.S.); (A.S.M.); (A.B.); (I.G.); (S.Y.); (A.K.)
| | - Arshavir Karapetyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.S.); (A.S.M.); (A.B.); (I.G.); (S.Y.); (A.K.)
| | - Afsal Kolloli
- Public Health Research Institute—New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.K.); (R.K.); (S.R.); (S.S.)
| | - Ranjeet Kumar
- Public Health Research Institute—New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.K.); (R.K.); (S.R.); (S.S.)
| | - Santhamani Ramasamy
- Public Health Research Institute—New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.K.); (R.K.); (S.R.); (S.S.)
| | - Selvakumar Subbian
- Public Health Research Institute—New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.K.); (R.K.); (S.R.); (S.S.)
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.S.); (A.S.M.); (A.B.); (I.G.); (S.Y.); (A.K.)
| |
Collapse
|
15
|
Li R, He T, Yang M, Xu J, Li Y, Wang X, Guo X, Li M, Xu L. Regulation of Bacillus Calmette-Guérin-induced macrophage autophagy and apoptosis by the AMPK-mTOR-ULK1 pathway. Microbiol Res 2025; 290:127952. [PMID: 39476518 DOI: 10.1016/j.micres.2024.127952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/02/2024] [Accepted: 10/24/2024] [Indexed: 12/12/2024]
Abstract
Tuberculosis (TB) is a chronic wasting infectious disease caused by Mycobacterium tuberculosis (MTB) or Mycobacterium bovis that can be transmitted among people and domestic animals. During the development of TB, macrophages of the innate immune system can act against MTB via autophagy and apoptosis to prevent the spread of the disease. Among the many autophagy regulatory pathways, the adenosine monophosphate (AMP)-activated protein kinase (AMPK)-mammalian rapamycin target protein (mTOR)-Unc-51-like kinase 1 (ULK1) pathway has received considerable attention. This study investigates the regulatory role of the AMPK-mTOR-ULK1 pathway in attenuating M. bovis Bacillus Calmette-Guérin (BCG)-induced autophagy and apoptosis in murine monocyte macrophages (RAW264.7). Changes in macrophage autophagy and apoptosis were analyzed using the AMPK activator AICAR and inhibitor Compound C to interfere with the AMPK-mTOR-ULK1 pathway and siRNA to silence the pathway. Consequently, BCG stimulation of macrophages significantly activated the AMPK-mTOR-ULK1 pathway while BCG-induced macrophage AMPK activation promoted macrophage autophagy and apoptosis. Activation of the AMPK-mTOR-ULK1 pathway by AICAR significantly improved autophagy occurrence in BCG-induced macrophages and increased apoptosis while Compound C with siRNA produced opposing effects by attenuating autophagy and apoptosis in BCG-induced macrophages. Thus, the AMPK-mTOR-ULK1 pathway has a dual regulatory role in BCG-induced macrophage autophagy and apoptosis and may have synergistic effects. This study analyzes the mechanism of resistance of host cells to MTB and provides a theoretical basis for new therapeutic strategies and related drug development.
Collapse
Affiliation(s)
- Ruiqian Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Tianle He
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Min Yang
- Guyuan Vocational and Technical School, Guyuan, Ningxia 756000, China
| | - Jinghua Xu
- COFCO Feed (Yinchuan) Co., Ltd., Lingwu, Ningxia 750499, China
| | - Yongqin Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Xueyan Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Xuelian Guo
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Mingzhu Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Lihua Xu
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China.
| |
Collapse
|
16
|
Jia L, Zhang L, Yang H, Li L, Zheng S, Ma Y, Xue Y, Zhang J, Li M, Su X, Wang K. Host-intestinal microbiota interactions in Edwardsiella piscicida-induced lethal enteritis in big-belly seahorses: Novel insights into the role of Carbohydrate-Active enzymes and host transcriptional responses. FISH & SHELLFISH IMMUNOLOGY 2025; 156:110024. [PMID: 39557374 DOI: 10.1016/j.fsi.2024.110024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Edwardsiella piscicida-induced lethal enteritis is a major threat to the sustainable development of seahorse aquaculture. The roles of Carbohydrate-Active enzymes (CAZymes) in interactions between the pathogen and the host are poorly understood. In this study, we found that 22 key CAZymes encoded by E. piscicida might involve in the coordination of five key stages of infection. Specifically, during the motility, adherence, and invasion stages, 10 CAZymes, including CE4, PL8, and CBM48, may significantly increase the activities of Lipid metabolism-associated pathways of the intestinal microbiota (P < 0.01), facilitating pathogen invasion of the host intestinal epithelium. During the replication stage, 11 CAZymes, including GH20, GT4, and GH3, may significantly increase activities of pathways associated with Carbohydrate metabolism (P < 0.01) to promote replication and proliferation of the pathogen. And for avoiding host defenses, GH2 and GH1 may enhance activities of both Carbohydrate and Amino acid metabolic pathways (P < 0.01), facilitating infection and immune evasion. Conjoint analysis showed that E. piscicida might mainly rely on Carbohydrate metabolism for infection, while the host might activate Amino acid metabolic pathways for self-defense. In addition, expressions of 10 key genes, Aldh9a1b, Aoc1, Tpi1b, PCK1, Ldha, Me1, Gla, Cel.2, Ugdh, and Mao, were significantly altered (P < 0.01) and may be used for characterizing host responses to E. piscicida infection. Activities of both Glycolysis/Gluconeogenesis and Tryptophan metabolism were found oppositely changed (P < 0.01) between pathogen and host, respectively, representing the primary focuses of the competition. Overall, this study provides new insights into E. piscicida-mediated intestinal enteritis in fish for the first time from the perspective of CAZymes, as well as a theoretical reference for the prevention and control of these diseases in the aquaculture of seahorses and other fish.
Collapse
Affiliation(s)
- Longwu Jia
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Lele Zhang
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Hongwei Yang
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Lin Li
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Shiyi Zheng
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Yicong Ma
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Yuanyuan Xue
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Jingyi Zhang
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Mingzhu Li
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Xiaolei Su
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China
| | - Kai Wang
- School of Fisheries, Ludong University, Yantai, 264025, China; Research and Development Center of Science, Technology and Industrialization of Seahorses, Ludong University, Yantai, 264025, China.
| |
Collapse
|
17
|
Doghish AS, Abulsoud AI, Nassar YA, Nasr SM, Mohammed OA, Abdel-Reheim MA, Rizk NI, Lutfy RH, Abdel Mageed SS, Ismail MA, Abd-Elhalim HM, Awad FA, Fayez SZ, Elimam H, Mansour RM. Harnessing miRNAs: A Novel Approach to Diagnosis and Treatment of Tuberculosis. J Biochem Mol Toxicol 2025; 39:e70119. [PMID: 39799557 DOI: 10.1002/jbt.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 01/15/2025]
Abstract
Mycobacterium tuberculosis (Mtb) complex, responsible for tuberculosis (TB) infection, continues to be a predominant global cause of mortality due to intricate host-pathogen interactions that affect disease progression. MicroRNAs (miRNAs), essential posttranscriptional regulators, have become pivotal modulators of these relationships. Recent findings indicate that miRNAs actively regulate immunological responses to Mtb complex by modulating autophagy, apoptosis, and immune cell activities. This has resulted in increased interest in miRNAs as prospective diagnostic indicators for TB, especially in differentiating active infection from latent or inactive stages. Variations in miRNA expression during Mtb infection indicate disease progression and offer insights into the immune response. Furthermore, miRNAs present potential as therapeutic targets in host-directed therapy (HDT) techniques for TB infection. This work examines the function of miRNAs in TB pathogenesis, with the objective of identifying particular miRNAs that regulate the immune response to the Mtb complex, evaluating their diagnostic value and exploring their therapeutic implications in host-directed therapy for TB infection. The objective is to enhance comprehension of how miRNAs can facilitate improved diagnosis and treatment of TB.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Yara A Nassar
- Department of Botany, Biotechnology and Its Application Program, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Sami Mohamed Nasr
- Biochemistry and Molecular Biology, Theodor Bilharz Research Institute, Giza, Egypt
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | | | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Radwa H Lutfy
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
| | - Menattallah A Ismail
- Applied Biotechnology Program, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Haytham M Abd-Elhalim
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
- Agricultural Research Center, Agricultural Genetic Engineering Research Institute, Giza, Egypt
| | - Farah A Awad
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Salma Zaki Fayez
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Reda M Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan, Egypt
- Molecular Biology and Biotechnology Department, School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
| |
Collapse
|
18
|
Hu D, Yang Z, Zhang J, Liu G, Pi J, Xu J, Wang Y, Zhao Y. Copper homeostasis; A rapier between mycobacteria and macrophages. FASEB Bioadv 2025; 7:e1484. [PMID: 39781425 PMCID: PMC11705462 DOI: 10.1096/fba.2024-00166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Copper is a vital trace element crucial for mediating interactions between Mycobacterium and macrophages. Within these immune cells, copper modulates oxidative stress responses and signaling pathways, enhancing macrophage immune functions and facilitating Mycobacterium clearance. Conversely, copper may promote Mycobacterium escape from macrophages through various mechanisms: inhibiting macrophage activity, diminishing phagocytic and bactericidal capacities, and supporting Mycobacterium survival and proliferation. This paradox has intensified research focus on the regulatory role of copper in immune cell-pathogen interactions. Interactions among metal ions can affect Mycobacterium concentration, distribution, and activity within an organism. In this review, we have elucidated the role of copper in these interactions, focusing on the mechanisms by which this metal influences both the immune defense mechanisms of macrophages and the survival strategies of Mycobacterium. The findings suggest that manipulating copper levels could enhance macrophage bactericidal functions and potentially limit Mycobacterium resistance. Therefore, elucidating the regulatory role of copper is pivotal for advancing our understanding of metal homeostasis in immune cell-pathogen dynamics and TB pathogenesis. Furthermore, we recommend further investigation into the role of copper in TB pathogenesis to advance tuberculosis diagnosis and treatment and gain comprehensive insights into metal homeostasis in infectious disease contexts.
