1
|
Cai J, Zhou H, Liu M, Zhang D, Lv J, Xue H, Zhou H, Zhang W. Host immunity and intracellular bacteria evasion mechanisms: Enhancing host-directed therapies with drug delivery systems. Int J Antimicrob Agents 2025; 65:107492. [PMID: 40107461 DOI: 10.1016/j.ijantimicag.2025.107492] [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: 10/31/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Host-directed therapies (HDTs) have been investigated as a potential solution to combat intracellular and drug-resistant bacteria. HDTs stem from extensive research on the intricate interactions between the host and intracellular bacteria, leading to a treatment approach that relies on immunoregulation. To improve the bioavailability and safety of HDTs, researchers have utilized diverse drug delivery systems (DDS) to encapsulate and transport therapeutic agents to target cells. In this review, we first introduce the three mechanisms of bactericidal action and intracellular bacterial evasion: autophagy, reactive oxygen species (ROS), and inflammatory cytokines, with a particular focus on autophagy. Special attention is given to the detailed mechanism of xenophagy in clearing intracellular bacteria, a crucial selective autophagy process that specifically targets and degrades intracellular pathogens. Following this, we present the application of DDS to modulate these regulatory methods for intracellular bacteria elimination. By integrating insights from immunology and nanomedicine, this review highlights the emerging role of DDS in advancing HDTs for intracellular bacterial infections and paving the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Jiayang Cai
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Han Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Mingwei Liu
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Dingjian Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Jingxuan Lv
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Haokun Xue
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Houcheng Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China.
| |
Collapse
|
2
|
Bi J, Song L, Guo Q, Chen X, Gong Y, Wu H, Zhang F, Wang J, Zhang G. Effect of urolithin A on intracellular survival of Mycobacterium tuberculosis by regulating AKT-FOXO1-mediated autophagy. mSphere 2025; 10:e0006125. [PMID: 40207917 DOI: 10.1128/msphere.00061-25] [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: 01/27/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Tuberculosis (TB), resulting from Mycobacterium tuberculosis (Mtb), is one of the leading causes of morbidity and mortality in humans worldwide. Host-directed therapy (HDT) is a novel approach for treating TB, particularly those with drug resistance. Urolithin A (UroA) produced through bioconversion of plant-derived ellagic acid by gut microbes has been proven to have multiple beneficial effects in a variety of diseases without showing undesired adverse reactions. However, whether UroA has antimycobacterial effect and the underlying mechanism has not yet been reported. Here, we found that UroA significantly inhibited Mtb growth within both macrophages and mice. Moreover, UroA promoted the activation of autophagy in Mtb-infected macrophages via the protein kinase B-Forkhead box protein O1 signaling pathway, which contributed to the antimycobacterial effect of UroA. Additionally, UroA suppressed the survival of clinically isoniazid (INH)-resistant Mtb (C2) within macrophages, and the combination of UroA and INH synergistically enhanced host elimination of Mtb H37Rv. Therefore, UroA may be utilized as a potential candidate for HDT and as an adjunctive therapy with first-line anti-TB drugs.IMPORTANCEHost-directed therapy (HDT) is a novel approach for treating tuberculosis (TB), particularly those with drug resistance. Urolithin A (UroA) produced through bioconversion of plant-derived ellagic acid by gut microbes has been proven to have multiple beneficial effects in a variety of diseases without showing undesired adverse reactions. We found that UroA significantly inhibited Mycobacterium tuberculosis (Mtb) growth within macrophages. Moreover, UroA suppressed the survival of clinically isoniazid (INH)-resistant Mtb (C2) within macrophages, and the combination of UroA and INH synergistically enhanced host elimination of Mtb H37Rv. Therefore, UroA may be utilized as a potential candidate for HDT and as an adjunctive therapy with first-line anti-TB drugs.
Collapse
Affiliation(s)
- Jing Bi
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southwest Medical University, Shenzhen, China
| | - Li Song
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southwest Medical University, Shenzhen, China
| | - Qinglong Guo
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southwest Medical University, Shenzhen, China
| | - Xi Chen
- School of Public Health, Guangdong Medical University, Dongguan, China
| | - Yaqi Gong
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Haojia Wu
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southwest Medical University, Shenzhen, China
| | - Fan Zhang
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southwest Medical University, Shenzhen, China
| | - Jingbin Wang
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Guoliang Zhang
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southwest Medical University, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
3
|
Zhang K, Du Z, Wang Z, Chen Y, Zhang L, Chen X, Guo A. sRNA ncBCG427 activates the expression of target gene MSMEG_4757 to enhance the survival of Mycobacterium smegmatis through lipid metabolism in adverse environments. World J Microbiol Biotechnol 2025; 41:185. [PMID: 40415042 DOI: 10.1007/s11274-025-04343-5] [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: 01/31/2025] [Accepted: 03/28/2025] [Indexed: 05/27/2025]
Abstract
The capability of mycobacteria to survive in adverse environments is crucial for successful infection, yet the underlying mechanisms remain unclear. A novel sRNA, ncBCG427, was previously identified in intracellular versus extracellular mycobacteria, with predicted targets clustering in lipid metabolism pathways in Mycobacterium smegmatis (M. sm). This study aimed to investigate how ncBCG427 regulates the survival of M. sm through lipid metabolism. Using lipidomics, metabolites from the ncBCG427-expressing strain (MS_ncBCG427) and the control strain (MS_Vector) were screened, revealing enrichment in lipid-associated pathways. The gene MSMEG_4757 (Fas) was identified as critical to this pathway and confirmed as a target of ncBCG427. Western blot analysis demonstrated that ncBCG427 increased Fas expression in THP-1 cells post-infection. Additionally, Oil Red O staining indicated that both ncBCG427 and MSMEG_4757 enhanced lipid droplet formation in A549 cells. Both MS_ncBCG427 and MS_4757 exhibited increased biofilm formation and enhanced survival under various adverse conditions, including carbon starvation, acid stress, membrane stress, and exposure to drugs such as rifampicin and streptomycin. In contrast, low-expression strains (MS_sh4757 and MS_ncBCG427_sh4757) showed reduced survival. In conclusion, ncBCG427 targets MSMEG_4757 to regulate lipid metabolism, enhancing biofilm formation and survival in adverse environments, revealing a novel mechanism of mycobacterial survival and potential antimicrobial targets.
Collapse
Affiliation(s)
- Kailun Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei Province, 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- National Professional Laboratory For Animal Tuberculosis (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zejin Du
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei Province, 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- National Professional Laboratory For Animal Tuberculosis (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zijian Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei Province, 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- National Professional Laboratory For Animal Tuberculosis (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yingyu Chen
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei Province, 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- National Professional Laboratory For Animal Tuberculosis (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Lei Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei Province, 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- National Professional Laboratory For Animal Tuberculosis (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xi Chen
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei Province, 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- National Professional Laboratory For Animal Tuberculosis (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Aizhen Guo
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei Province, 430070, China.
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China.
- National Professional Laboratory For Animal Tuberculosis (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.
- International Research Center for Animal Disease, Ministry of Science and Technology, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
4
|
Li X, Luo X, Wang B, Fu L, Chen X, Lu Y. Sudapyridine (WX-081) inhibits Mycobacterium tuberculosis by targeting ATP synthase and upregulating host innate immunity. mSphere 2025:e0014925. [PMID: 40396746 DOI: 10.1128/msphere.00149-25] [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: 03/06/2025] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
Drug-resistant tuberculosis (DR-TB) urgently requires safer, more accessible alternatives to bedaquiline (BDQ), which faces critical flaws like cardiotoxicity, high costs, and emerging resistance. WX-081, a promising BDQ alternative, has demonstrated superior anti-TB activity and improved safety in clinical studies. However, its mechanism of action remains unexplored, underscoring the need for further research to optimize its potential in advancing global TB elimination efforts. This study reveals WX-081's dual mechanisms: targeting atpE to disrupt ATP synthase and proton motive force via resistance screening, gene sequencing, and functional assays while enhancing host immunity through macrophage transcriptomics. Molecular docking confirmed atpE binding sites, and immune activation pathways (NF-κB/MAPK) were identified, positioning WX-081 as a potent, safe anti-DR-TB candidate despite unresolved mechanistic details.IMPORTANCEBedaquiline, a key drug for drug-resistant tuberculosis, is restricted by safety issues impacting its clinical utility. Its next-generation alternative, WX-081, has advanced to Phase III trials but lacks in-depth studies on its mechanism and host immune-modulatory effects, necessitating further research before broad clinical adoption.
Collapse
Affiliation(s)
- Xinda Li
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaoyi Luo
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Bin Wang
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Lei Fu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xi Chen
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Lu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
5
|
Qiu YH, Zhang YH, Wu ZC, Huang JY, Chen BC, Xiao J, Chen FF. 3,4-Dimethoxychalcone alleviates limb ischemia/reperfusion injury by TFEB-mediated autophagy enhancement and antioxidative response. FASEB J 2025; 39:e70496. [PMID: 40162605 DOI: 10.1096/fj.202402609rr] [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: 10/27/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Caloric restriction mimetics (CRMs) replicate the positive effects of caloric restriction (CR) and have demonstrated therapeutic benefits in neuroinflammatory and cardiovascular diseases. However, it remains uncertain whether CRMs enhance functional recovery following ischemia/reperfusion (I/R) injury, as well as the specific mechanisms involved in this process. This study examines the therapeutic potential of the CRM 3,4-dimethoxychalcone (3,4-DC) in limb I/R injury. Histology, tissue swelling analysis, and laser doppler imaging (LDI) were used to assess the viability of the limbs. Western blotting and immunofluorescence were utilized to examine apoptosis levels, oxidative stress (OS), autophagy, transcription factor EB (TFEB) activity, and mucolipin 1 (MCOLN1)-calcineurin signaling pathway. The administration of 3,4-DC notably alleviated hypoperfusion, tissue swelling, skeletal muscle fiber damage, and cellular injury in the limb caused by I/R. The pharmacological blockade of autophagy negated the antioxidant and antiapoptotic effects of 3,4-DC. Moreover, RNA interference-mediated TFEB silencing eliminated the 3,4-DC-induced restoration of autophagy, antioxidant response, and antiapoptotic effects. Additionally, our findings revealed that 3,4-DC modulates TFEB activity via the MCOLN1-calcineurin signaling pathway. 3,4-DC facilitates functional recovery by enhancing TFEB-driven autophagy, while simultaneously suppressing oxidative stress and apoptosis following I/R injury, suggesting its potential value in clinical applications.
Collapse
Affiliation(s)
- Yi-Hui Qiu
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yin-He Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Zi-Chang Wu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Jing-Yong Huang
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bi-Cheng Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Xiao
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Fan-Feng Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Pfau DJ, Bryk R. High throughput screening assay for the identification of ATF4 and TFEB activating compounds. AUTOPHAGY REPORTS 2025; 4:2473765. [PMID: 40265045 PMCID: PMC11980509 DOI: 10.1080/27694127.2025.2473765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 04/24/2025]
Abstract
Macrophages act to defend against infection, but can fail to completely prevent bacterial replication and dissemination in an immunocompetent host. Recent studies have shown that activation of a host transcription factor, TFEB, a regulator of lysosomal biogenesis, could restrict intramacrophage replication of the human pathogen Mycobacterium tuberculosis and synergize with suboptimal levels of the antibiotic rifampin to reduce bacterial loads. Currently available small molecule TFEB activators lack selectivity and potency, but could be potentially useful in a variety of pathological conditions with suboptimal lysosomal activity. TFEB nuclear translocation and activation depend on its phosphorylation status which is controlled by multiple cellular pathways. We devised a whole cell, high throughput screening assay to identify small molecules that activate TFEB by establishing a stably transfected HEK293T reporter cell line for ATF4, a basic leucine zipper transcription factor induced by stress response and activated in parallel to TFEB. We optimized its use in vitro using compounds that target endoplasmic reticulum stress and intracellular calcium signaling. We report results from screening the commercially available LOPAC library and the Selleck Chemicals library modified to include only FDA-approved drugs and clinical research compounds. We identified twenty-one compounds across six clinical use categories that activate ATF4, and confirmed that two proteasome inhibitors promote TFEB activation. The results of this study provide an assay that could be used to screen for small molecules that activate ATF4 and TFEB and a potential list of compounds identified as activators of the ATF4 transcription factor in response to cellular stress.