Collapse
Affiliation(s)
- Di Hu
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
| | - Zisha Yang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
| | - Jun‐ai Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
| | - Ganbin Liu
- Department of RespirationDongguan 6th HospitalDongguanGuangdongChina
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
| | - Junfa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
| | - Yan Wang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Microbiology and Immunology DepartmentGuangdong Medical UniversityDongguanGuangdongChina
| | - Yi Zhao
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
- Microbiology and Immunology DepartmentGuangdong Medical UniversityDongguanGuangdongChina
| |
Collapse
|
19
|
Gao J, Huang X, Zhu Q, He H, Zhang J, Chen J, Wei C, Luo S, Yang S, Xie Z. Mtb/HIV co-infection immune microenvironment subpopulations heterogeneity. Int Immunopharmacol 2024; 143:113341. [PMID: 39405943 DOI: 10.1016/j.intimp.2024.113341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/29/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND The co-infection of human immunodeficiency virus type 1 (HIV-1) and tuberculosis poses a lethal threat. Currently, our understanding of the altered immune responses and diverse immune cell subpopulations triggered by dual pathogen infections remains inadequate. METHODS We utilized single-cell RNA sequencing data from the Gene Expression Omnibus database and the China National GeneBank Nucleotide Sequence Archive to study peripheral blood mononuclear cells from individuals infected with HIV-1 and those co-infected with Mycobacterium tuberculosis (Mtb)/HIV. We investigated cellular components, signaling pathways, biological functions, developmental trajectories, and gene regulatory networks among different cells to determine cellular heterogeneity in the progression of Mtb/HIV co-infection. RESULTS We constructed a single-cell global transcriptional landscape of Mtb/HIV co-infection, revealing heterogeneity among various cell subpopulations. CD4+ T_RACK1_STAT1 subpopulation may participate in the JAK-STAT signaling pathway through RACK1-mediated transcriptional regulation of STAT1, potentially mediating the immune response in patients. Targeting CD8+ T_RACK1_TIGIT subpopulation via RACK1 may help restore the effector capacity of CD8+ T cells. Additionally, Mono_HSP90AA1 and Mono_APOBEC3A subpopulations were positioned at the endpoints of monocyte differentiation trajectories in different patients, suggesting their significant roles in distinct types of immune responses. CTL_GNLY and NK_HSPA1A subpopulations were specifically enriched in three distinct HIV-infected patient groups, indicating their crucial roles in the immune cytotoxicity associated with Mtb/HIV co-infection. CONCLUSION The immune system disruptions caused by HIV-1 infection are further exacerbated by co-infection with Mtb. This compounded effect leads to significant heterogeneity in immune cell subpopulations among co-infected individuals, promoting immune system dysfunction.
Collapse
Affiliation(s)
- Jiamin Gao
- Laboratory of Infectious Disease, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning) and The Fourth People's Hospital of Nanning, Nanning 530023, China; Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning 530023, China.
| | - Xianzhen Huang
- Laboratory of Infectious Disease, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning) and The Fourth People's Hospital of Nanning, Nanning 530023, China; Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning 530023, China
| | - Qingdong Zhu
- Laboratory of Infectious Disease, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning) and The Fourth People's Hospital of Nanning, Nanning 530023, China; Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning 530023, China
| | - Huawei He
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning 530023, China
| | - Jie Zhang
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning 530023, China
| | - Jieling Chen
- Laboratory of Infectious Disease, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning) and The Fourth People's Hospital of Nanning, Nanning 530023, China
| | - Cailing Wei
- Laboratory of Infectious Disease, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning) and The Fourth People's Hospital of Nanning, Nanning 530023, China
| | - Shunda Luo
- Department of Clinical Laboratory, The Fourth People's Hospital of Nanning, Nanning 530023, China
| | - Shixiong Yang
- Laboratory of Infectious Disease, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning) and The Fourth People's Hospital of Nanning, Nanning 530023, China; Administrative Office, The Fourth People's Hospital of Nanning, Nanning 530023, China.
| | - Zhouhua Xie
- Laboratory of Infectious Disease, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning) and The Fourth People's Hospital of Nanning, Nanning 530023, China; Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning 530023, China; Administrative Office, The Fourth People's Hospital of Nanning, Nanning 530023, China.
| |
Collapse
|
20
|
Nasiri MJ, Lutfy K, Venketaraman V. Challenges of Multidrug-Resistant Tuberculosis Meningitis: Current Treatments and the Role of Glutathione as an Adjunct Therapy. Vaccines (Basel) 2024; 12:1397. [PMID: 39772057 PMCID: PMC11728556 DOI: 10.3390/vaccines12121397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/05/2025] Open
Abstract
Multidrug-resistant tuberculosis (MDR-TB) poses a significant global health threat, especially when it involves the central nervous system (CNS). Tuberculous meningitis (TBM), a severe manifestation of TB, is linked to high mortality rates and long-term neurological complications, further exacerbated by drug resistance and immune evasion mechanisms employed by Mycobacterium tuberculosis (Mtb). Although pulmonary TB remains the primary focus of research, MDR-TBM introduces unique challenges in diagnosis, treatment, and patient outcomes. The effectiveness of current treatments is frequently compromised by poor CNS penetration of anti-TB drugs and the necessity for prolonged therapy, which often involves considerable toxicity. This review explores the potential of cytokine-based adjunct immunotherapies for MDR-TBM, addressing the challenges of balancing pro-inflammatory and anti-inflammatory signals within the CNS. A central focus is the prospective role of glutathione, not only in reducing oxidative stress but also in enhancing host immune defenses against Mtb's immune evasion strategies. Furthermore, the development of vaccines aimed at upregulating glutathione synthesis in macrophages represents a promising strategy to bolster the immune response and improve treatment outcomes. By integrating glutathione and innovative vaccine approaches into MDR-TBM management, this review proposes a comprehensive strategy that targets Mtb directly while supporting immune modulation, with the potential to enhance patient outcomes and reduce treatment related adverse effects. We underscore the urgent need for further research into adjunctive therapies and immunomodulatory strategies to more effectively combat MDR-TBM.
Collapse
Affiliation(s)
- Mohammad J. Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-69411, Iran;
| | - Kabir Lutfy
- College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766-1854, USA;
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| |
Collapse
|
21
|
Khlebnikova A, Kirshina A, Zakharova N, Ivanov R, Reshetnikov V. Current Progress in the Development of mRNA Vaccines Against Bacterial Infections. Int J Mol Sci 2024; 25:13139. [PMID: 39684849 DOI: 10.3390/ijms252313139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Bacterial infections have accompanied humanity for centuries. The discovery of the first antibiotics and the subsequent golden era of their discovery temporarily shifted the balance in this confrontation to the side of humans. Nevertheless, the excessive and improper use of antibacterial drugs and the evolution of bacteria has gotten the better of humans again. Therefore, today, the search for new antibacterial drugs or the development of alternative approaches to the prevention and treatment of bacterial infections is relevant and topical again. Vaccination is one of the most effective strategies for the prevention of bacterial infections. The success of new-generation vaccines, such as mRNA vaccines, in the fight against viral infections has prompted many researchers to design mRNA vaccines against bacterial infections. Nevertheless, the biology of bacteria and their interactions with the host's immunity are much more complex compared to viruses. In this review, we discuss structural features and key mechanisms of evasion of an immune response for nine species of bacterial pathogens against which mRNA vaccines have been developed and tested in animals. We focus on the results of experiments involving the application of mRNA vaccines against various bacterial pathogens in animal models and discuss possible options for improving the vaccines' effectiveness. This is one of the first comprehensive reviews of the use of mRNA vaccines against bacterial infections in vivo to improve our knowledge.
Collapse
Affiliation(s)
- Alina Khlebnikova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Anna Kirshina
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Natalia Zakharova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Roman Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Vasiliy Reshetnikov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| |
Collapse
|
22
|
Shleeva MO, Demina GR, Savitsky AP. A systematic overview of strategies for photosensitizer and light delivery in antibacterial photodynamic therapy for lung infections. Adv Drug Deliv Rev 2024; 215:115472. [PMID: 39549920 DOI: 10.1016/j.addr.2024.115472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/25/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
Antimicrobial photodynamic therapy (aPDT) emerges as a viable treatment strategy for infections resistant to conventional antibiotics. A complex interplay of factors, including intracellular photosensitizer (PS) accumulation, photochemical reaction type, and oxygen levels, determines the efficacy of aPDT. Recent progress includes the development of modified PSs with enhanced lipophilicity and target-specific strategies to improve bacterial cell wall penetration and targeting. Nanotechnology-based approaches, such as using nanomaterials for targeted PS delivery, have shown promise in enhancing aPDT efficacy. Advancements in light delivery methods for aPDT, such as transillumination of large lesions and local light delivery using fiber optic techniques, are also being explored to optimize treatment efficacy in clinical settings. The limited number of animal models and clinical trials specifically designed to assess the efficacy of aPDT for lung infections highlights the need for further research in this critical area. The potential prospects of aPDT for lung tissue infections originating from antibiotic-resistant bacterial infections are also discussed in this review.
Collapse
Affiliation(s)
- Margarita O Shleeva
- A.N. Bach Institute of Biochemistry, Federal Research Centre 'Fundamentals of Biotechnology' of the Russian Academy of Sciences, Moscow, Russia.
| | - Galina R Demina
- A.N. Bach Institute of Biochemistry, Federal Research Centre 'Fundamentals of Biotechnology' of the Russian Academy of Sciences, Moscow, Russia
| | - Alexander P Savitsky
- A.N. Bach Institute of Biochemistry, Federal Research Centre 'Fundamentals of Biotechnology' of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
23
|
Liu Y, Zhang J, Zhao H, Zhong F, Li J, Zhao L. VBNC Cronobacter sakazakii survives in macrophages by resisting oxidative stress and evading recognition by macrophages. BMC Microbiol 2024; 24:458. [PMID: 39506633 PMCID: PMC11539806 DOI: 10.1186/s12866-024-03595-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Survival in host macrophages is an effective strategy for pathogenic bacterial transmission and pathogenesis. Our previous study found that viable but non-culturable (VBNC) Cronobacter Sakazakii (C. sakazakii) can survive in macrophages, but its survival mechanism is not clear. In this study, we investigated the possible mechanisms of VBNC C. sakazakii survival in macrophages in terms of environmental tolerance within macrophages and evasion of macrophages recognition. The results revealed that VBNC C. sakazakii survived under oxidative conditions at a higher rate than the culturable C. sakazakii. Moreover, the stringent response gene (relA and spoT) and the antioxidant-related genes (sodA, katG, and trxA) were up-regulated, indicating that VBNC C. sakazakii may regulate antioxidation through stringent response. On the other hand, compared with culturable C. sakazakii, VBNC C. sakazakii caused reduced response (Toll-like receptor 4) in macrophages, which was attributed to the suppression of biosynthesis of the lipopolysaccharides (LPS). Furthermore, we found that ellagic acid can reduce the survival rate of bacteria in macrophages by improving the immune TLR4 recognition ability of macrophages. In conclusion, VBNC C. sakazakii may survive in macrophages by regulating oxidative tolerance through stringent response and altering LPS synthesis to evade TLR4 recognition by macrophages, which suggests the pathogenic risk of VBNC C. sakazakii.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Jingfeng Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Haoqing Zhao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Feifeng Zhong
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Jianyu Li
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Lichao Zhao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China.
| |
Collapse
|
24
|
Megawati D, Armitige LY, Tazi L. Differential Host Gene Expression in Response to Infection by Different Mycobacterium tuberculosis Strains-A Pilot Study. Microorganisms 2024; 12:2146. [PMID: 39597535 PMCID: PMC11596623 DOI: 10.3390/microorganisms12112146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Tuberculosis (TB) represents a global public health threat and is a leading cause of morbidity and mortality worldwide. Effective control of TB is complicated with the emergence of multidrug resistance. Yet, there is a fundamental gap in understanding the complex and dynamic interactions between different Mycobacterium tuberculosis strains and the host. In this pilot study, we investigated the host immune response to different M. tuberculosis strains, including drug-sensitive avirulent or virulent, and rifampin-resistant or isoniazid-resistant virulent strains in human THP-1 cells. We identified major differences in the gene expression profiles in response to infection with these strains. The expression of IDO1 and IL-1β in the infected cells was stronger in all virulent M. tuberculosis strains. The most striking result was the overexpression of many interferon-stimulated genes (ISGs) in cells infected with the isoniazid-resistant strain, compared to the rifampin-resistant and the drug-sensitive strains. Our data indicate that infection with the isoniazid-resistant M. tuberculosis strain preferentially resulted in cGAS-STING/STAT1 activation, which induced a characteristic host immune response. These findings reveal complex gene signatures and a dynamic variation in the immune response to infection by different M. tuberculosis strains.
Collapse
Affiliation(s)
- Dewi Megawati
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616, USA;
- Department of Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Warmadewa University, Denpasar 80239, Bali, Indonesia
| | | | - Loubna Tazi
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616, USA;
| |
Collapse
|
25
|
Georgakopoulou VE, Papalexis P, Trakas N. Nanotechnology-based approaches for targeted drug delivery for the treatment of respiratory tract infections. J Biol Methods 2024; 11:e99010032. [PMID: 39839091 PMCID: PMC11744063 DOI: 10.14440/jbm.2024.0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 01/23/2025] Open
Abstract
Background Nanotechnology has emerged as a promising field for the diagnosis, monitoring, and treatment of respiratory tract infections (RTIs). By leveraging the unique properties of nanoscale delivery systems, nanotechnology can significantly enhance the selectivity and efficacy of antimicrobials, thereby reducing off-target effects. Objective This review explores the development and application of targeted nanosystems in combating viral, bacterial, and fungal RTIs. Nanotechnology-based systems, including biological and non-biological nanoparticles, offer innovative solutions for overcoming antimicrobial resistance, improving drug bioavailability, and minimizing systemic side effects. RTIs are a leading cause of morbidity and mortality globally, particularly affecting vulnerable populations such as children, the elderly, and immunocompromised individuals. Traditional drug delivery methods face numerous challenges, such as rapid clearance, poor tissue penetration, and drug degradation. Nanoparticle-based delivery systems address these issues by enhancing tissue penetration, providing sustained drug release, and enabling targeted delivery to infection sites. These systems include liposomal delivery, polymeric nanoparticles, dendrimers, and metal-based nanoparticles, each offering unique advantages in treating RTIs. Nanotechnology also plays a crucial role in vaccine development by offering new strategies to enhance immune responses and improve antigen delivery. Furthermore, the review discusses the clinical translation and regulatory considerations for nanotechnology-based drug delivery, emphasizing the need for rigorous testing and quality control to ensure safety and efficacy. Conclusion Nanotechnology offers promising advancements in the treatment, and prevention of RTIs by enhancing drug delivery and efficacy. By addressing challenges such as antimicrobial resistance and poor tissue penetration, nanotechnology-based systems have the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
| | - Petros Papalexis
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, Athens 12243, Greece
| | - Nikolaos Trakas
- Department of Biochemistry, Sismanogleio Hospital, Athens 15126, Greece
| |
Collapse
|
26
|
Qin Y, Zhou G, Jiao F, Cheng C, Meng C, Wang L, Wu S, Fan C, Li J, Zhou B, Chu Y, Jiao H. Brucella mediates autophagy, inflammation, and apoptosis to escape host killing. Front Cell Infect Microbiol 2024; 14:1408407. [PMID: 39507949 PMCID: PMC11537862 DOI: 10.3389/fcimb.2024.1408407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/26/2024] [Indexed: 11/08/2024] Open
Abstract
Brucellosis is a serious zoonosis caused by Brucella spp. infection, which not only seriously jeopardizes the health of humans and mammals, but also causes huge economic losses to the livestock industry. Brucella is a Gram-negative intracellular bacterium that relies primarily on its virulence factors and a variety of evolved survival strategies to replicate and proliferate within cells. Currently, the mechanisms of autophagy, inflammation, and apoptosis in Brucella-infected hosts are not fully understood and require further research and discussion. This review focuses on the relationship between Brucella and autophagy, inflammation, and apoptosis to provide the scientific basis for revealing the pathogenesis of Brucella.
Collapse
Affiliation(s)
- Yaqiong Qin
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Gengxu Zhou
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Fengyuan Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chuan Cheng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chi Meng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Lingjie Wang
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Shengping Wu
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Cailiang Fan
- The College of Veterinary Medicine, Southwest University, Chongqing, China
- Animal Epidemic Prevention and Control Center of Rongchang, Chongqing, China
| | - Jixiang Li
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Bo Zhou
- Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Yuefeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Hanwei Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
27
|
Li F, Zhang X, Xu J, Zhang Y, Li G, Yang X, Deng G, Dai Y, Liu B, Kosan C, Chen X, Cai Y. SIRT7 remodels the cytoskeleton via RAC1 to enhance host resistance to Mycobacterium tuberculosis. mBio 2024; 15:e0075624. [PMID: 39287444 PMCID: PMC11481912 DOI: 10.1128/mbio.00756-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Phagocytosis of Mycobacterium tuberculosis (Mtb) followed by its integration into the matured lysosome is critical in the host defense against tuberculosis. How Mtb escapes this immune attack remains elusive. In this study, we unveiled a novel regulatory mechanism by which SIRT7 regulates cytoskeletal remodeling by modulating RAC1 activation. We discovered that SIRT7 expression was significantly reduced in CD14+ monocytes of TB patients. Mtb infection diminished SIRT7 expression by macrophages at both the mRNA and protein levels. SIRT7 deficiency impaired actin cytoskeleton-dependent macrophage phagocytosis, LC3II expression, and bactericidal activity. In a murine tuberculosis model, SIRT7 deficiency detrimentally impacted host resistance to Mtb, while Sirt7 overexpression significantly increased the host defense against Mtb, as determined by bacterial burden and inflammatory-histopathological damage in the lung. Mechanistically, we demonstrated that SIRT7 limits Mtb infection by directly interacting with and activating RAC1, through which cytoskeletal remodeling is modulated. Therefore, we concluded that SIRT7, in its role regulating cytoskeletal remodeling through RAC1, is critical for host responses during Mtb infection and proposes a potential target for tuberculosis treatment.IMPORTANCETuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a significant global health issue. Critical to macrophages' defense against Mtb is phagocytosis, governed by the actin cytoskeleton. Previous research has revealed that Mtb manipulates and disrupts the host's actin network, though the specific mechanisms have been elusive. Our study identifies a pivotal role for SIRT7 in this context: Mtb infection leads to reduced SIRT7 expression, which, in turn, diminishes RAC1 activation and consequently impairs actin-dependent phagocytosis. The significance of our research is that SIRT7 directly engages with and activates Rac Family Small GTPase 1 (RAC1), thus promoting effective phagocytosis and the elimination of Mtb. This insight into the dynamic between host and pathogen in TB not only broadens our understanding but also opens new avenues for therapeutic development.