Collapse
Affiliation(s)
- Daniel J. Pfau
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Ruslana Bryk
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
7
|
Omoteso OA, Fadaka AO, Walker RB, Khamanga SM. Innovative Strategies for Combating Multidrug-Resistant Tuberculosis: Advances in Drug Delivery Systems and Treatment. Microorganisms 2025; 13:722. [PMID: 40284559 PMCID: PMC12029526 DOI: 10.3390/microorganisms13040722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Multidrug-resistant tuberculosis (MDR-TB) is a significant public health challenge globally, exacerbated by the limited efficacy of existing therapeutic approaches, prolonged treatment duration, and severe side effects. As drug resistance continues to emerge, innovative drug delivery systems and treatment strategies are critical to combating this crisis. This review highlights the molecular mechanisms underlying resistance to drugs in Mycobacterium tuberculosis, such as genetic mutation, efflux pump activity, and biofilm formation, contributing to the persistence and difficulty in eradicating MDR-TB. Current treatment options, including second-line drugs, offer limited effectiveness, prompting the need for innovation of advanced therapies and drug delivery systems. The progression in drug discovery has resulted in the approval of innovative therapeutics, including bedaquiline and delamanid, amongst other promising candidates under investigation. However, overcoming the limitations of traditional drug delivery remains a significant challenge. Nanotechnology has emerged as a promising solution, with nanoparticle-based drug delivery systems offering improved bioavailability and targeted and controlled release delivery, particularly for pulmonary targeting and intracellular delivery to macrophages. Furthermore, the development of inhalable formulations and the potential of nanomedicines to bypass drug resistance mechanisms presents a novel approach to enhancing drug efficacy. Moreover, adjunctive therapies, including immune modulation and host-directed therapies, are being explored to improve treatment outcomes. Immunotherapies, such as cytokine modulation and novel TB vaccines, offer complementary strategies to the use of antibiotics in combating MDR-TB. Personalized medicine approaches, leveraging genomic profiling of both the pathogen and the host, offer promise in optimizing treatment regimens and minimizing drug resistance. This review underscores the importance of multidisciplinary approaches, combining drug discovery, advanced delivery system development, and immune modulation to address the complexities of treating MDR-TB. Continued innovation, global collaboration, and improved diagnostics are essential to developing practical, accessible, and affordable treatments for MDR-TB.
Collapse
Affiliation(s)
- Omobolanle A. Omoteso
- Division of Pharmaceutics, Faculty of Pharmacy, Rhodes University, Makhanda 6139, South Africa; (R.B.W.); (S.M.K.)
| | - Adewale O. Fadaka
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Roderick B. Walker
- Division of Pharmaceutics, Faculty of Pharmacy, Rhodes University, Makhanda 6139, South Africa; (R.B.W.); (S.M.K.)
| | - Sandile M. Khamanga
- Division of Pharmaceutics, Faculty of Pharmacy, Rhodes University, Makhanda 6139, South Africa; (R.B.W.); (S.M.K.)
| |
Collapse
|
8
|
Bi J, Guo Q, Gong Y, Chen X, Wu H, Song L, Xu Y, Ou M, Wang Z, Chen J, Jiang C, Liu A, Li G, Zhang G. Troglitazone Reduction of Intracellular Mycobacterium tuberculosis Survival Via Macrophage Autophagy Through LKB1-AMPKα Signaling. J Infect Dis 2025; 231:e553-e565. [PMID: 39450555 PMCID: PMC11911799 DOI: 10.1093/infdis/jiae523] [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: 04/03/2024] [Revised: 10/07/2024] [Accepted: 10/23/2024] [Indexed: 10/26/2024] Open
Abstract
Tuberculosis caused by Mycobacterium tuberculosis (Mtb), results in significant disease and death worldwide. Host-directed therapy, including conventional drugs, is a promising antituberculosis strategy that shows synergistic antibacterial effects when combined with antituberculosis drugs. Here, the mycobactericidal effect of 3 antidiabetic drugs was examined. Of these, only troglitazone (Trog) enhanced the antimycobacterial effect in vitro and in vivo. This was due to Trog-mediated autophagy activation. Moreover, a knock-down experiment revealed that Trog activated autophagy and exhibited antimycobacterial activity through the serine/threonine-protein kinase STK11 (LKB1)-5'-AMP-activated protein kinase (AMPK) signaling pathway. Molecular docking and coimmunoprecipitation experiments demonstrated that Trog promoted LKB1 phosphorylation and activation by targeting STE20-related kinase adapter protein alpha (STRADA). Finally, we found that Trog inhibited the intracellular survival of clinical isoniazid-resistant Mtb, and the combination of Trog and isoniazid showed additive antibacterial effects against Mtb H37Rv. Taken together, antidiabetic Trog may be repurposed as a candidate for host-directed therapy and combined with first-line antituberculosis drugs.
Collapse
Affiliation(s)
- Jing Bi
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Qinglong Guo
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yaqi Gong
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Xi Chen
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Haojia Wu
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Li Song
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yating Xu
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Min Ou
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Zhaoqin Wang
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Jiean Chen
- Shenzhen Bay Laboratory, Pingshan Translational Medicine Center, Shenzhen, China
| | - Chenran Jiang
- Shenzhen Bay Laboratory, Pingshan Translational Medicine Center, Shenzhen, China
| | - Aimei Liu
- Department of Tuberculosis, Guangxi Chest Hospital, Liuzhou, China
| | - Guobao Li
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Guoliang Zhang
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
9
|
Fadda H, Khan RH, Shqair Y, Uslu C, Panov AV, Lyakhovich A. Antibacterials exert toxic effects on aquatic organisms by inhibiting respiration, inducing oxidative stress, mitochondrial dysfunction and autophagy. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 280:107284. [PMID: 39951902 DOI: 10.1016/j.aquatox.2025.107284] [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: 10/23/2024] [Revised: 02/04/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Although bactericidal antibiotics are an integral part of infectious disease medicine, the uncontrolled use of these drugs in recent years is beginning to affect the environment, especially water resources. With the exception of a few well-documented toxic effects, antimicrobials are thought to act mainly on bacteria and, at low doses, have no deleterious effects on eukaryotic organisms. However, since mitochondria are direct ancient relatives of prokaryotes, mitochondrial dysfunction induced by antibiotic exposure can also occur in higher eukaryotes. In this work, we used the microcrustacean Artemia salina to show how some known antibacterial drugs exert toxic effects on mitochondria, suppressing cellular and organismal respiration in aquatic organisms. We have demonstrated that exposure to antimicrobials of different classes, namely kanamycin A, tetracycline hydrochloride, erythromycin, and chloramphenicol, leads to the accumulation of reactive oxygen species and is accompanied by autophagy. Our results suggest that the emergence of antibactericidal drugs, increasingly appearing in wastewater, may have far-reaching consequences for aquatic areas adjacent to large cities.
Collapse
Affiliation(s)
- Hagar Fadda
- Faculty of Medicine and Science University of Fribourg Avenue de l'Europe 20, Fribourg 1700, Switzerland; Molecular Biology, Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956 Turkey
| | - Raheema Hassan Khan
- Molecular Biology, Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956 Turkey
| | - Yara Shqair
- Molecular Biology, Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956 Turkey
| | - Cemile Uslu
- Molecular Biology, Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956 Turkey
| | - Alexander V Panov
- Department of Biomedical Sciences, School of Medicine, Mercer University, Macon, GA 31201, USA
| | - Alex Lyakhovich
- Molecular Biology, Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956 Turkey.
| |
Collapse
|
10
|
Frandsen JR, Yuan Z, Bedi B, Prasla Z, Choi SR, Narayanasamy P, Sadikot RT. PGC-1α activation to enhance macrophage immune function in mycobacterial infections. PLoS One 2025; 20:e0310908. [PMID: 39913377 PMCID: PMC11801632 DOI: 10.1371/journal.pone.0310908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 09/09/2024] [Indexed: 02/09/2025] Open
Abstract
Nontuberculous Mycobacteria (NTM) are a heterogeneous group of environmental microorganisms with distinct human pathogenesis. Their incidence and prevalence are rising worldwide, due in part to elevated antimicrobial resistance which complicates treatment and potential successful outcomes. Although information exists on the clinical significance of NTMs, little is known about host immune response to infection. NTM infections alter macrophage mitochondrial capacity and decrease ATP production, efficient immune response, and bacterial clearance. Transcription factor peroxisome proliferator activated receptor (PPAR) γ coactivator-1α (PGC-1α) is a master regulator of mitochondrial biogenesis, influencing metabolism, mitochondrial pathways, and antioxidant response. Mitochondrial transcription factor A (TFAM) is a protein essential for mitochondrial DNA (mtDNA) genome stability, integrity, and metabolism. Both PGC-1α and TFAM regulate mitochondrial biogenesis and activity, and their disruption is linked to inflammatory signaling and altered macrophage function. We show that NTM causes macrophage mitochondrial damage and disrupted bioenergetics. Mechanistically we show that this is related to attenuation of expression of PGC-1α and TFAM in infected macrophages. Importantly, rescuing expression of PGC-1α and TFAM using pharmacologic approaches restored macrophage immune function. Our results suggest that pharmacologic approaches to enhance mitochondrial function provide a novel approach to target macrophage immune function and means to combat NTM infections.
Collapse
Affiliation(s)
- Joel R. Frandsen
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Zhihong Yuan
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Brahmchetna Bedi
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Zohra Prasla
- Pulmonology and Critical Care Department, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Seoung-Ryoung Choi
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Prabagaran Narayanasamy
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ruxana T. Sadikot
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
11
|
Harikishore A, Grüber G. Mycobacterium tuberculosis F-ATP Synthase Inhibitors and Targets. Antibiotics (Basel) 2024; 13:1169. [PMID: 39766559 PMCID: PMC11672644 DOI: 10.3390/antibiotics13121169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/23/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Mycobacteria tuberculosis (Mtb) infection causes tuberculosis (TB). TB is one of the most intractable infectious diseases, causing over 1.13 million deaths annually. Under harsh growing conditions, the innate response of mycobacteria is to shut down its respiratory metabolism to a basal level, transit into a dormant, non-replicating phase to preserve viability, and establish latent infection. Mtb utilizes non-canonical regulatory mechanisms, such as alternative oxidase pathways, to survive in low oxygen/nutrient conditions. The bacterium's survival in its native microenvironmental niches is aided by its ability to evolve mutations to drug binding sites, enhance overexpression of various enzymes that activate β-lactam antibiotics hydrolysis, or stimulate efflux pathways to ward off the effect of antibiotics. Bedaquiline and its 3,5-dialkoxypyridine analogs, sudapyridine and squaramide S31f, have been shown to be potent Mtb F1FO-ATP synthase inhibitors of replicating and non-replicating Mtb and have brought oxidative phosphorylation into focus as an anti-TB target. In this review, we attempt to highlight non-canonical structural and regulatory pathogen-specific epitopes of the F1-domain, ligand development on such sites, structural classes of inhibitors targeting the Fo-domain, and alternative respiratory metabolic responses that Mtb employs in response to bedaquiline to ensure its survival and establish latent infection.