Collapse
Affiliation(s)
- Fuxiang Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
| | - Ximeng Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Jinjin Xu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yue Zhang
- School of Pharmaceutical Sciences, Shenzhen University Medical School, Shenzhen, China
| | - Guo Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Xirui Yang
- Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Guofang Deng
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Youchao Dai
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, China
| | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yi Cai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
28
|
Liu Y, Wang J, Yang J, Xia J, Yu J, Chen D, Huang Y, Yang F, Ruan Y, Xu JF, Pi J. Nanomaterial-mediated host directed therapy of tuberculosis by manipulating macrophage autophagy. J Nanobiotechnology 2024; 22:608. [PMID: 39379986 PMCID: PMC11462893 DOI: 10.1186/s12951-024-02875-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
Tuberculosis (TB), induced by Mycobacterium tuberculosis (Mtb) infection, remains a major public health issue worldwide. Mtb has developed complicated strategies to inhibit the immunological clearance of host cells, which significantly promote TB epidemic and weaken the anti-TB treatments. Host-directed therapy (HDT) is a novel approach in the field of anti-infection for overcoming antimicrobial resistance by enhancing the antimicrobial activities of phagocytes through phagosomal maturation, autophagy and antimicrobial peptides. Autophagy, a highly conserved cellular event within eukaryotic cells that is effective against a variety of bacterial infections, has been shown to play a protective role in host defense against Mtb. In recent decades, the introduction of nanomaterials into medical fields open up a new scene for novel therapeutics with enhanced efficiency and safety against different diseases. The active modification of nanomaterials not only allows their attractive targeting effects against the host cells, but also introduce the potential to regulate the host anti-TB immunological mechanisms, such as apoptosis, autophagy or macrophage polarization. In this review, we introduced the mechanisms of host cell autophagy for intracellular Mtb clearance, and how functional nanomaterials regulate autophagy for disease treatment. Moreover, we summarized the recent advances of nanomaterials for autophagy regulations as novel HDT strategies for anti-TB treatment, which may benefit the development of more effective anti-TB treatments.
Collapse
Affiliation(s)
- Yilin Liu
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China
| | - Jiajun Wang
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China
| | - Jiayi Yang
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China
| | - Jiaojiao Xia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Jiaqi Yu
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China
| | - Dongsheng Chen
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China
| | - Yuhe Huang
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China
| | - Fen Yang
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China
| | - Yongdui Ruan
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China.
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China.
| | - Jun-Fa Xu
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China.
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China.
| | - Jiang Pi
- Research Center of Nano Technology and Application Engineering, School of Medical Technology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, China.
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
29
|
Shetty A, Kwas H, Rajhi H, Rangareddy H, Fryer J. Revolutionizing Tuberculosis Management With Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas Technology: A Comprehensive Literature Review. Cureus 2024; 16:e71697. [PMID: 39552996 PMCID: PMC11568648 DOI: 10.7759/cureus.71697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 11/19/2024] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas systems have gained attention for their revolutionary potential in tuberculosis (TB) management, providing a novel approach to both diagnostics and treatment. This technology, renowned for its ability to accurately target and modify genetic material, offers a promising solution to the limitations of current TB diagnostic methods, which often rely on time-consuming culture techniques or polymerase chain reaction (PCR)-based assays. One of the key advantages of CRISPR-Cas systems is their high specificity and sensitivity, making them well-suited for detecting Mycobacterium tuberculosis, even in low-bacterial-load samples. Techniques such as CRISPR-Cas12 and Cas13 have been employed for rapid detection, utilizing their trans-cleavage activity to produce a fluorescent signal upon recognition of the TB genome. Furthermore, these methods often use isothermal amplification techniques like recombinase polymerase amplification (RPA) or loop-mediated isothermal amplification (LAMP), which require less equipment compared to traditional PCR. Beyond diagnostics, CRISPR-Cas technologies show promise in studying TB resistance mechanisms and potentially treating drug-resistant strains. Genome-editing capabilities enable researchers to manipulate the M. tuberculosis genome, investigating genes linked to virulence or antibiotic resistance. Although challenges such as the development of multiplexed CRISPR assays for detecting multiple mutations simultaneously remain, advancements continue to improve the technology's practicality for clinical use. Incorporating CRISPR into TB management could enhance early detection, inform personalized treatment, and potentially contribute to developing more effective therapies, especially in regions where TB remains a significant public health threat.
Collapse
Affiliation(s)
- Achal Shetty
- Community Medicine, Father Muller Medical College, Mangalore, IND
| | - Hamida Kwas
- Pulmonology, University of Sfax, Faculty of Medicine of Sfax, Gabès University Hospital, Gabès, TUN
| | - Hayfa Rajhi
- Analysis Laboratory Research, University Hospital of Gabès, Gabès, TUN
| | | | | |
Collapse
|
30
|
Fan S, Zhao D, Wang J, Ma Y, Chen D, Huang Y, Zhang T, Liu Y, Xia J, Huang X, Lu Y, Ruan Y, Xu JF, Shen L, Yang F, Pi J. Photothermal and host immune activated therapy of cutaneous tuberculosis using macrophage targeted mesoporous polydopamine nanoparticles. Mater Today Bio 2024; 28:101232. [PMID: 39315396 PMCID: PMC11418140 DOI: 10.1016/j.mtbio.2024.101232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/25/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
Tuberculosis (TB) remains the leading cause of deaths among infectious diseases worldwide. Cutaneous Tuberculosis (CTB), caused by Mycobacterium tuberculosis (Mtb) infection in the skin, is still a harmful public health issue that requires more effective treatment strategy. Herein, we introduced mannose-modified mesoporous polydopamine nanosystems (Man-mPDA NPs) as the macrophage-targeted vectors to deliver anti-TB drug rifampicin and as photothermal agent to facilitate photothermal therapy (PTT) against Mtb infected macrophages for synergistic treatment of CTB. Based on the selective macrophage targeting effects, the proposed Rif@Man-mPDA NPs also showed excellent photothermal properties to develop Rif@Man-mPDA NPs-mediated PTT for intracellular Mtb killings in macrophages. Importantly, Rif@Man-mPDA NPs could inhibit the immune escape of Mtb by effectively chelating intracellular Fe2+ and inhibiting lipid peroxidation, and up-regulating GPX4 expression to inhibit ferroptosis of Mtb infected macrophages through activating Nrf2/HO-1 signaling. Moreover, Rif@Man-mPDA NPs-mediated PTT could effectively activate host cell immune responses by promoting autophagy of Mtb infected macrophages, which thus synergizes targeted drug delivery and ferroptosis inhibition for more effective intracellular Mtb clearance. This Rif@Man-mPDA NPs-mediated PTT strategy could also effectively inhibit the Mtb burdens and alleviate the pathological lesions induced by Mtb infection without significant systemic side effects in mouse CTB model. These results indicate that Rif@Man-mPDA NPs-mediated PTT can be served as a novel anti-TB strategy against CTB by synergizing macrophage targeted photothermal therapy and host immune defenses, thus holding promise for more effective treatment strategy development against CTB.
Collapse
Affiliation(s)
- Shuhao Fan
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Daina Zhao
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiajun Wang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yuhe Ma
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Dongsheng Chen
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yuhe Huang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Tangxin Zhang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yilin Liu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiaojiao Xia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Xueqin Huang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yujia Lu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yongdui Ruan
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
| | - Jun-Fa Xu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Ling Shen
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| | - Fen Yang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
31
|
Malik AA, Shariq M, Sheikh JA, Fayaz H, Srivastava G, Thakuri D, Ahuja Y, Ali S, Alam A, Ehtesham NZ, Hasnain SE. Regulation of Type I Interferon and Autophagy in Immunity against Mycobacterium Tuberculosis: Role of CGAS and STING1. Adv Biol (Weinh) 2024; 8:e2400174. [PMID: 38977406 DOI: 10.1002/adbi.202400174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/22/2024] [Indexed: 07/10/2024]
Abstract
Mycobacterium tuberculosis (M. tb) is a significant intracellular pathogen responsible for numerous infectious disease-related deaths worldwide. It uses ESX-1 T7SS to damage phagosomes and to enter the cytosol of host cells after phagocytosis. During infection, M. tb and host mitochondria release dsDNA, which activates the CGAS-STING1 pathway. This pathway leads to the production of type I interferons and proinflammatory cytokines and activates autophagy, which targets and degrades bacteria within autophagosomes. However, the role of type I IFNs in immunity against M. tb is controversial. While previous research has suggested a protective role, recent findings from cgas-sting1 knockout mouse studies have contradicted this. Additionally, a study using knockout mice and non-human primate models uncovered a new mechanism by which neutrophils recruited to lung infections form neutrophil extracellular traps. Activating plasmacytoid dendritic cells causes them to produce type I IFNs, which interfere with the function of interstitial macrophages and increase the likelihood of tuberculosis. Notably, M. tb uses its virulence proteins to disrupt the CGAS-STING1 signaling pathway leading to enhanced pathogenesis. Investigating the CGAS-STING1 pathway can help develop new ways to fight tuberculosis.
Collapse
Affiliation(s)
- Asrar Ahmad Malik
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Mohd Shariq
- ICMR-National Institute of Pathology, Ansari Nagar West, New Delhi, 110029, India
| | - Javaid Ahmad Sheikh
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Haleema Fayaz
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Gauri Srivastava
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Deeksha Thakuri
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Yashika Ahuja
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Saquib Ali
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Anwar Alam
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Nasreen Z Ehtesham
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
| | - Seyed E Hasnain
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201306, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), Hauz Khas, New Delhi, 110 016, India
| |
Collapse
|
32
|
Zhang D, Zhao H, Li P, Wu X, Liang Y. Research Progress on Liposome Pulmonary Delivery of Mycobacterium tuberculosis Nucleic Acid Vaccine and Its Mechanism of Action. J Aerosol Med Pulm Drug Deliv 2024; 37:284-298. [PMID: 38669118 PMCID: PMC11502632 DOI: 10.1089/jamp.2023.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 03/07/2024] [Indexed: 04/28/2024] Open
Abstract
Traditional vaccines have played an important role in the prevention and treatment of infectious diseases, but they still have problems such as low immunogenicity, poor stability, and difficulty in inducing lasting immune responses. In recent years, the nucleic acid vaccine has emerged as a relatively cheap and safe new vaccine. Compared with traditional vaccines, nucleic acid vaccine has some unique advantages, such as easy production and storage, scalability, and consistency between batches. However, the direct administration of naked nucleic acid vaccine is not ideal, and safer and more effective vaccine delivery systems are needed. With the rapid development of nanocarrier technology, the combination of gene therapy and nanodelivery systems has broadened the therapeutic application of molecular biology and the medical application of biological nanomaterials. Nanoparticles can be used as potential drug-delivery vehicles for the treatment of hereditary and infectious diseases. In addition, due to the advantages of lung immunity, such as rapid onset of action, good efficacy, and reduced adverse reactions, pulmonary delivery of nucleic acid vaccine has become a hot spot in the field of research. In recent years, lipid nanocarriers have become safe, efficient, and ideal materials for vaccine delivery due to their unique physical and chemical properties, which can effectively reduce the toxic side effects of drugs and achieve the effect of slow release and controlled release, and there have been a large number of studies using lipid nanocarriers to efficiently deliver target components into the body. Based on the delivery of tuberculosis (TB) nucleic acid vaccine by lipid carrier, this article systematically reviews the advantages and mechanism of liposomes as a nucleic acid vaccine delivery carrier, so as to lay a solid foundation for the faster and more effective development of new anti-TB vaccine delivery systems in the future.
Collapse
Affiliation(s)
- Danyang Zhang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
- Postgraduate Department of Heibei North University, Zhangjiakou, China
| | - Haimei Zhao
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
- Postgraduate Department of Heibei North University, Zhangjiakou, China
| | - Ping Li
- Postgraduate Department of Heibei North University, Zhangjiakou, China
| | - Xueqiong Wu
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Yan Liang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
33
|
Wang C, Jiang Y, Yang Z, Xu H, Khalid AK, Iftakhar T, Peng Y, Lu L, Zhang L, Bermudez L, Guo A, Chen Y. Host factor RBMX2 promotes epithelial cell apoptosis by downregulating APAF-1's Retention Intron after Mycobacterium bovis infection. Front Immunol 2024; 15:1431207. [PMID: 39308873 PMCID: PMC11412827 DOI: 10.3389/fimmu.2024.1431207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
The Mycobacterium tuberculosis variant bovis (M. bovis) is a highly pathogenic environmental microorganism that causes bovine tuberculosis (bTB), a significant zoonotic disease. Currently, "test and culling" is the primary measure for controlling bTB, but it has been proven to be inadequate in animals due to their high susceptibility to the pathogen. Selective breeding for increased host resistance to bTB to reduce its prevalence is feasible. In this study, we found a vital host-dependent factor, RBMX2, that can potentially promote M. bovis infection. By knocking RBMX2 out, we investigated its function during M. bovis infection. Through transcriptome sequencing and alternative splicing transcriptome sequencing, we concluded that after M. bovis infection, embryo bovine lung (EBL) cells were significantly enriched in RNA splicing associated with apoptosis compared with wild-type EBL cells. Through protein/molecular docking, molecular dynamics simulations, and real-time quantitative PCR, we demonstrated that RBMX2 promotes the apoptosis of epithelial cells by upregulating and binding to apoptotic peptidase activating factor 1 (APAF-1), resulting in the alternative splicing of APAF-1 as a retention intron. To our knowledge, this is the first report of M. bovis affecting host epithelial cell apoptosis by hijacking RBMX2 to promote the intron splicing of downstream APAF-1. These findings may represent a significant contribution to the development of novel TB prevention and control strategies.
Collapse
Affiliation(s)
- Chao Wang
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| | - Yanzhu Jiang
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| | - Zhiming Yang
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haojun Xu
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Abdul Karim Khalid
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tahira Iftakhar
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yongchong Peng
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| | - Lu Lu
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| | - Lei Zhang
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| | - Luiz Bermudez
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States
| | - Aizhen Guo
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Yingyu Chen
- The National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
34
|
Peng X, Pu F, Zhou F, Dai X, Xu F, Wang J, Feng J, Xia P. Exploring expression levels of the cGAS-STING pathway genes in peripheral blood mononuclear cells of spinal tuberculosis patients. BMC Infect Dis 2024; 24:915. [PMID: 39232642 PMCID: PMC11373091 DOI: 10.1186/s12879-024-09815-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND This study aimed to investigate the differential expression levels of the cGAS-STING pathway in peripheral blood mononuclear cells (PBMCs) of spinal tuberculosis (TB) patients with different progression and its feasibility as a diagnostic marker. METHODS Peripheral blood and medical records of 25 patients with spinal TB and 10 healthy individuals, were prospectively collected and analyzed. PBMCs and serum were extracted from peripheral blood and the expression levels of the cGAS-STING pathway in PBMCs were measured by real-time PCR (RT-PCR) and serum interferon β (IFN-β) expression levels were measured by enzyme-linked immunosorbent assay (ELISA). The expression of Interferon regulatory Factor 3 (IRF3) in PBMCs was measured using western blot. Statistical analysis was performed using the SPSS 26.0 statistical package. RESULTS The results showed that the expression level of the TANK-binding kinase 1 (TBK1) and IRF3 was significantly higher in PBMCs (P < 0.05), in patients with active lesions than in patients with stable lesions. The serum concentration of IFN-β was significantly higher in patients with active lesions (P = 0.028). Compared with healthy individuals, the expression level of the cGAS-STING pathway was elevated in PBMCs of TB patients (P < 0.05), and the difference in the expression level of IFN-β was not statistically significant (P > 0.05), and the serum IFN-β concentration was elevated (P < 0.05). The calculated AUC values for TBK1 and IRF3 in PBMCs, IFN-β in serum and erythrocyte sedimentation rate (ESR) to distinguish between patients with active and stable lesions were 0.732, 0.714, 0.839, and 0.714 respectively. CONCLUSIONS The expression level of TBK1 and IRF3 in PBMCs, and IFN-β in the serum of patients with spinal TB is positively correlated with disease activity. TBK1 has higher specificity and IFN-β in serum has higher sensitivity when used to differentiate between patients with active and stable lesions.
Collapse
Affiliation(s)
- Xianglin Peng
- Department of Orthopedics, Tongji Medical College, Traditional Chinese and Western Medicine Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Feifei Pu
- Department of Orthopedics, Tongji Medical College, Traditional Chinese and Western Medicine Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Fangzheng Zhou
- Department of Orthopedics, Tongji Medical College, Traditional Chinese and Western Medicine Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Xiyong Dai
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430022, China
| | - Feng Xu
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430022, China
| | - Junwen Wang
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China
| | - Jing Feng
- Department of Orthopedics, Tongji Medical College, Traditional Chinese and Western Medicine Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China.
| | - Ping Xia
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China.
| |
Collapse
|
35
|
Liu L, Wen C, Cai X, Gong W. A Novel Bi-Directional Channel for Nutrient Uptake across Mycobacterial Outer Envelope. Microorganisms 2024; 12:1827. [PMID: 39338501 PMCID: PMC11434571 DOI: 10.3390/microorganisms12091827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
Nutrients are absorbed by special transport proteins on the cell membrane; however, there is less information regarding transporters across the mycobacterial outer envelope, which comprises dense and intricate structures. In this study, we focus on the model organism Mycolicibacterium smegmatis, which has a cell envelope similar to that of Mycobacterium tuberculosis, as well as on the TiME protein secretion tube across the mycobacterial outer envelope. We present transcriptome results and analyze the protein compositions of a mycobacterial surface envelope, determining that more transporters and porins are induced to complement the deletion of the time gene in Mycolicibacterium smegmatis. The TiME protein is essential for nutrient utilization, as demonstrated in the uptake experiments and growth on various monosaccharides or with amino acids as the sole carbon source. Its deletion caused bacteria to be more sensitive to anti-TB drugs and to show a growth defect at an acid pH level, indicating that TiME promotes the survival of M. smegmatis in antibiotic-containing and acidic environments. These results suggest that TiME tubes facilitate bi-directional processes for both protein secretion and nutrient uptake across the mycobacterial outer envelope.
Collapse
Affiliation(s)
- Lei Liu
- Division of Life Sciences and Medicine, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Chongzheng Wen
- Division of Life Sciences and Medicine, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Xiaoying Cai
- Division of Life Sciences and Medicine, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Weimin Gong
- Division of Life Sciences and Medicine, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
36
|
Sinha V, Singh A, Singh A, Saraswati SSK, Rana AK, Kalra K, Natarajan K. Potassium ion channel Kir2.1 negatively regulates protective responses to Mycobacterium bovis BCG. J Leukoc Biol 2024; 116:644-656. [PMID: 38489665 DOI: 10.1093/jleuko/qiae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Tuberculosis caused by the pathogen Mycobacterium tuberculosis leads to increased mortality and morbidity worldwide. The prevalence of highly drug-resistant strains has reinforced the need for greater understanding of host-pathogen interactions at the cellular and molecular levels. Our previous work demonstrated critical roles of calcium ion channels in regulating protective responses to mycobacteria. In this report, we deciphered the roles of inwardly rectifying K+ ion channel Kir2.1 in epithelial cells. Data showed that infection of epithelial cells (and macrophages) increases the surface expression of Kir2.1. This increased expression of Kir2.1 results in higher intracellular mycobacterial survival, as either inhibiting or knocking down Kir2.1 results in mounting of a higher oxidative burst leading to a significant attenuation of mycobacterial survival. Further, inhibiting Kir2.1 also led to increased expression of T cell costimulatory molecules accompanied with increased activation of MAP kinases and transcription factors nuclear factor κB and phosphorylated CREB. Furthermore, inhibiting Kir2.1 induced increased autophagy and apoptosis that could also contribute to decreased bacterial survival. Interestingly, an increased association of heat shock protein 70 kDa with Kir2.1 was observed. These results showed that mycobacteria modulate the expression and function of Kir2.1 in epithelial cells to its advantage.