Collapse
Affiliation(s)
- Amaravadhi Harikishore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Gerhard Grüber
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| |
Collapse
|
12
|
Kavanagh ME, McLean KJ, Gilbert SH, Amadi C, Snee M, Tunnicliffe RB, Arora K, Boshoff HI, Fanourakis A, Rebello-Lopez MJ, Ortega-Muro F, Levy CW, Munro AW, Leys D, Abell C, Coyne AG. Fragment-based development of small molecule inhibitors targeting Mycobacterium tuberculosis cholesterol metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620643. [PMID: 39803573 PMCID: PMC11722527 DOI: 10.1101/2024.10.28.620643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Mycobacterium tuberculosis (Mtb) is the world's most deadly infectious pathogen and new drugs are urgently required to combat the emergence of multi- (MDR) and extensively- (XDR) drug resistant strains. The bacterium specifically upregulates sterol uptake pathways in infected macrophages and the metabolism of host-derived cholesterol is essential for Mtb's long-term survival in vivo. Here, we report the development of antitubercular small molecules that inhibit the Mtb cholesterol oxidases CYP125 and CYP142, which catalyze the initial step of cholesterol metabolism. An efficient biophysical fragment screen was used to characterize the structure-activity relationships of CYP125 and CYP142, and identify a non-azole small molecule 1a that can bind to the heme cofactor of both enzymes. A structure-guided fragment-linking strategy was used to optimize the binding affinity of 1a, yielding a potent dual CYP125/142 inhibitor 5m (KD CYP125/CYP142 = 0.04/0.16 μM). Compound 5m potently inhibits the catalytic activity of CYP125 and CYP142 in vitro (KI values < 0.1 μM), and rapidly depletes Mtb intracellular ATP (IC50 = 0.15 μM). The compound has antimicrobial activity against both drug susceptible and MDR Mtb (MIC99 values 0.4 - 1.5 μM) in extracellular assays, and inhibits the growth of Mtb in human macrophages (MIC = 1.7 μM) with good selectivity over mammalian cytotoxicity (LD50 ≥ 50 μM). The combination of small molecule inhibitors and structural data reported here provide useful tools to study the role of cholesterol metabolism in Mtb and are a promising step towards novel antibiotics targeting bioenergetic pathways, which could be used to help combat MDR-TB.
Collapse
Affiliation(s)
- Madeline E. Kavanagh
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Kirsty J. McLean
- Centre for Synthetic Biology of Fine and Specialty Chemicals (SYNBIOCHEM), Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Sophie H. Gilbert
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Cecilia Amadi
- Centre for Synthetic Biology of Fine and Specialty Chemicals (SYNBIOCHEM), Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Matthew Snee
- Centre for Synthetic Biology of Fine and Specialty Chemicals (SYNBIOCHEM), Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Richard B. Tunnicliffe
- Centre for Synthetic Biology of Fine and Specialty Chemicals (SYNBIOCHEM), Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Kriti Arora
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - Helena I. Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexander Fanourakis
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | | | | | - Colin W. Levy
- Manchester Protein Structure Facility (MPSF), Manchester Institute of Biotechnology, University of Manchester, Manchester, M1 7DN, UK
| | - Andrew W. Munro
- Centre for Synthetic Biology of Fine and Specialty Chemicals (SYNBIOCHEM), Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - David Leys
- Department of Chemistry, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Chris Abell
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Anthony G. Coyne
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| |
Collapse
|
13
|
Rahman F. Characterizing the immune response to Mycobacterium tuberculosis: a comprehensive narrative review and implications in disease relapse. Front Immunol 2024; 15:1437901. [PMID: 39650648 PMCID: PMC11620876 DOI: 10.3389/fimmu.2024.1437901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/07/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Tuberculosis remains the leading cause of death from infectious diseases among adults worldwide. To date, an overarching review of the immune response to Mtb in humans has not been fully elucidated, with innate immunity remaining poorly understood due to historic focus on adaptive immunity. Specifically, there is a major gap concerning the contribution of the immune system to overall bacterial clearance, particularly residual bacteria. This review aims to describe the time course of interactions between the host immune system and Mtb, from the start of the infection to the development of the adaptive response. Concordantly, we aim to crystallize the pathogenic effects and immunoevasive mechanisms of Mtb. The translational value of animal data is also discussed. Methods The literature search was conducted in the PubMed, ScienceDirect, and Google Scholar databases, which included reported research from 1990 until 2024. A total of 190 publications were selected and screened, of which 108 were used for abstraction and 86 were used for data extraction. Graphical summaries were created using the narrative information (i.e., recruitment, recognition, and response) to generate clear visual representations of the immune response at the cellular and molecular levels. Results The key cellular players included airway epithelial cells, alveolar epithelial cells, neutrophils, natural killer cells, macrophages, dendritic cells, T cells, and granulomatous lesions; the prominent molecular players included IFN-γ, TNF-α, and IL-10. The paper also sheds light on the immune response to residual bacteria and applications of the data. Discussion We provide a comprehensive characterization of the key immune players that are implicated in pulmonary tuberculosis, in line with the organs or compartments in which mycobacteria reside, offering a broad vignette of the immune response to Mtb and how it responds to residual bacteria. Ultimately, the data presented could provide immunological insights to help establish optimized criteria for identifying efficacious treatment regimens and durations for relapse prevention in the modeling and simulation space and wider fields.
Collapse
Affiliation(s)
- Fatima Rahman
- Department of Pharmacology, University College London, London, United Kingdom
- Istituto per le Applicazioni del Calcolo, Consiglio Nazionale delle Ricerche, Rome, Italy
| |
Collapse
|
14
|
Yang S, Tong L, Li X, Zhang Y, Chen H, Zhang W, Zhang H, Chen Y, Chen R. A novel clinically relevant human fecal microbial transplantation model in humanized mice. Microbiol Spectr 2024; 12:e0043624. [PMID: 39162553 PMCID: PMC11448399 DOI: 10.1128/spectrum.00436-24] [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: 02/16/2024] [Accepted: 07/12/2024] [Indexed: 08/21/2024] Open
Abstract
The intact immune system of mice exhibits resistance to colonization by exogenous microorganisms, but the gut microbiota profiles of the humanized mice and the patterns of human fecal microbiota colonization remain unexplored. Humanized NCG (huNCG) mice were constructed by injected CD34 +stem cells. 16S rRNA sequencing and fecal microbiota transplantation (FMT) technologies were used to detect the differences in microbiota and selective colonization ability for exogenous community colonization among three mice cohorts (C57BL/6J, NCG, and huNCG). Flow cytometry analysis showed that all huNCG mice had over 25% hCD45 +in peripheral blood. 16S rRNA gene sequence analysis showed that compared with NCG mice, the gut microbiota of huNCG mice were significantly altered. After FMT, the principal coordinates analysis (PCoA) showed that the gut microbial composition of huNCG mice (huNCG-D9) was similar to that of donors. The relative abundance of Firmicutes and Bacteroidetes were significantly increased in huNCG mice compared to NCG mice. Further comparison of ASV sequences revealed that Bacteroides plebeius, Bacteroides finegoldii, Escherichia fergusonii, Escherichia albertii, Klebsiella pneumoniae, and Klebsiella variicola exhibited higher abundance and stability in huNCG mice after FMT. Furthermore, PICRUSt2 analysis showed that huNCG mice had significantly enhanced metabolism and immunity. This study demonstrated that humanized mice are more conducive to colonization within the human gut microbiota, which provides a good method for studying the association between human diseases and microbiota.IMPORTANCEThe gut microbiota and biomarkers of humanized mice are systematically revealed for the first time. The finding that human fecal microbiota colonize humanized mice more stably provides new insights into the study of interactions between immune responses and gut microbiota.
Collapse
Affiliation(s)
- Shuai Yang
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Linglin Tong
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Li
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuchen Zhang
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hao Chen
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wei Zhang
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - He Zhang
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Chen
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renjin Chen
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
15
|
Nitschke J, Huber R, Vossio S, Moreau D, Marcourt L, Gindro K, Queiroz EF, Soldati T, Hanna N. Discovery of anti-infective compounds against Mycobacterium marinum after biotransformation of simple natural stilbenes by a fungal secretome. Front Microbiol 2024; 15:1439814. [PMID: 39355425 PMCID: PMC11443511 DOI: 10.3389/fmicb.2024.1439814] [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: 05/28/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024] Open
Abstract
Introduction Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, remains a serious threat to human health worldwide and the quest for new anti-tubercular drugs is an enduring and demanding journey. Natural products (NPs) have played a significant role in advancing drug therapy of infectious diseases. Methods This study evaluated the suitability of a high-throughput infection system composed of the host amoeba Dictyostelium discoideum (Dd) and Mycobacterium marinum (Mm), a close relative of Mtb, to identify anti-infective compounds. Growth of Dd and intracellular Mm were quantified by using luminescence and fluorescence readouts in phenotypic assays. The system was first benchmarked with a set of therapeutic anti-Mtb antibiotics and then used to screen a library of biotransformed stilbenes. Results The study confirmed both efficacy of established antibiotics such as rifampicin and bedaquiline, with activities below defined anti-mycobacterium susceptibility breakpoints, and the lack of activity of pyrazinamide against Mm. The screening revealed the promising anti-infective activities of trans-δ-viniferins and in particular of two compounds 17 and 19 with an IC50 of 18.1 μM, 9 μM, respectively. Both compounds had no activity on Mm in broth. Subsequent exploration via halogenation and structure-activity relationship studies led to the identification of derivatives with improved selectivity and potency. The modes of action of the anti-infective compounds may involve inhibition of mycobacterial virulence factors or boosting of host defense. Discussion The study highlights the potential of biotransformation and NP-inspired derivatization approaches for drug discovery and underscores the utility of the Dd-Mm infection system in identifying novel anti-infective compounds.
Collapse
Affiliation(s)
- Jahn Nitschke
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Robin Huber
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, Geneva, Switzerland
| | - Stefania Vossio
- ACCESS Screening Platform, NCCR Chemical Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Dimitri Moreau
- ACCESS Screening Platform, NCCR Chemical Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Laurence Marcourt
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, Geneva, Switzerland
| | - Katia Gindro
- Mycology Group, Research Department Plant Protection, Agroscope, Nyon, Switzerland
| | - Emerson F. Queiroz
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, Geneva, Switzerland
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Nabil Hanna
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| |
Collapse
|
16
|
Putera I, Schrijver B, Kolijn PM, van Stigt AC, Ten Berge JCEM, IJspeert H, Nagtzaam NMA, Swagemakers SMA, van Laar JAM, Agrawal R, Rombach SM, van Hagen PM, La Distia Nora R, Dik WA. A serum B-lymphocyte activation signature is a key distinguishing feature of the immune response in sarcoidosis compared to tuberculosis. Commun Biol 2024; 7:1114. [PMID: 39256610 PMCID: PMC11387424 DOI: 10.1038/s42003-024-06822-1] [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: 04/06/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
Sarcoidosis and tuberculosis (TB) are two granulomatous diseases that often share overlapping clinical features, including uveitis. We measured 368 inflammation-related proteins in serum in both diseases, with and without uveitis from two distinct geographically separated cohorts: sarcoidosis from the Netherlands and TB from Indonesia. A total of 192 and 102 differentially expressed proteins were found in sarcoidosis and active pulmonary TB compared to their geographical healthy controls, respectively. While substantial overlap exists in the immune-related pathways involved in both diseases, activation of B cell activating factor (BAFF) signaling and proliferation-inducing ligand (APRIL) mediated signaling pathways was specifically associated with sarcoidosis. We identified a B-lymphocyte activation signature consisting of BAFF, TNFRSF13B/TACI, TRAF2, IKBKG, MAPK9, NFATC1, and DAPP1 that was associated with sarcoidosis, regardless of the presence of uveitis. In summary, a difference in B-lymphocyte activation is a key discriminative immunological feature between sarcoidosis/ocular sarcoidosis (OS) and TB/ocular TB (OTB).
Collapse
Affiliation(s)
- Ikhwanuliman Putera
- Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine Section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Benjamin Schrijver
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - P Martijn Kolijn
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Astrid C van Stigt
- Department of Internal Medicine Section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Academic Center for Rare Immunological Diseases (Rare Immunological Disease Center), Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Hanna IJspeert
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Academic Center for Rare Immunological Diseases (Rare Immunological Disease Center), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Nicole M A Nagtzaam
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sigrid M A Swagemakers
- Department of Bioinformatics, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jan A M van Laar
- Department of Internal Medicine Section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rupesh Agrawal
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke NUS University, Singapore, Singapore
- Singapore Eye Research Institute, Singapore, Singapore
- Moorfields Eye Hospital, London, United Kingdom
| | - Saskia M Rombach
- Department of Internal Medicine Section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - P Martin van Hagen
- Department of Internal Medicine Section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rina La Distia Nora
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Willem A Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
17
|
Wei L, Liu L, Meng Z, Qi K, Gao X, Feng J, Luo J. Recognition of Mycobacterium tuberculosis by macrophage Toll-like receptor and its role in autophagy. Inflamm Res 2024; 73:753-770. [PMID: 38563966 DOI: 10.1007/s00011-024-01864-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: 08/18/2023] [Revised: 12/25/2023] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND The pathogen responsible for tuberculosis is called Mycobacterium tuberculosis. Its interaction with macrophages has a significant impact on the onset and progression of the disease. METHODS The respiratory pathway allows Mycobacterium tuberculosis to enter the body's lungs where it battles immune cells before being infected latently or actively. In the progress of tuberculosis, Mycobacterium tuberculosis activates the body's immune system and creates inflammatory factors, which cause tissue inflammation to infiltrate and the creation of granulomas, which seriously harms the body. Toll-like receptors of macrophage can mediate host recognition of Mycobacterium tuberculosis, initiate immune responses, and participate in macrophage autophagy. New host-directed therapeutic approaches targeting autophagy for drug-resistant Mycobacterium tuberculosis have emerged, providing new ideas for the effective treatment of tuberculosis. CONCLUSIONS In-depth understanding of the mechanisms by which macrophage autophagy interacts with intracellular Mycobacterium tuberculosis, as well as the study of potent and specific autophagy-regulating molecules, will lead to much-needed advances in drug discovery and vaccine design, which will improve the prevention and treatment of human tuberculosis.
Collapse
Affiliation(s)
- Linna Wei
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, China
| | - Liping Liu
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, China
| | - Zudi Meng
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, China
| | - Kai Qi
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, China
| | - Xuehan Gao
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, China
| | - Jihong Feng
- Department of Oncology, Lishui People's Hospital, Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China
| | - Junmin Luo
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
18
|
Maure A, Lawarée E, Fiorentino F, Pawlik A, Gona S, Giraud-Gatineau A, Eldridge MJG, Danckaert A, Hardy D, Frigui W, Keck C, Gutierrez C, Neyrolles O, Aulner N, Mai A, Hamon M, Barreiro LB, Brodin P, Brosch R, Rotili D, Tailleux L. A host-directed oxadiazole compound potentiates antituberculosis treatment via zinc poisoning in human macrophages and in a mouse model of infection. PLoS Biol 2024; 22:e3002259. [PMID: 38683873 PMCID: PMC11081512 DOI: 10.1371/journal.pbio.3002259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 05/09/2024] [Accepted: 03/13/2024] [Indexed: 05/02/2024] Open
Abstract
Antituberculosis drugs, mostly developed over 60 years ago, combined with a poorly effective vaccine, have failed to eradicate tuberculosis. More worryingly, multiresistant strains of Mycobacterium tuberculosis (MTB) are constantly emerging. Innovative strategies are thus urgently needed to improve tuberculosis treatment. Recently, host-directed therapy has emerged as a promising strategy to be used in adjunct with existing or future antibiotics, by improving innate immunity or limiting immunopathology. Here, using high-content imaging, we identified novel 1,2,4-oxadiazole-based compounds, which allow human macrophages to control MTB replication. Genome-wide gene expression analysis revealed that these molecules induced zinc remobilization inside cells, resulting in bacterial zinc intoxication. More importantly, we also demonstrated that, upon treatment with these novel compounds, MTB became even more sensitive to antituberculosis drugs, in vitro and in vivo, in a mouse model of tuberculosis. Manipulation of heavy metal homeostasis holds thus great promise to be exploited to develop host-directed therapeutic interventions.
Collapse
Affiliation(s)
- Alexandra Maure
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Emeline Lawarée
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Francesco Fiorentino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Alexandre Pawlik
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Saideep Gona
- Department of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | | | | | - Anne Danckaert
- Institut Pasteur, Université Paris Cité, UTechS BioImaging-C2RT, Paris, France
| | - David Hardy
- Institut Pasteur, Université Paris Cité, Histopathology Platform, Paris, France
| | - Wafa Frigui
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Camille Keck
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Claude Gutierrez
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Nathalie Aulner
- Institut Pasteur, Université Paris Cité, UTechS BioImaging-C2RT, Paris, France
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci-bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| | - Mélanie Hamon
- Institut Pasteur, Université Paris Cité, Chromatine et Infection unit, Paris, France
| | - Luis B. Barreiro
- Department of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Priscille Brodin
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Roland Brosch
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Ludovic Tailleux
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| |
Collapse
|
19
|
Wu Y, Gong X, Shen J, Zhu K. Postantibiotic leukocyte enhancement-mediated reduction of intracellular bacteria by macrophages. J Adv Res 2024; 58:117-128. [PMID: 37290606 PMCID: PMC10982861 DOI: 10.1016/j.jare.2023.05.010] [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: 10/28/2022] [Revised: 05/22/2023] [Accepted: 05/28/2023] [Indexed: 06/10/2023] Open
Abstract
INTRODUCTION Potentiation of the bactericidal activities of leukocytes, including macrophages, upon antibacterial agent administration has been observed for several decades and is summarized as the postantibiotic leukocyte enhancement (PALE) theory. Antibiotics-induced bacterial sensitization to leukocytes is commonly recognized as the mechanism of PALE. However, the degree of sensitization drastically varies with antibiotic classes, and little is known about whether and how the potentiation of leukocytes contributes to PALE. OBJECTIVES In this study, we aim to develop a mechanistic understanding of PALE by investigating the immunoregulation of traditional antibiotics on macrophages. METHODS Interaction models between bacteria and macrophages were constructed to identify the effects of different antibiotics on the bactericidal activities of macrophages. Oxygen consumption rate, expression of oxidases, and antioxidants were then measured to evaluate the effects of fluoroquinolones (FQs) on the oxidative stress of macrophages. Furthermore, the modulation in endoplasmic reticulum stress and inflammation upon antibiotic treatment was detected to analyze the mechanisms. At last, the peritoneal infection model was utilized to verify the PALE in vivo. RESULTS Enrofloxacin significantly reduced the intracellular burden of diverse bacterial pathogens through promoting the accumulation of reactive oxygen species (ROS). The upregulated oxidative response accordingly reprograms the electron transport chain with decreased production of antioxidant enzymes to reduce internalized pathogens. Additionally, enrofloxacin modulated the expression and spatiotemporal localization of myeloperoxidase (MPO) to facilitate ROS accumulation to target invaded bacteria and downregulated inflammatory response to alleviate cellular injury. CONCLUSION Our findings demonstrate the crucial role of leukocytes in PALE, shedding light on the development of new host-directed antibacterial therapies and the design of rational dosage regimens.
Collapse
Affiliation(s)
- Yifan Wu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiaoxia Gong
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
20
|
Kumar M, Sharma S, Kumar J, Barik S, Mazumder S. Mitochondrial electron transport chain in macrophage reprogramming: Potential role in antibacterial immune response. CURRENT RESEARCH IN IMMUNOLOGY 2024; 5:100077. [PMID: 38572399 PMCID: PMC10987323 DOI: 10.1016/j.crimmu.2024.100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024] Open
Abstract
Macrophages restrain microbial infection and reinstate tissue homeostasis. The mitochondria govern macrophage metabolism and serve as pivot in innate immunity, thus acting as immunometabolic regulon. Metabolic pathways produce electron flows that end up in mitochondrial electron transport chain (mtETC), made of super-complexes regulating multitude of molecular and biochemical processes. Cell-intrinsic and extrinsic factors influence mtETC structure and function, impacting several aspects of macrophage immunity. These factors provide the macrophages with alternate fuel sources and metabolites, critical to gain functional competence and overcoming pathogenic stress. Mitochondrial reactive oxygen species (mtROS) and oxidative phosphorylation (OXPHOS) generated through the mtETC are important innate immune attributes, which help macrophages in mounting antibacterial responses. Recent studies have demonstrated the role of mtETC in governing mitochondrial dynamics and macrophage polarization (M1/M2). M1 macrophages are important for containing bacterial pathogens and M2 macrophages promote tissue repair and wound healing. Thus, mitochondrial bioenergetics and metabolism are intimately coupled with innate immunity. In this review, we have addressed mtETC function as innate rheostats that regulate macrophage reprogramming and innate immune responses. Advancement in this field encourages further exploration and provides potential novel macrophage-based therapeutic targets to control unsolicited inflammation.
Collapse
Affiliation(s)
- Manmohan Kumar
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Shagun Sharma
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Department of Zoology, Gargi College, University of Delhi, Delhi, India
| | - Jai Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Sailen Barik
- EonBio, 3780 Pelham Drive, Mobile, AL 36619, USA
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Faculty of Life Sciences and Biotechnology, South Asian University, Delhi, India
| |
Collapse
|
21
|
Shan L, Wang Z, Wu L, Qian K, Peng G, Wei M, Tang B, Jun X. Statistical and network analyses reveal mechanisms for the enhancement of macrophage immunity by manganese in Mycobacterium tuberculosis infection. Biochem Biophys Rep 2024; 37:101602. [PMID: 38155943 PMCID: PMC10753046 DOI: 10.1016/j.bbrep.2023.101602] [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: 08/12/2023] [Revised: 11/01/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023] Open
Abstract
Tuberculosis is a significant infectious disease that poses a serious risk to human health. Our previous research has indicated that manganese ions reduce the bacterial load of Mycobacterium tuberculosis in macrophages, but the exact immune defense mechanism remains unknown. Several critical proteins and pathways involved in the host's immune response during this process are still unidentified. Our research aims to identify these proteins and pathways and provide a rationale for the use of manganese ions in the adjuvant treatment of tuberculosis. We downloaded GSE211666 data from the GEO database and selected the RM (Post-infection manganese ion treatment group) and Ra (single-infection group) groups for comparison and analysis to identify differential genes. These differential genes were then enriched and analyzed using STRING, Cytoscape, and NDEx tools to identify the two most relevant pathways of the "Host Response Signature Network." After conducting an in-depth analysis of these two pathways, we found that manganese ions mainly mediate (1) the interferon -gamma (IFN-γ) and its receptor IFNGR and the downstream JAK-STAT pathway and (2) the NFκB pathway to enhance macrophage response to interferon, autophagy, polarization, and cytokine release. Using qPCR experiments, we verified the increased expression of CXCL10, MHCII, IFNγ, CSF2, and IL12, all of which are cytokines that play a key role in resistance to Mycobacterium tuberculosis infection, suggesting that macrophages enter a state of pro-inflammatory and activation after the addition of manganese ions, which enhances their immunosuppressive effect against Mycobacterium tuberculosis. We conclude that our study provides evidence of manganese ion's ability to treat tuberculosis adjuvantly.