Collapse
Affiliation(s)
- Vishal Sinha
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Akshita Singh
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Aarti Singh
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Shakuntala Surender Kumar Saraswati
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Ankush Kumar Rana
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Kanika Kalra
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Krishnamurthy Natarajan
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| |
Collapse
|
37
|
Zhang J, He Y, Ruan Q, Bi A, Zhou J, Weng S, Ma H, Lin T, Wang H, Xu Y. The hsa_circ_0002371/hsa-miR-502-5p/ATG16L1 axis modulates the survival of intracellular Mycobacterium tuberculosis and autophagy in macrophages. Cell Signal 2024; 121:111271. [PMID: 38944259 DOI: 10.1016/j.cellsig.2024.111271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/11/2024] [Accepted: 06/22/2024] [Indexed: 07/01/2024]
Abstract
Circular RNAs (circRNAs) play a critical role in pathological mechanisms of Mycobacterium tuberculosis (Mtb) and can be used as a new biomarker for active tuberculosis (ATB) diagnosis. Therefore, we identified significantly dysregulated circRNAs in ATB patients and healthy controls (HC) and explored their molecular mechanism. We found that hsa_circ_0002371 was significantly up-regulated in PBMCs of ATB patients and Mycobacterium tuberculosis H37Rv- or Mycobacterium bovis bacillus Calmette Guerin (BCG)-infected THP-1 cells. Functional experiments demonstrated that hsa_circ_0002371 inhibited autophagy in BCG-infected THP-1 cells and promoted intracellular BCG survival rate. In terms of mechanism, hsa_circ_0002371 facilitated the expression of hsa-miR-502-5p, as shown by bioinformatics and dual-luciferase reporter gene analysis, respectively. Notably, hsa-miR-502-5p inhibited autophagy via suppressing autophagy related 16 like 1 (ATG16L1) in BCG-infected macrophages and thus promoting intracellular BCG growth. In summation, hsa_circ_0002371 increased the suppression of hsa-miR-502-5p on ATG16L1 and inhibited autophagy to promote Mtb growth in macrophages. In Conclusion, our data suggested that hsa_circ_0002371 was significantly up-regulated in the PBMCs of ATB patients compared with HC. The hsa_circ_0002371/hsa-miR-502-5p/ATG16L1 axis promoted the survival of intracellular Mtb and inhibited autophagy in macrophages. Our findings suggested hsa_circ_0002371 could act as a potential diagnostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Jinyi Zhang
- Institute of Genetics, School of Life Science, Fudan University, Shanghai 200433, China
| | - Yumo He
- Institute of Genetics, School of Life Science, Fudan University, Shanghai 200433, China
| | - Qiaoling Ruan
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Aixiao Bi
- Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Jingyu Zhou
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shufeng Weng
- Institute of Genetics, School of Life Science, Fudan University, Shanghai 200433, China
| | - Huixia Ma
- Institute of Genetics, School of Life Science, Fudan University, Shanghai 200433, China
| | - Taiyue Lin
- Institute of Genetics, School of Life Science, Fudan University, Shanghai 200433, China
| | - Honghai Wang
- Institute of Genetics, School of Life Science, Fudan University, Shanghai 200433, China
| | - Ying Xu
- Institute of Genetics, School of Life Science, Fudan University, Shanghai 200433, China.
| |
Collapse
|
38
|
Carnero Canales CS, Marquez Cazorla JI, Marquez Cazorla RM, Roque-Borda CA, Polinário G, Figueroa Banda RA, Sábio RM, Chorilli M, Santos HA, Pavan FR. Breaking barriers: The potential of nanosystems in antituberculosis therapy. Bioact Mater 2024; 39:106-134. [PMID: 38783925 PMCID: PMC11112550 DOI: 10.1016/j.bioactmat.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/17/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, continues to pose a significant threat to global health. The resilience of TB is amplified by a myriad of physical, biological, and biopharmaceutical barriers that challenge conventional therapeutic approaches. This review navigates the intricate landscape of TB treatment, from the stealth of latent infections and the strength of granuloma formations to the daunting specters of drug resistance and altered gene expression. Amidst these challenges, traditional therapies often fail, contending with inconsistent bioavailability, prolonged treatment regimens, and socioeconomic burdens. Nanoscale Drug Delivery Systems (NDDSs) emerge as a promising beacon, ready to overcome these barriers, offering better drug targeting and improved patient adherence. Through a critical approach, we evaluate a spectrum of nanosystems and their efficacy against MTB both in vitro and in vivo. This review advocates for the intensification of research in NDDSs, heralding their potential to reshape the contours of global TB treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Cesar Augusto Roque-Borda
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Giulia Polinário
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | | | - Rafael Miguel Sábio
- School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, the Netherlands
| | - Marlus Chorilli
- School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Hélder A. Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Fernando Rogério Pavan
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| |
Collapse
|
39
|
Kassem AF, Sabt A, Korycka-Machala M, Shaldam MA, Kawka M, Dziadek B, Kuzioła M, Dziadek J, Batran RZ. New coumarin linked thiazole derivatives as antimycobacterial agents: Design, synthesis, enoyl acyl carrier protein reductase (InhA) inhibition and molecular modeling. Bioorg Chem 2024; 150:107511. [PMID: 38870705 DOI: 10.1016/j.bioorg.2024.107511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Tuberculosis is a global serious problem that imposes major health, economic and social challenges worldwide. The search for new antitubercular drugs is extremely important which could be achieved via inhibition of different druggable targets. Mycobacterium tuberculosis enoyl acyl carrier protein reductase (InhA) enzyme is essential for the survival of M. tuberculosis. In this investigation, a series of coumarin based thiazole derivatives was synthesized relying on a molecular hybridization approach and was assessed against thewild typeMtb H37Rv and its mutant strain (ΔkatG) via inhibiting InhA enzyme. Among the synthesized derivatives, compounds 2b, 3i and 3j were the most potent against wild type M. tuberculosis with MIC values ranging from 6 to 8 μg/ mL and displayed low cytotoxicity towards mouse fibroblasts at concentrations 8-13 times higher than the MIC values. The three hybrids could also inhibit the growth of ΔkatGmutant strain which is resistant to isoniazid (INH). Compounds 2b and 3j were able to inhibit the growth of mycobacteria inside human macrophages, indicating their ability to penetrate human professional phagocytes. The two derivatives significantly suppress mycobacterial biofilm formation by 10-15 %. The promising target compounds were also assessed for their inhibitory effect against InhA and showed potent effectiveness with IC50 values of 0.737 and 1.494 µM, respectively. Molecular docking studies revealed that the tested compounds occupied the active site of InhA in contact with the NAD+ molecule. The 4-phenylcoumarin aromatic system showed binding interactions within the hydrophobic pocket of the active site. Furthermore, H-bond formation and π -π stacking interactions were also recorded for the promising derivatives.
Collapse
Affiliation(s)
- Asmaa F Kassem
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Ahmed Sabt
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Małgorzata Korycka-Machala
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Malwina Kawka
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Bożena Dziadek
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Magdalena Kuzioła
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, Lodz, Poland
| | - Jarosław Dziadek
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland.
| | - Rasha Z Batran
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt.
| |
Collapse
|
40
|
Zheng Q, Li Z, Zhou Y, Li Y, Gong M, Sun H, Deng X, Ma Y. Heparin-Binding Hemagglutinin of Mycobacterium tuberculosis Inhibits Autophagy via Toll-like Receptor 4 and Drives M2 Polarization in Macrophages. J Infect Dis 2024; 230:323-335. [PMID: 38266152 DOI: 10.1093/infdis/jiae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 01/09/2024] [Accepted: 01/23/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Tuberculosis (TB), predominantly caused by Mycobacterium tuberculosis (MTB) infection, remains a prominent global health challenge. Macrophages are the frontline defense against MTB, relying on autophagy for intracellular bacterial clearance. However, MTB can combat and evade autophagy, and it influences macrophage polarization, facilitating immune evasion and promoting infection. We previously found that heparin-binding hemagglutinin (HBHA) inhibits autophagy in A549 cells; however, its role in macrophage autophagy and polarization remains unclear. METHODS Bacterial cultures, cell cultures, Western blotting, immunofluorescence, macrophage infection assays, siRNA knockdown, and enzyme-linked immunosorbent assay were used to investigate HBHA's impact on macrophages and its relevance in Mycobacterium infection. RESULTS HBHA inhibited macrophage autophagy. Expression of recombinant HBHA in Mycobacterium smegmatis (rMS-HBHA) inhibited autophagy, promoting bacterial survival within macrophages. Conversely, HBHA knockout in the Mycobacterium bovis bacillus Calmette-Guérin (BCG) mutant (BCG-ΔHBHA) activated autophagy and reduced bacterial survival. Mechanistic investigations revealed that HBHA may inhibit macrophage autophagy through the Toll-like receptor 4-dependent PI3K-AKT-mTOR signaling pathway. Furthermore, HBHA induced macrophage M2 polarization. CONCLUSIONS Mycobacterium may exploit HBHA to suppress the antimicrobial immune response in macrophages, facilitating intracellular survival and immune evasion through autophagy inhibition and M2 polarization induction. Our findings may help identify novel therapeutic targets and develop more effective treatments against MTB infection.