Collapse
Affiliation(s)
- Lidong Shan
- College of Life Science, Bengbu Medical University, China
| | - Zihai Wang
- College of Life Science, Bengbu Medical University, China
| | - Lingshan Wu
- College of Life Science, Bengbu Medical University, China
| | - Kaiqiang Qian
- College of Life Science, Bengbu Medical University, China
| | - Guisen Peng
- College of Life Science, Bengbu Medical University, China
| | - MeiLi Wei
- College of Life Science, Bengbu Medical University, China
| | - Bikui Tang
- College of Life Science, Bengbu Medical University, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, China
| | - Xi Jun
- College of Life Science, Bengbu Medical University, China
| |
Collapse
|
22
|
Steshin IS, Vasyankin AV, Shirokova EA, Rozhkov AV, Livshits GD, Panteleev SV, Radchenko EV, Ignatov SK, Palyulin VA. Free Energy Barriers for Passive Drug Transport through the Mycobacterium tuberculosis Outer Membrane: A Molecular Dynamics Study. Int J Mol Sci 2024; 25:1006. [PMID: 38256079 PMCID: PMC10815926 DOI: 10.3390/ijms25021006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The emergence of multi-drug-resistant tuberculosis strains poses a significant challenge to modern medicine. The development of new antituberculosis drugs is hindered by the low permeability of many active compounds through the extremely strong bacterial cell wall of mycobacteria. In order to estimate the ability of potential antimycobacterial agents to diffuse through the outer mycolate membrane, the free energy profiles, the corresponding activation barriers, and possible permeability modes of passive transport for a series of known antibiotics, modern antituberculosis drugs, and prospective active drug-like molecules were determined using molecular dynamics simulations with the all-atom force field and potential of mean-force calculations. The membranes of different chemical and conformational compositions, density, thickness, and ionization states were examined. The typical activation barriers for the low-mass molecules penetrating through the most realistic membrane model were 6-13 kcal/mol for isoniazid, pyrazinamide, and etambutol, and 19 and 25 kcal/mol for bedaquilin and rifampicin. The barriers for the ionized molecules are usually in the range of 37-63 kcal/mol. The linear regression models were derived from the obtained data, allowing one to estimate the permeability barriers from simple physicochemical parameters of the diffusing molecules, notably lipophilicity and molecular polarizability.
Collapse
Affiliation(s)
- Ilya S. Steshin
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia; (I.S.S.); (A.V.V.); (E.A.S.); (A.V.R.); (G.D.L.); (S.V.P.); (E.V.R.)
| | - Alexander V. Vasyankin
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia; (I.S.S.); (A.V.V.); (E.A.S.); (A.V.R.); (G.D.L.); (S.V.P.); (E.V.R.)
| | - Ekaterina A. Shirokova
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia; (I.S.S.); (A.V.V.); (E.A.S.); (A.V.R.); (G.D.L.); (S.V.P.); (E.V.R.)
| | - Alexey V. Rozhkov
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia; (I.S.S.); (A.V.V.); (E.A.S.); (A.V.R.); (G.D.L.); (S.V.P.); (E.V.R.)
| | - Grigory D. Livshits
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia; (I.S.S.); (A.V.V.); (E.A.S.); (A.V.R.); (G.D.L.); (S.V.P.); (E.V.R.)
| | - Sergey V. Panteleev
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia; (I.S.S.); (A.V.V.); (E.A.S.); (A.V.R.); (G.D.L.); (S.V.P.); (E.V.R.)
| | - Eugene V. Radchenko
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia; (I.S.S.); (A.V.V.); (E.A.S.); (A.V.R.); (G.D.L.); (S.V.P.); (E.V.R.)
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, Moscow 119991, Russia
| | - Stanislav K. Ignatov
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia; (I.S.S.); (A.V.V.); (E.A.S.); (A.V.R.); (G.D.L.); (S.V.P.); (E.V.R.)
| | - Vladimir A. Palyulin
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia; (I.S.S.); (A.V.V.); (E.A.S.); (A.V.R.); (G.D.L.); (S.V.P.); (E.V.R.)
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, Moscow 119991, Russia
| |
Collapse
|
23
|
Rodríguez-Fernández P, Botella L, Cavet JS, Domínguez J, Gutierrez MG, Suckling CJ, Scott FJ, Tabernero L. MptpB Inhibitor Improves the Action of Antibiotics against Mycobacterium tuberculosis and Nontuberculous Mycobacterium avium Infections. ACS Infect Dis 2024; 10:170-183. [PMID: 38085851 PMCID: PMC10788870 DOI: 10.1021/acsinfecdis.3c00446] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024]
Abstract
Treatment of Mycobacterium tuberculosis and Mycobacterium avium infections requires multiple drugs for long time periods. Mycobacterium protein-tyrosine-phosphatase B (MptpB) is a key M. tuberculosis virulence factor that subverts host antimicrobial activity to promote intracellular survival. Inhibition of MptpB reduces the infection burden in vivo and offers new opportunities to improve current treatments. Here, we demonstrate that M. avium produces an MptpB orthologue and that the MptpB inhibitor C13 reduces the M. avium infection burden in macrophages. Combining C13 with the antibiotics rifampicin or bedaquiline showed an additive effect, reducing intracellular infection of both M. tuberculosis and M. avium by 50%, compared to monotreatment with antibiotics alone. This additive effect was not observed with pretomanid. Combining C13 with the minor groove-binding compounds S-MGB-362 and S-MGB-363 also reduced the M. tuberculosis intracellular burden. Similar additive effects of C13 and antibiotics were confirmed in vivo using Galleria mellonella infections. We demonstrate that the reduced mycobacterial burden in macrophages observed with C13 treatments is due to the increased trafficking to lysosomes.
Collapse
Affiliation(s)
- Pablo Rodríguez-Fernández
- School
of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health
Science Centre, M13 9PT Manchester, U.K.
| | - Laure Botella
- Host
Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, NW1 1AT London, U.K.
| | - Jennifer S. Cavet
- School
of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health
Science Centre, M13 9PT Manchester, U.K.
- Lydia
Becker Institute for Immunology and Inflammation, University of Manchester, M13 9PT Manchester, U.K.
| | - Jose Domínguez
- Institut
d’Investigació Germans Trias i Pujol, CIBER Enfermedades
Respiratorias (CIBERES), Universitat Autònoma
de Barcelona, 08916 Barcelona, Spain
| | - Maximiliano G. Gutierrez
- Host
Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, NW1 1AT London, U.K.
| | - Colin J. Suckling
- Department
of Pure and Applied Chemistry, University
of Strathclyde, 295 Cathedral Street, G1 1XL Glasgow, U.K.
| | - Fraser J. Scott
- Department
of Pure and Applied Chemistry, University
of Strathclyde, 295 Cathedral Street, G1 1XL Glasgow, U.K.
| | - Lydia Tabernero
- School
of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health
Science Centre, M13 9PT Manchester, U.K.
- Lydia
Becker Institute for Immunology and Inflammation, University of Manchester, M13 9PT Manchester, U.K.
| |
Collapse
|
24
|
Kiselinova M, Naesens L, Huis In ’t Veld D, Boelens J, Van Braeckel E, Vande Weygaerde Y, Callens S. Management Challenges of Extrapulmonary Nontuberculous Mycobacterial Infection: A Single-Center Case Series and Literature Review. Pathogens 2023; 13:12. [PMID: 38276158 PMCID: PMC10819148 DOI: 10.3390/pathogens13010012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024] Open
Abstract
Extrapulmonary nontuberculous mycobacterial (NTM) disease remains largely enigmatic, yet these mycobacteria are increasingly acknowledged as important opportunistic pathogens in humans. Traditionally, NTM infections have been identified across various anatomical locations, with the respiratory system being the most affected and best understood. Historically, extrapulmonary NTM infection was predominantly associated with HIV/AIDS, with Mycobacterium avium lymphadenopathy being the most commonly reported. Today, however, because of the expanding utilization of immunosuppressive therapies and the demographic shift towards an aging population, an increasing number of NTM infections are expected and seen. Hence, a heightened index of suspicion is essential, necessitating a multifaceted approach to identification and drug sensitivity testing to improve treatment outcomes. In extrapulmonary NTM management, expert consultation is strongly recommended to determine the most efficacious treatment regimen, as individualized, patient-tailored therapies are often required. Furthermore, the economic burden of NTM disease is considerable, accompanied by high rates of hospitalization. To optimize the management of these intricate infections, there is an urgent need for comprehensive data on incidence, prevalence, and outcomes. This case-based series delves into the intricate nature of extrapulmonary NTM infections, focusing on both rapid and slow-growing NTM species, and explores therapeutic options, resistance mechanisms, and host-related immunological factors.
Collapse
Affiliation(s)
- Maja Kiselinova
- Department of General Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (L.N.); (S.C.)
| | - Leslie Naesens
- Department of General Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (L.N.); (S.C.)
| | - Diana Huis In ’t Veld
- Department of General Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (L.N.); (S.C.)
| | - Jerina Boelens
- Department of Microbiology, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Eva Van Braeckel
- Department of Respiratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium (Y.V.W.)
- Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | | | - Steven Callens
- Department of General Internal Medicine, Ghent University Hospital, 9000 Ghent, Belgium; (L.N.); (S.C.)
- Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
25
|
Dwivedi R, Baindara P. Differential Regulation of TFEB-Induced Autophagy during Mtb Infection and Starvation. Microorganisms 2023; 11:2944. [PMID: 38138088 PMCID: PMC10746089 DOI: 10.3390/microorganisms11122944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Through the promotion of phagolysosome formation, autophagy has emerged as a crucial mechanism to eradicate intracellular Mycobacterium tuberculosis (Mtb). A cell-autonomous host defense mechanism called lysosome biogenesis and autophagy transports cytoplasmic cargos and bacterial phagosomes to lysosomes for destruction during infection. Similar occurrences occurred in stressful or starvation circumstances and led to autophagy, which is harmful to the cell. It is interesting to note that under both hunger and infection states, the transcription factor EB (TFEB) acts as a master regulator of lysosomal activities and autophagy. This review highlighted recent research on the multitier regulation of TFEB-induced autophagy by a variety of host effectors and Mtb sulfolipid during Mtb infection and starvation. In general, the research presented here sheds light on how lysosome biogenesis and autophagy are differentially regulated by the TFEB during Mtb infection and starvation.
Collapse
Affiliation(s)
- Richa Dwivedi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Piyush Baindara
- Radiation Oncology, NextGen Precision Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
26
|
Abstract
Autophagy is the initial defense response of the host against pathogens. Autophagy can be either non-selective or selective. It selectively targets the degradation of autophagic substrates through the sorting and transportation of autophagic receptor proteins. However, excessive autophagy activity will trigger cell death especially ferroptosis, which was characterized by the accumulation of lipid peroxide and free iron. Several certain types of selective autophagy degrade antioxidant systems and ferritin. Here, we summarized the latest researches of autophagy in infection and discuss the regulatory mechanisms and signaling pathways of autophagy-dependent ferroptosis.