Collapse
Affiliation(s)
- Qing Zheng
- Department of Laboratory Medicine, Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital
| | - Zhi Li
- Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences
| | - Yu Zhou
- Department of Laboratory Medicine, Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital
| | - Yuru Li
- Department of Laboratory Medicine, Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital
| | - Meiliang Gong
- Department of Laboratory Medicine, Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital
| | - Heqiang Sun
- Department of Laboratory Medicine, Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital
| | - Xinli Deng
- Department of Laboratory Medicine, Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital
| | - Yueyun Ma
- Department of Clinical Laboratory, Air Force Medical Center, Beijing, China
| |
Collapse
|
41
|
Wang J, Cao H, Xie Y, Xu Z, Li Y, Luo H. Mycobacterium tuberculosis infection induces a novel type of cell death: Ferroptosis. Biomed Pharmacother 2024; 177:117030. [PMID: 38917759 DOI: 10.1016/j.biopha.2024.117030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024] Open
Abstract
Ferroptosis is a lipid peroxidation-driven and iron-dependent form of programmed cell death, which is involved in a variety of physical processes and multiple diseases. Numerous reports have demonstrated that ferroptosis is closely related to the pathophysiological processes of Mycobacterium tuberculosis (M. tuberculosis) infection and is characterized by the accumulation of excess lipid peroxides on the cell membrane. In this study, the various functions of ferroptosis, and the therapeutic strategies and diagnostic biomarkers of tuberculosis, were summarized. Notably, this review provides insights into the molecular mechanisms and functions of M. tuberculosis-induced ferroptosis, suggesting potential future therapeutic and diagnostic markers for tuberculosis.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou 215300, PR China
| | - Hui Cao
- Department of Food and Nutrition Safety, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu 210009, PR China
| | - Yiping Xie
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou 215300, PR China
| | - Zi Xu
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou 215300, PR China
| | - Yujie Li
- Department of Clinical Laboratory, Kunshan Hospital Affiliated to Jiangsu University, Suzhou 215300, PR China
| | - Hao Luo
- Department of Clinical Laboratory, The Second People's Hospital of Kunshan, Suzhou, China.
| |
Collapse
|
42
|
Priyanka, Sharma S, Sharma M. Role of PE/PPE proteins of Mycobacterium tuberculosis in triad of host mitochondria, oxidative stress and cell death. Microb Pathog 2024; 193:106757. [PMID: 38908454 DOI: 10.1016/j.micpath.2024.106757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The PE and PPE family proteins of Mycobacterium tuberculosis (Mtb) is exclusively found in pathogenic Mycobacterium species, comprising approximately 8-10 % of the Mtb genome. These emerging virulent factors have been observed to play pivotal roles in Mtb pathogenesis and immune evasion through various strategies. These immunogenic proteins are known to modulate the host immune response and cell-death pathways by targeting the powerhouse of the cell, the mitochondria to support Mtb survival. In this article, we are focused on how PE/PPE family proteins target host mitochondria to induce mitochondrial perturbations, modulate the levels of cellular ROS (Reactive oxygen species) and control cell death pathways. We observed that the time of expression of these proteins at different stages of infection is crucial for elucidating their impact on the cell death pathways and eventually on the outcome of infection. This article focuses on understanding the contributions of the PE/PPE proteins by unravelling the triad of host mitochondria, oxidative stress and cell death pathways that facilitate the Mtb persistence. Understanding the role of these proteins in host cellular pathways and the intricate mechanisms paves the way for the development of novel therapeutic strategies to combat TB infections.
Collapse
Affiliation(s)
- Priyanka
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Sadhna Sharma
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Monika Sharma
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
43
|
Zhang X, Ding G, Yang X, Lu H, Xu Y, Hu Y, Liu S, Zhang H, Huang K, Deng G, Ye T, Yu Q, Cai Y, Xie S, Wang W, Chen X. Myriocin enhances the clearance of M. tuberculosis by macrophages through the activation of PLIN2. mSphere 2024; 9:e0025724. [PMID: 38920406 PMCID: PMC11288015 DOI: 10.1128/msphere.00257-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/19/2024] [Indexed: 06/27/2024] Open
Abstract
Myriocin is an inhibitor of de novo synthesis of sphingolipids and ceramides. In this research, we showed myriocin could significantly reduce Mtb burden and histopathological inflammation in mice. However, the underlying mechanism remains unclear. RNA-seq analysis revealed a significant increase in gene expression of PLIN2/CD36/CERT1 after myriocin treatment. The reduced bactericidal burden was only reversed after silencing the lipid droplets (LDs) surface protein PLIN2. This suggests that myriocin enhances the ability of macrophages to clear Mtb depending on the PLIN2 gene, which is part of the PPARγ pathway. Indeed, we observed a significant increase in the number of LDs following myriocin treatment.IMPORTANCEMycobacterium tuberculosis has the ability to reprogram host cell lipid metabolism and alter the antimicrobial functions of infected macrophages. The sphingolipids, such as ceramides, are the primary host lipids utilized by the bacteria, making the sphingomyelinase/ceramide system critical in Mtb infections. Surprisingly, the antimicrobial effect of myriocin was found to be independent of its role in reducing ceramides, but instead, it depends on the lipid droplets surface protein PLIN2. Our findings provide a novel mechanism for how myriocin enhances Mtb clearance in macrophages.
Collapse
Affiliation(s)
- Ximeng Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Guanggui Ding
- Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xirui Yang
- Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Hailin Lu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yuzhong Xu
- Department of Clinical Laboratory, Shenzhen Baoan Hospital, Shenzhen, China
| | - Yunlong Hu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Song Liu
- Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Huihua Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Kaisong Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Guofang Deng
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Taosheng Ye
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Qing Yu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yi Cai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Shuixiang Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Wenfei Wang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
44
|
Tseng YH, Pan SW, Huang JR, Lee CC, Hung JJ, Hsu PK, Chen NJ, Su WJ, Chen YM, Feng JY. Promoter methylation and increased expression of PD-L1 in patients with active tuberculosis. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:278. [PMID: 39081906 PMCID: PMC11287217 DOI: 10.15698/mic2024.07.832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 05/04/2024] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
The PD-1/PD-L1 pathway plays a pivotal role in T cell activity and is involved in the pathophysiology of Mycobacterium tuberculosis (MTB) infection. DNA methylation is a mechanism that modulates PD-L1 expression in cancer cells. However, its effect on PD-L1 expression in macrophages after MTB infection remains unknown. We prospectively enrolled patients with active tuberculosis (TB) and non-TB subjects. The expression of PD-L1 and methylation-related genes in peripheral blood mononuclear cells (PBMCs) were investigated and their correlation with disease severity and treatment outcomes were examined. PD-L1 promoter methylation status was evaluated using bisulfite sequencing. Immunohistochemistry (IHC) and immunofluorescence (IF) staining were used to visualize PD-L1- and TET-1-expressing cells in lung tissues from patients with TB and in macrophage cell lines with MTB-related stimulation. In total, 80 patients with active TB and 40 non-TB subjects were enrolled in the analysis. Patients with active TB had significantly higher expression of PD-L1, DNMT3b, TET1, TET2, and lower expression of DNMT1, compared to that in the non-TB subjects. The expression of PD-L1 and TET-1 was significantly associated with 1-month smear and culture non-conversion. IHC and IF staining demonstrated the co-localization of PD-L1- and TET-1-expressing macrophages in patients with pulmonary TB and in human macrophage cell lines after MTB-related stimulation. DNMT inhibition and TET-1 knockdown in human macrophages increased and decreased PD-L1 expression, respectively. Overall, PD-L1 expression is increased in patients with active TB and is correlated with treatment outcomes. DNA methylation is involved in modulating PD-L1 expression in human macrophages.
Collapse
Affiliation(s)
- Yen-Han Tseng
- Department of Chest Medicine, Taipei Veterans General HospitalTaipei, 112Taiwan
- School of Medicine, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
| | - Sheng-Wei Pan
- Department of Chest Medicine, Taipei Veterans General HospitalTaipei, 112Taiwan
- School of Medicine, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
- Institute of Public Health, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
| | - Jhong-Ru Huang
- School of Medicine, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
- Division of Chest Medicine, Department of Internal Medicine, Taichung Hospital, Ministry of Health and WelfareTaichungTaiwan
| | - Chang-Ching Lee
- Department of Chest Medicine, Taipei Veterans General HospitalTaipei, 112Taiwan
| | - Jung-Jyh Hung
- School of Medicine, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General HospitalTaipei, 112Taiwain
| | - Po-Kuei Hsu
- School of Medicine, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General HospitalTaipei, 112Taiwain
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, School of Life Sciences, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
| | - Wei-Juin Su
- School of Medicine, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
- Division of Chest Medicine, China Medical University Hospital, Taipei BranchTaipei, 114Taiwan
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General HospitalTaipei, 112Taiwan
- School of Medicine, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
| | - Jia-Yih Feng
- Department of Chest Medicine, Taipei Veterans General HospitalTaipei, 112Taiwan
- School of Medicine, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung UniversityTaipei, 112Taiwan
| |
Collapse
|
45
|
Lo TH, Weng IC, Chen HL, Liu FT. The role of galectins in the regulation of autophagy and inflammasome in host immunity. Semin Immunopathol 2024; 46:6. [PMID: 39042263 DOI: 10.1007/s00281-024-01018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024]
Abstract
Galectins, a family of glycan-binding proteins have been shown to bind a wide range of glycans. In the cytoplasm, these glycans can be endogenous (or "self"), originating from damaged endocytic vesicles, or exogenous (or "non-self"), found on the surface of invading microbial pathogens. Galectins can detect these unusual cytosolic exposures to glycans and serve as critical regulators in orchestrating immune responses in innate and adaptive immunity. This review provides an overview of how galectins modulate host cellular responses, such as autophagy, xenophagy, and inflammasome-dependent cell death program, to infection.