Collapse
Affiliation(s)
- Jiarou Li
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin150086, Heilongjiang Province, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin150086, Heilongjiang Province, China
| | - Hongliang Wang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin150086, Heilongjiang Province, China
| |
Collapse
|
27
|
Kim H, Shin SJ. Revolutionizing control strategies against Mycobacterium tuberculosis infection through selected targeting of lipid metabolism. Cell Mol Life Sci 2023; 80:291. [PMID: 37704889 PMCID: PMC11072447 DOI: 10.1007/s00018-023-04914-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/12/2023] [Accepted: 08/07/2023] [Indexed: 09/15/2023]
Abstract
Lipid species play a critical role in the growth and virulence expression of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). During Mtb infection, foamy macrophages accumulate lipids in granulomas, providing metabolic adaptation and survival strategies for Mtb against multiple stresses. Host-derived lipid species, including triacylglycerol and cholesterol, can also contribute to the development of drug-tolerant Mtb, leading to reduced efficacy of antibiotics targeting the bacterial cell wall or transcription. Transcriptional and metabolic analyses indicate that lipid metabolism-associated factors of Mtb are highly regulated by antibiotics and ultimately affect treatment outcomes. Despite the well-known association between major antibiotics and lipid metabolites in TB treatment, a comprehensive understanding of how altered lipid metabolites in both host and Mtb influence treatment outcomes in a drug-specific manner is necessary to overcome drug tolerance. The current review explores the controversies and correlations between lipids and drug efficacy in various Mtb infection models and proposes novel approaches to enhance the efficacy of anti-TB drugs. Moreover, the review provides insights into the efficacious control of Mtb infection by elucidating the impact of lipids on drug efficacy. This review aims to improve the effectiveness of current anti-TB drugs and facilitate the development of innovative therapeutic strategies against Mtb infection by making reverse use of Mtb-favoring lipid species.
Collapse
Affiliation(s)
- Hagyu Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
28
|
Zheng W, Chang IC, Limberis J, Budzik J, Zha BS, Howard Z, Chen L, Ernst J. Mycobacterium tuberculosis resides in lysosome-poor monocyte-derived lung cells during chronic infection. RESEARCH SQUARE 2023:rs.3.rs-3049913. [PMID: 37398178 PMCID: PMC10312915 DOI: 10.21203/rs.3.rs-3049913/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Mycobacterium tuberculosis (Mtb) persists in lung myeloid cells during chronic infection. However, the mechanisms allowing Mtb to evade elimination are not fully understood. Here, we determined that in chronic phase, CD11clo monocyte-derived lung cells termed MNC1 (mononuclear cell subset 1), harbor more live Mtb than alveolar macrophages (AM), neutrophils, and less permissive CD11chi MNC2. Transcriptomic and functional studies of sorted cells revealed that the lysosome biogenesis pathway is underexpressed in MNC1, which have less lysosome content, acidification, and proteolytic activity than AM, and less nuclear TFEB, a master regulator of lysosome biogenesis. Mtb infection does not drive lysosome deficiency in MNC1. Instead, Mtb recruits MNC1 and MNC2 to the lungs for its spread from AM to these cells via its ESX-1 secretion system. The c-Abl tyrosine kinase inhibitor nilotinib activates TFEB and enhances lysosome function of primary macrophages and MNC1 and MNC2 in vivo, improving control of Mtb infection. Our results indicate that Mtb exploits lysosome-poor monocyte-derived cells for in vivo persistence, suggesting a potential target for host-directed tuberculosis therapy.
Collapse
|
29
|
Kamal A, Arnold C, Claringbould A, Moussa R, Servaas NH, Kholmatov M, Daga N, Nogina D, Mueller‐Dott S, Reyes‐Palomares A, Palla G, Sigalova O, Bunina D, Pabst C, Zaugg JB. GRaNIE and GRaNPA: inference and evaluation of enhancer-mediated gene regulatory networks. Mol Syst Biol 2023; 19:e11627. [PMID: 37073532 PMCID: PMC10258561 DOI: 10.15252/msb.202311627] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 04/20/2023] Open
Abstract
Enhancers play a vital role in gene regulation and are critical in mediating the impact of noncoding genetic variants associated with complex traits. Enhancer activity is a cell-type-specific process regulated by transcription factors (TFs), epigenetic mechanisms and genetic variants. Despite the strong mechanistic link between TFs and enhancers, we currently lack a framework for jointly analysing them in cell-type-specific gene regulatory networks (GRN). Equally important, we lack an unbiased way of assessing the biological significance of inferred GRNs since no complete ground truth exists. To address these gaps, we present GRaNIE (Gene Regulatory Network Inference including Enhancers) and GRaNPA (Gene Regulatory Network Performance Analysis). GRaNIE (https://git.embl.de/grp-zaugg/GRaNIE) builds enhancer-mediated GRNs based on covariation of chromatin accessibility and RNA-seq across samples (e.g. individuals), while GRaNPA (https://git.embl.de/grp-zaugg/GRaNPA) assesses the performance of GRNs for predicting cell-type-specific differential expression. We demonstrate their power by investigating gene regulatory mechanisms underlying the response of macrophages to infection, cancer and common genetic traits including autoimmune diseases. Finally, our methods identify the TF PURA as a putative regulator of pro-inflammatory macrophage polarisation.
Collapse
Affiliation(s)
- Aryan Kamal
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
- Faculty of BiosciencesCollaboration for Joint PhD Degree between EMBL and Heidelberg UniversityHeidelbergGermany
| | - Christian Arnold
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
| | - Annique Claringbould
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
| | - Rim Moussa
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
| | - Nila H Servaas
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
| | - Maksim Kholmatov
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
| | - Neha Daga
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
| | - Daria Nogina
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
| | - Sophia Mueller‐Dott
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
| | - Armando Reyes‐Palomares
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
- Present address:
Department of Biochemistry and Molecular BiologyComplutense University of MadridMadridSpain
| | - Giovanni Palla
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
- Present address:
Institute of Computational BiologyHelmholtz Center MunichOberschleißheimGermany
| | - Olga Sigalova
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
- Faculty of BiosciencesCollaboration for Joint PhD Degree between EMBL and Heidelberg UniversityHeidelbergGermany
| | - Daria Bunina
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
| | - Caroline Pabst
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Molecular Medicine Partnership UnitUniversity of HeidelbergHeidelbergGermany
| | - Judith B Zaugg
- European Molecular Biology Laboratory, Structural and Computational Biology UnitHeidelbergGermany
- Molecular Medicine Partnership UnitUniversity of HeidelbergHeidelbergGermany
| |
Collapse
|
30
|
Malwal SR, Mazurek B, Ko J, Xie P, Barnes C, Varvitsiotis C, Zimmerman MD, Olatunji S, Lee J, Xie M, Sarathy J, Caffrey M, Strynadka NCJ, Dartois V, Dick T, Rin Lee BN, Russell DG, Oldfield E. Investigation into the Mechanism of Action of the Tuberculosis Drug Candidate SQ109 and Its Metabolites and Analogues in Mycobacteria. J Med Chem 2023; 66:7553-7569. [PMID: 37235809 PMCID: PMC10330530 DOI: 10.1021/acs.jmedchem.3c00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
We tested a series of SQ109 analogues against Mycobacterium tuberculosis and M. smegmatis, in addition to determining their uncoupling activity. We then investigated potential protein targets, involved in quinone and cell wall biosynthesis, using "rescue" experiments. There was little effect of menaquinone on growth inhibition by SQ109, but there were large increases in the IC50 of SQ109 and its analogues (up to 20×) on addition of undecaprenyl phosphate (Up), a homologue of the mycobacterial decaprenyl (C50) diphosphate. Inhibition of an undecaprenyl diphosphate phosphatase, an ortholog of the mycobacterial phosphatase, correlated with cell growth inhibition, and we found that M. smegmatis cell growth inhibition could be well predicted by using uncoupler and Up-rescue results. We also investigated whether SQ109 was metabolized inside Mycobacterium tuberculosis, finding only a single metabolite, previously shown to be inactive. The results are of general interest since they help explain the mechanism of SQ109 in mycobacteria.
Collapse
Affiliation(s)
- Satish R. Malwal
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Ben Mazurek
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Jihee Ko
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Pujun Xie
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Chikako Barnes
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Christine Varvitsiotis
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Matthew D. Zimmerman
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, New Jersey 07110, United States
| | - Samir Olatunji
- Schools of Medicine and Biochemistry & Immunology, Trinity College Dublin, D02 R590, Ireland
| | - Jaeyong Lee
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Min Xie
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, New Jersey 07110, United States
| | - Jansy Sarathy
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, New Jersey 07110, United States
| | - Martin Caffrey
- Schools of Medicine and Biochemistry & Immunology, Trinity College Dublin, D02 R590, Ireland
| | - Natalie C. J. Strynadka
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Véronique Dartois
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, New Jersey 07110, United States
- Hackensack Meridian School of Medicine, Department of Medical Sciences, Nutley, NJ 07110, United States
| | - Thomas Dick
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, New Jersey 07110, United States
- Hackensack Meridian School of Medicine, Department of Medical Sciences, Nutley, NJ 07110, United States
- Department of Microbiology and Immunology, Georgetown University, Washington, DC 20007, USA
| | - Bom Nae Rin Lee
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853 USA
| | - David G. Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853 USA
| | - Eric Oldfield
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
31
|
Zheng W, Chang IC, Limberis J, Budzik JM, Zha BS, Howard Z, Chen L, Ernst JD. Mycobacterium tuberculosis resides in lysosome-poor monocyte-derived lung cells during chronic infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524758. [PMID: 36711606 PMCID: PMC9882350 DOI: 10.1101/2023.01.19.524758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Mycobacterium tuberculosis (Mtb) infects cells in multiple lung myeloid cell subsets and causes chronic infection despite innate and adaptive immune responses. However, the mechanisms allowing Mtb to evade elimination are not fully understood. Here, using new methods, we determined that after T cell responses have developed, CD11clo monocyte-derived lung cells termed MNC1 (mononuclear cell subset 1), harbor more live Mtb compared to alveolar macrophages (AM), neutrophils, and less permissive CD11chi MNC2. Bulk RNA sequencing of sorted cells revealed that the lysosome biogenesis pathway is underexpressed in MNC1. Functional assays confirmed that Mtb-permissive MNC1 have less lysosome content, acidification, and proteolytic activity than AM, and less nuclear TFEB, a master regulator of lysosome biogenesis. Mtb infection does not drive lysosome deficiency in MNC1 in vivo. Instead, Mtb recruits MNC1 and MNC2 to the lungs for its spread from AM to these cell subsets as a virulence mechanism that requires the Mtb ESX-1 secretion system. The c-Abl tyrosine kinase inhibitor nilotinib activates TFEB and enhances lysosome function of primary macrophages in vitro and MNC1 and MNC2 in vivo, improving control of Mtb infection. Our results indicate that Mtb exploits lysosome-poor monocyte-derived cells for in vivo persistence, suggesting a potential target for host-directed tuberculosis therapy.
Collapse
Affiliation(s)
- Weihao Zheng
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - I-Chang Chang
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Jason Limberis
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Jonathan M. Budzik
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - B. Shoshana Zha
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Zach Howard
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Lucas Chen
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Joel D. Ernst
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
32
|
Lee BS, Singh S, Pethe K. Inhibiting respiration as a novel antibiotic strategy. Curr Opin Microbiol 2023; 74:102327. [PMID: 37235914 DOI: 10.1016/j.mib.2023.102327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023]
Abstract
The approval of the first-in-class antibacterial bedaquiline for tuberculosis marks a breakthrough in antituberculosis drug development. The drug inhibits mycobacterial respiration and represents the validation of a wholly different metabolic process as a druggable target space. In this review, we discuss the advances in the development of mycobacterial respiratory inhibitors, as well as the potential of applying this strategy to other pathogens. The non-fermentative nature of mycobacteria explains their vulnerability to respiration inhibition, and we caution that this strategy may not be equally effective in other organisms. Conversely, we also showcase fundamental studies that reveal ancillary functions of the respiratory pathway, which are crucial to some pathogens' virulence, drug susceptibility and fitness, introducing another perspective of targeting bacterial respiration as an antibiotic strategy.
Collapse
Affiliation(s)
- Bei Shi Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore.
| | - Samsher Singh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| | - Kevin Pethe
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 637551, Singapore; National Centre for Infectious Diseases, Singapore 308442, Singapore.
| |
Collapse
|
33
|
Khan U, Karmakar BC, Basak P, Paul S, Gope A, Sarkar D, Mukhopadhyay AK, Dutta S, Bhattacharya S. Glycyrrhizin, an inhibitor of HMGB1 induces autolysosomal degradation function and inhibits Helicobacter pylori infection. Mol Med 2023; 29:51. [PMID: 37038107 PMCID: PMC10088177 DOI: 10.1186/s10020-023-00641-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/21/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Helicobacter pylori is a key agent for causing gastric complications linked with gastric disorders. In response to infection, host cells stimulate autophagy to maintain cellular homeostasis. However, H. pylori have evolved the ability to usurp the host's autophagic machinery. High mobility group box1 (HMGB1), an alarmin molecule is a regulator of autophagy and its expression is augmented during infection and gastric cancer. Therefore, this study aims to explore the role of glycyrrhizin (a known inhibitor of HMGB1) in autophagy during H. pylori infection. MAIN METHODS Human gastric cancer (AGS) cells were infected with the H. pylori SS1 strain and further treatment was done with glycyrrhizin. Western blot was used to examine the expression of autophagy proteins. Autophagy and lysosomal activity were monitored by fluorescence assays. A knockdown of HMGB1 was performed to verify the effect of glycyrrhizin. H. pylori infection in in vivo mice model was established and the effect of glycyrrhizin treatment was studied. RESULTS The autophagy-lysosomal pathway was impaired due to an increase in lysosomal membrane permeabilization during H. pylori infection in AGS cells. Subsequently, glycyrrhizin treatment restored the lysosomal membrane integrity. The recovered lysosomal function enhanced autolysosome formation and concomitantly attenuated the intracellular H. pylori growth by eliminating the pathogenic niche. Additionally, glycyrrhizin treatment inhibited inflammation and improved gastric tissue damage in mice. CONCLUSION This study showed that inhibiting HMGB1 restored lysosomal activity to ameliorate H. pylori infection. It also demonstrated the potential of glycyrrhizin as an antibacterial agent to address the problem of antimicrobial resistance.
Collapse
Affiliation(s)
- Uzma Khan
- Division of Biochemistry ICMR-NICED, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), Kolkata, 700010, India
| | - Bipul Chandra Karmakar
- Division of Bacteriology ICMR-NICED, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), Kolkata, 700010, India
| | - Priyanka Basak
- Division of Biochemistry ICMR-NICED, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), Kolkata, 700010, India
| | - Sangita Paul
- Division of Bacteriology ICMR-NICED, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), Kolkata, 700010, India
| | - Animesh Gope
- Division of Clinical Medicine, ICMR-NICED, ICMR- National Institute of Cholera and Enteric Diseases (ICMR-NICED), Kolkata, India
| | - Deotima Sarkar
- Division of Biochemistry ICMR-NICED, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), Kolkata, 700010, India
| | - Asish Kumar Mukhopadhyay
- Division of Bacteriology ICMR-NICED, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), Kolkata, 700010, India
| | - Shanta Dutta
- Division of Bacteriology ICMR-NICED, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), Kolkata, 700010, India
| | - Sushmita Bhattacharya
- Division of Biochemistry ICMR-NICED, ICMR-National Institute of Cholera and Enteric Diseases (ICMR-NICED), Kolkata, 700010, India.
| |
Collapse
|
34
|
Setyawan MF, Mertaniasih NM, Soedarsono S, Nuha Z, Maladan Y, Matsumoto S. Mycobacterium tuberculosis - atpE gene profile of bedaquiline-treated pulmonary tuberculosis patients at the referral hospital Dr. Soetomo, Indonesia. Int J Mycobacteriol 2023; 12:122-128. [PMID: 37338471 DOI: 10.4103/ijmy.ijmy_40_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023] Open
Abstract
Background The atpE gene is a target for bedaquiline (Bdq)-activating drug action and mutations in the gene are fixed to cause resistance. However, changes in the amino acid of ATPase have been little reported from a clinical setting since it was first used in 2015 in Indonesia. This study aims to observe the sequence of nucleotide and amino acid from rifampicin-resistant (RR) pulmonary tuberculosis (TB) patients, both new and relapse cases treated with Bdq. Methods This is an observational descriptive study performed in the referral hospital Dr Soetomo, Indonesia, at August 2022-November 2022. We performed Sanger sequencing and comparison of the atpE gene from the patient's sputum from August to November 2022 to wild-type Mycobacterium tuberculosis H37Rv and species of mycobacteria using BioEdit version 7.2 and BLAST NCBI software. We also conducted an epidemiological study on patients' characteristics. This study uses a descriptive statistic to show the percentage of data. Results The total of 12 M. tuberculosis isolates showed that the atpE gene sequence was 100% similar to the wild-type M. tuberculosis H37Rv. No single-nucleotide polymorphisms or mutations were found, and no change in the amino acid structure at position 28 (Asp), 61 (Glu), 63 (Ala), and 66 (Ile). The percentage identity of atpE to M. tuberculosis H37Rv and M. tuberculosis complex was 99%-100%, while the similarity with the other mycobacteria species other than TB (Mycobacterium avium complex, Mycobacterium abscessus, and Mycobacterium lepraemurium) was 88%-91%. Conclusions This study revealed M. tuberculosis -atpE gene sequence profile of RR-TB patients had no mutations, as the specific gene region, and no change in the amino acid structure. Therefore, Bdq can be continually trusted as an effective anti-tubercular drug in RR-TB patients.
Collapse
Affiliation(s)
- Muhamad Frendy Setyawan
- Master Program in Tropical Medicine; Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ni Made Mertaniasih
- Department of Clinical Microbiology, Faculty of Medicine, Airlangga University; Department of Clinical Microbiology, Dr. Soetomo Academic Hospital, Surabaya, Indonesia
| | - S Soedarsono
- Department of Clinical Microbiology, Dr. Soetomo Academic Hospital; Sub-Pulmonology Department of Internal Medicine, Faculty of Medicine, Hang Tuah University, Surabaya, Indonesia
| | - Zakiyathun Nuha
- Laboratory of Tuberculosis, Institute of Tropical Disease, Universitas Airlangga, East Java, Surabaya, Indonesia
| | - Yustinus Maladan
- Eijkman Research Center for Molecular Biology, The National Research and Innovation Agency, Cibinong, Bogor, Indonesia
| | - Sohkichi Matsumoto
- Department of Bacteriology, Niigata University School of Medicine, Niigata, Japan
| |
Collapse
|
35
|
Mackieh R, Al-Bakkar N, Kfoury M, Roufayel R, Sabatier JM, Fajloun Z. Inhibitors of ATP Synthase as New Antibacterial Candidates. Antibiotics (Basel) 2023; 12:antibiotics12040650. [PMID: 37107012 PMCID: PMC10135114 DOI: 10.3390/antibiotics12040650] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
ATP, the power of all cellular functions, is constantly used and produced by cells. The enzyme called ATP synthase is the energy factory in all cells, which produces ATP by adding inorganic phosphate (Pi) to ADP. It is found in the inner, thylakoid and plasma membranes of mitochondria, chloroplasts and bacteria, respectively. Bacterial ATP synthases have been the subject of multiple studies for decades, since they can be genetically manipulated. With the emergence of antibiotic resistance, many combinations of antibiotics with other compounds that enhance the effect of these antibiotics have been proposed as approaches to limit the spread of antibiotic-resistant bacteria. ATP synthase inhibitors, such as resveratrol, venturicidin A, bedaquiline, tomatidine, piceatannol, oligomycin A and N,N-dicyclohexylcarbodiimide were the starting point of these combinations. However, each of these inhibitors target ATP synthase differently, and their co-administration with antibiotics increases the susceptibility of pathogenic bacteria. After a brief description of the structure and function of ATP synthase, we aim in this review to highlight therapeutic applications of the major bacterial ATP synthase inhibitors, including animal’s venoms, and to emphasize their importance in decreasing the activity of this enzyme and subsequently eradicating resistant bacteria as ATP synthase is their source of energy.
Collapse
|
36
|
Im H, Pearson ML, Martinez E, Cichos KH, Song X, Kruckow KL, Andrews RM, Ghanem ES, Orihuela CJ. Targeting NAD+ regeneration enhances antibiotic susceptibility of Streptococcus pneumoniae during invasive disease. PLoS Biol 2023; 21:e3002020. [PMID: 36928033 PMCID: PMC10019625 DOI: 10.1371/journal.pbio.3002020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 02/02/2023] [Indexed: 03/18/2023] Open
Abstract
Anaerobic bacteria are responsible for half of all pulmonary infections. One such pathogen is Streptococcus pneumoniae (Spn), a leading cause of community-acquired pneumonia, bacteremia/sepsis, and meningitis. Using a panel of isogenic mutants deficient in lactate, acetyl-CoA, and ethanol fermentation, as well as pharmacological inhibition, we observed that NAD(H) redox balance during fermentation was vital for Spn energy generation, capsule production, and in vivo fitness. Redox balance disruption in fermentation pathway-specific fashion substantially enhanced susceptibility to killing in antimicrobial class-specific manner. Blocking of alcohol dehydrogenase activity with 4-methylpyrazole (fomepizole), an FDA-approved drug used as an antidote for toxic alcohol ingestion, enhanced susceptibility of multidrug-resistant Spn to erythromycin and reduced bacterial burden in the lungs of mice with pneumonia and prevented the development of invasive disease. Our results indicate fermentation enzymes are de novo targets for antibiotic development and a novel strategy to combat multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Hansol Im
- Department of Microbiology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Madison L. Pearson
- Department of Microbiology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Eriel Martinez
- Department of Microbiology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Kyle H. Cichos
- Department of Orthopaedic Surgery Arthroplasty Section, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Xiuhong Song
- Department of Microbiology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Katherine L. Kruckow
- Department of Microbiology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Rachel M. Andrews
- Department of Microbiology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Elie S. Ghanem
- Department of Orthopaedic Surgery Arthroplasty Section, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Carlos J. Orihuela
- Department of Microbiology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
37
|
Thu VTA, Dat LD, Jayanti RP, Trinh HKT, Hung TM, Cho YS, Long NP, Shin JG. Advancing personalized medicine for tuberculosis through the application of immune profiling. Front Cell Infect Microbiol 2023; 13:1108155. [PMID: 36844400 PMCID: PMC9950414 DOI: 10.3389/fcimb.2023.1108155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/17/2023] [Indexed: 02/12/2023] Open
Abstract
While early and precise diagnosis is the key to eliminating tuberculosis (TB), conventional methods using culture conversion or sputum smear microscopy have failed to meet demand. This is especially true in high-epidemic developing countries and during pandemic-associated social restrictions. Suboptimal biomarkers have restricted the improvement of TB management and eradication strategies. Therefore, the research and development of new affordable and accessible methods are required. Following the emergence of many high-throughput quantification TB studies, immunomics has the advantages of directly targeting responsive immune molecules and significantly simplifying workloads. In particular, immune profiling has been demonstrated to be a versatile tool that potentially unlocks many options for application in TB management. Herein, we review the current approaches for TB control with regard to the potentials and limitations of immunomics. Multiple directions are also proposed to hopefully unleash immunomics' potential in TB research, not least in revealing representative immune biomarkers to correctly diagnose TB. The immune profiles of patients can be valuable covariates for model-informed precision dosing-based treatment monitoring, prediction of outcome, and the optimal dose prediction of anti-TB drugs.