Collapse
Affiliation(s)
- Tzu-Han Lo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - I-Chun Weng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Hung-Lin Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
- Department of Dermatology, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
46
|
AlSaeed H, Haider MJA, Alzaid F, Al-Mulla F, Ahmad R, Al-Rashed F. PPARdelta: A key modulator in the pathogenesis of diabetes mellitus and Mycobacterium tuberculosis co-morbidity. iScience 2024; 27:110046. [PMID: 38989454 PMCID: PMC11233913 DOI: 10.1016/j.isci.2024.110046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/20/2024] [Accepted: 05/17/2024] [Indexed: 07/12/2024] Open
Abstract
The interplay between lipid metabolism and immune response in macrophages plays a pivotal role in various infectious diseases, notably tuberculosis (TB). Herein, we illuminate the modulatory effect of heat-killed Mycobacterium tuberculosis (HKMT) on macrophage lipid metabolism and its implications on the inflammatory cascade. Our findings demonstrate that HKMT potently activates the lipid scavenger receptor, CD36, instigating lipid accumulation. While CD36 inhibition mitigated lipid increase, it unexpectedly exacerbated the inflammatory response. Intriguingly, this paradoxical effect was linked to an upregulation of PPARδ. Functional analyses employing PPARδ modulation revealed its central role in regulating both lipid dynamics and inflammation, suggesting it as a potential therapeutic target. Moreover, primary monocytic cells from diabetic individuals, a demographic at amplified risk of TB, exhibited heightened PPARδ expression and inflammation, further underscoring its pathological relevance. Targeting PPARδ in these cells effectively dampened the inflammatory response, offering a promising therapeutic avenue against TB.
Collapse
Affiliation(s)
- Halemah AlSaeed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, State of Kuwait
| | - Mohammed J A Haider
- Department of Biological Sciences, Faculty of Science, Kuwait University, PO BOX 5969, Safat 13060, State of Kuwait
| | - Fawaz Alzaid
- Bioenergetics Department, Dasman Diabetes Institute, Kuwait City 15462, Kuwait
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, 75015 Paris, France
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, State of Kuwait
| | - Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, State of Kuwait
| |
Collapse
|
47
|
Batran RZ, Sabt A, Dziadek J, Kassem AF. Design, synthesis and computational studies of new azaheterocyclic coumarin derivatives as anti- Mycobacterium tuberculosis agents targeting enoyl acyl carrier protein reductase (InhA). RSC Adv 2024; 14:21763-21777. [PMID: 38984262 PMCID: PMC11232110 DOI: 10.1039/d4ra02746a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024] Open
Abstract
In this study, we designed and synthesized a series of coumarin derivatives as antitubercular agents targeting the enoyl acyl carrier protein reductase (InhA) enzyme. Among the synthesized compounds, the tetrazole derivative 4c showed the most potent antitubercular effect with a minimum inhibitory concentration value (MIC) of 15 μg mL-1 against Mtb H37Rv and could also inhibit the growth of the mutant strain (ΔkatG). Compound 4c was able to penetrate Mtb-infected human macrophages and suppress the intracellular growth of tubercle bacilli. Moreover, the target derivative 4c showed a potent inhibitory effect against InhA enzyme with an IC50 value of 0.565 μM, which was superior to the reference InhA inhibitor triclosan. Molecular docking of compound 4c within the InhA active site revealed the importance of the 4-phenylcoumarin ring system and tetrazole moiety for activity. Finally, the physicochemical properties and pharmacokinetic parameters of 4c were investigated.
Collapse
Affiliation(s)
- Rasha Z Batran
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki Cairo 12622 Egypt
| | - Ahmed Sabt
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki Cairo 12622 Egypt
| | - Jarosław Dziadek
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology of the Polish Academy of Sciences Lodz Poland
| | - Asmaa F Kassem
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki Cairo 12622 Egypt
| |
Collapse
|
48
|
Ramachandran G, Yeruva CV, Swarup G, Raghunand TR. A cytoprotective role for optineurin during mycobacterial infection of macrophages. Biochem Biophys Rep 2024; 38:101672. [PMID: 38434142 PMCID: PMC10907145 DOI: 10.1016/j.bbrep.2024.101672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
Autophagy has emerged as a critical innate immune mechanism for host elimination of intracellular pathogens, however, the role of the autophagy receptor Optineurin during mycobacterial infection is not fully understood. To address this lacuna, we infected bone marrow-derived macrophages (BMDMs) derived from Optn+/+ and Optn-/- mice with Mycobacterium smegmatis, and observed the infection outcome at sequential time points. While low multiplicity of infection (MOI) did not show any significant difference between BMDMs from the two groups, at high MOI Optn-/- mice-derived BMDMs showed significantly lower colony forming unit counts, as well as lower cell counts at 12 h and 24 h post-infection. Quantification of cell numbers and nuclear morphologies at various time points post-infection indicated a markedly higher cell death in the Optineurin-deficient macrophages. Optineurin-deficient BMDMs showed significantly lower levels of the autophagosomal protein LC3-II upon infection, indicating a potential role for Optineurin in regulating autophagy during mycobacterial infection. Moreover, when stimulated by bacterial LPS, Optineurin deficient macrophages, showed altered levels of the inflammatory cytokine pro-IL-1β. These observations taken together suggest a novel regulatory role for Optineurin during mycobacterial infection. Its deficiency leads to an impairment in macrophage responses, directly impacting the pathophysiology of infection.
Collapse
Affiliation(s)
| | | | - Ghanshyam Swarup
- CSIR - Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tirumalai R. Raghunand
- CSIR - Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
49
|
Kaur J, Deshmukh PT, Gaurkar SS. Otorhinolaryngologic Manifestations of Tuberculosis: A Comprehensive Review of Clinical and Diagnostic Challenges. Cureus 2024; 16:e64586. [PMID: 39144871 PMCID: PMC11323964 DOI: 10.7759/cureus.64586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Tuberculosis (TB) is a significant global health issue, predominantly affecting the lungs but also capable of involving the otorhinolaryngologic (ear, nose, and throat) regions. This comprehensive review explores the epidemiology, pathophysiology, clinical presentation, diagnostic challenges, management strategies, and public health implications of otorhinolaryngologic TB. The disease's diverse clinical manifestations, such as chronic ear discharge, nasal obstruction, and hoarseness, often mimic other common conditions, complicating diagnosis and delaying treatment. Diagnostic confirmation requires a combination of clinical assessment, laboratory tests, and imaging techniques, each with inherent limitations. Effective management necessitates a multidisciplinary approach, integrating medical and surgical interventions tailored to individual patient needs. Potential complications, including airway obstruction and hearing loss, highlight the importance of timely and appropriate treatment. The review underscores the critical role of public health measures in TB control. It also identifies emerging trends in diagnosis and treatment, emphasizing the need for ongoing research to improve patient outcomes and contribute to the global effort to control and eventually eradicate TB. This review aims to give healthcare providers a deeper understanding of otorhinolaryngologic TB, enhancing diagnostic and therapeutic approaches and improving patient care.
Collapse
Affiliation(s)
- Jasleen Kaur
- Otolaryngology - Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Prasad T Deshmukh
- Otolaryngology - Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sagar S Gaurkar
- Otolaryngology - Head and Neck Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
50
|
Wu Y, Riehle A, Pollmeier B, Kadow S, Schumacher F, Drab M, Kleuser B, Gulbins E, Grassmé H. Caveolin-1 affects early mycobacterial infection and apoptosis in macrophages and mice. Tuberculosis (Edinb) 2024; 147:102493. [PMID: 38547568 DOI: 10.1016/j.tube.2024.102493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/09/2024] [Accepted: 02/11/2024] [Indexed: 06/14/2024]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis, remains one of the deadliest infections in humans. Because Mycobacterium bovis Bacillus Calmette-Guérin (BCG) share genetic similarities with Mycobacterium tuberculosis, it is often used as a model to elucidate the molecular mechanisms of more severe tuberculosis infection. Caveolin-1 has been implied in many physiological processes and diseases, but it's role in mycobacterial infections has barely been studied. We isolated macrophages from Wildtype or Caveolin-1 deficient mice and analyzed hallmarks of infection, such as internalization, induction of autophagy and apoptosis. For in vivo assays we intravenously injected mice with BCG and investigated tissues for bacterial load with colony-forming unit assays, bioactive lipids with mass spectrometry and changes of protein expressions by Western blotting. Our results revealed that Caveolin-1 was important for early killing of BCG infection in vivo and in vitro, controlled acid sphingomyelinase (Asm)-dependent ceramide formation, apoptosis and inflammatory cytokines upon infection with BCG. In accordance, Caveolin-1 deficient mice and macrophages showed higher bacterial burdens in the livers. The findings indicate that Caveolin-1 plays a role in infection of mice and murine macrophages with BCG, by controlling cellular apoptosis and inflammatory host response. These clues might be useful in the fight against tuberculosis.
Collapse
Affiliation(s)
- Yuqing Wu
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Andrea Riehle
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Barbara Pollmeier
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Stephanie Kadow
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | | | - Marek Drab
- Unit of Nanostructural Biointeractions, Department of Immunology of Infectious Diseases, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 12 Weigla Street, 53-114, Wroclaw, Poland
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Erich Gulbins
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Heike Grassmé
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|