Collapse
Affiliation(s)
- Vo Thuy Anh Thu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Ly Da Dat
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Rannissa Puspita Jayanti
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Hoang Kim Tu Trinh
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh, Ho Chi Minh City, Vietnam
| | - Tran Minh Hung
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Yong-Soon Cho
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea,*Correspondence: Jae-Gook Shin, ; Nguyen Phuoc Long,
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea,Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea,*Correspondence: Jae-Gook Shin, ; Nguyen Phuoc Long,
| |
Collapse
|
38
|
Putra ON, Yulistiani Y, Soedarsono S, Subay S. Favorable outcome of individual regimens containing bedaquiline and delamanid in drug-resistant tuberculosis: A systematic review. Int J Mycobacteriol 2023; 12:1-9. [PMID: 36926755 DOI: 10.4103/ijmy.ijmy_217_22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Background Drug-resistant tuberculosis (DR-TB) is a public health concern that is difficult to treat, requiring long and complex treatment with highly effective drugs. Bedaquiline and/or delamanid have already shown promising outcomes in patients with DR-TB, increasing the rate of culture conversion and lowering TB-related mortality. Methods We comprehensively searched and evaluated the effectiveness of individual regimens containing bedaquiline and delamanid on culture conversion and treatment success. We assessed for quality either observational or experimental studies. Results We identified 14 studies that met the inclusion criteria using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses flowchart with 12 observational and 2 experimental studies. Of 1691 DR-TB patients enrolled in the included studies, 1407 of them concomitantly received regimens containing bedaquiline and delamanid. Overall multidrug resistant (MDR), preextensively drug resistant (XDR), and XDR-TB were seen in 21.4%, 44.1%, and 34.5%, respectively. Of 14 studies, 8 of them reported favorable outcomes including sputum culture conversion and cure rate at the end of treatment, meanwhile 6 studies only reported sputum culture conversion. Sputum culture conversion at the end of the 6th month was 63.6%-94.7% for observational studies, and 87.6%-95.0% for experimental studies. The favorable outcome at the end of treatment was 67.5%-91.4%. With high pre-XDR and XDR cases among DR-TB patients with limited treatment options, regimens containing bedaquiline and delamanid provide successful treatment. Conclusion In DR-TB patients receiving regimens containing bedaquiline and delamanid, favorable outcomes were high including sputum conversion and cure rate.
Collapse
Affiliation(s)
- Oki Nugraha Putra
- Doctoral Program of Pharmacy, Faculty of Pharmacy, Airlangga University; Study Program of Pharmacy, Faculty of Medicine, Hang Tuah University, Surabaya, Indonesia
| | - Yulistiani Yulistiani
- Doctoral Program of Pharmacy, Faculty of Pharmacy, Airlangga University, Surabaya, Indonesia
| | - Soedarsono Soedarsono
- Study Program of Pharmacy, Faculty of Medicine, Hang Tuah University; Department of Pulmonology and Respiratory Medicine, Dr. Soetomo Hospital, Surabaya, Indonesia
| | - Susi Subay
- Department of Pulmonology and Respiratory Medicine, Dr. Soetomo Hospital, Surabaya, Indonesia
| |
Collapse
|
39
|
Larkins-Ford J, Aldridge BB. Advances in the design of combination therapies for the treatment of tuberculosis. Expert Opin Drug Discov 2023; 18:83-97. [PMID: 36538813 PMCID: PMC9892364 DOI: 10.1080/17460441.2023.2157811] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Tuberculosis requires lengthy multi-drug therapy. Mycobacterium tuberculosis occupies different tissue compartments during infection, making drug access and susceptibility patterns variable. Antibiotic combinations are needed to ensure each compartment of infection is reached with effective drug treatment. Despite drug combinations' role in treating tuberculosis, the design of such combinations has been tackled relatively late in the drug development process, limiting the number of drug combinations tested. In recent years, there has been significant progress using in vitro, in vivo, and computational methodologies to interrogate combination drug effects. AREAS COVERED This review discusses the advances in these methodologies and how they may be used in conjunction with new successful clinical trials of novel drug combinations to design optimized combination therapies for tuberculosis. Literature searches for approaches and experimental models used to evaluate drug combination effects were undertaken. EXPERT OPINION We are entering an era richer in combination drug effect and pharmacokinetic/pharmacodynamic data, genetic tools, and outcome measurement types. Application of computational modeling approaches that integrate these data and produce predictive models of clinical outcomes may enable the field to generate novel, effective multidrug therapies using existing and new drug combination backbones.
Collapse
Affiliation(s)
- Jonah Larkins-Ford
- Department of Molecular Biology and Microbiology and Tufts University School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (CIMAR), Tufts University, Boston, MA, USA
- Current address: MarvelBiome Inc, Woburn, MA, USA
| | - Bree B. Aldridge
- Department of Molecular Biology and Microbiology and Tufts University School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (CIMAR), Tufts University, Boston, MA, USA
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA, USA
| |
Collapse
|
40
|
Zhang D, Xu S, Wu H, Liu J, Wang Y, Zhu G. Melatonin Is Neuroprotective in Escherichia coli Meningitis Depending on Intestinal Microbiota. Int J Mol Sci 2022; 24:ijms24010298. [PMID: 36613745 PMCID: PMC9820133 DOI: 10.3390/ijms24010298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Avian meningitis Escherichia coli (E. coli) can cause acute bacterial meningitis which threatens poultry health, causes great economic losses in the poultry industry, and has recently been speculated as a potential zoonotic pathogen. Melatonin can counteract bacterial meningitis-induced disruption of the blood-brain barrier (BBB), neuroinflammation, and reduce mortality. There are increasing data showing that melatonin's beneficial effects on bacterial meningitis are associated with intestinal microbiota. In this study, our data showed that melatonin alleviated neurological symptoms, enhanced survival rate, protected the integrity of the BBB, reduced the bacterial load in various tissues and blood, and inhibited inflammation and neutrophil infiltration of brain tissue in an APEC TW-XM-meningitis mice model. The results of 16S rRNA showed that melatonin pretreatment significantly maintained the composition of intestinal microbiota in APEC-meningitis mice. The abundance and diversity of intestinal microbiota were disturbed in APEC TW-XM-meningitis mice, with a decreased ratio of Firmicutes to Bacteroides and an increased the abundance of Proteobacteria. Melatonin pretreatment could significantly improve the composition and abundance of harmful bacteria and alleviate the decreased abundance of beneficial bacteria. Importantly, melatonin failed to affect the meningitis neurologic symptoms caused by APEC TW-XM infection in antibiotic-pretreated mice. In conclusion, the results suggest that melatonin can effectively prevent meningitis induced by APEC TW-XM infection in mice, depending on the intestinal microbiota. This finding is helpful to further explore the specific target mechanism of melatonin-mediated intestinal microbiota in the prevention of and protection against Escherichia coli meningitis.
Collapse
Affiliation(s)
- Dong Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Shu Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Hucong Wu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jiaqi Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yiting Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
41
|
Cioetto-Mazzabò L, Boldrin F, Beauvineau C, Speth M, Marina A, Namouchi A, Segafreddo G, Cimino M, Favre-Rochex S, Balasingham S, Trastoy B, Munier-Lehmann H, Griffiths G, Gicquel B, Guerin M, Manganelli R, Alonso-Rodríguez N. SigH stress response mediates killing of Mycobacterium tuberculosis by activating nitronaphthofuran prodrugs via induction of Mrx2 expression. Nucleic Acids Res 2022; 51:144-165. [PMID: 36546765 PMCID: PMC9841431 DOI: 10.1093/nar/gkac1173] [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: 10/03/2022] [Revised: 11/17/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
The emergence of drug-resistant Mycobacterium tuberculosis strains highlights the need to discover anti-tuberculosis drugs with novel mechanisms of action. Here we discovered a mycobactericidal strategy based on the prodrug activation of selected chemical derivatives classified as nitronaphthofurans (nNFs) mediated by the coordinated action of the sigH and mrx2 genes. The transcription factor SigH is a key regulator of an extensive transcriptional network that responds to oxidative, nitrosative, and heat stresses in M. tuberculosis. The nNF action induced the SigH stress response which in turn induced the mrx2 overexpression. The nitroreductase Mrx2 was found to activate nNF prodrugs, killing replicating, non-replicating and intracellular forms of M. tuberculosis. Analysis of SigH DNA sequences obtained from spontaneous nNF-resistant M. tuberculosis mutants suggests disruption of SigH binding to the mrx2 promoter site and/or RNA polymerase core, likely promoting the observed loss of transcriptional control over Mrx2. Mutations found in mrx2 lead to structural defects in the thioredoxin fold of the Mrx2 protein, significantly impairing the activity of the Mrx2 enzyme against nNFs. Altogether, our work brings out the SigH/Mrx2 stress response pathway as a promising target for future drug discovery programs.
Collapse
Affiliation(s)
| | | | - Claire Beauvineau
- Chemical Library Institut Curie/CNRS, CNRS UMR9187, INSERM U1196 and CNRS UMR3666, INSERM U1193, Université Paris-Saclay, Orsay 91405, France
| | - Martin Speth
- Department Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo 0371, Norway
| | - Alberto Marina
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio 48160 Spain
| | - Amine Namouchi
- Génétique Mycobactérienne, Institute Pasteur, Paris 75015, France,Centre for Ecological and Evolutionary Synthesis (CEES), Department of Biosciences, University of Oslo, Oslo 0371, Norway
| | - Greta Segafreddo
- Department of Molecular Medicine, University of Padova, Padova 35122, Italy
| | - Mena Cimino
- Génétique Mycobactérienne, Institute Pasteur, Paris 75015, France
| | | | | | - Beatriz Trastoy
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio 48160 Spain,Structural Glycobiology Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Bizkaia 48903, Spain
| | - Hélène Munier-Lehmann
- Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR3523, Université de Paris, Paris 75015, France
| | - Gareth Griffiths
- Department Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo 0371, Norway
| | - Brigitte Gicquel
- Génétique Mycobactérienne, Institute Pasteur, Paris 75015, France,Department of Tuberculosis Control and Prevention, Shenzhen Nanshan Centre for Chronic Disease Control, Shenzhen 518054, China
| | - Marcelo E Guerin
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio 48160 Spain,Structural Glycobiology Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Bizkaia 48903, Spain,IKERBASQUE, Basque Foundation for Science, Bilbao 48009, Spain
| | - Riccardo Manganelli
- Correspondence may also be addressed to Riccardo Manganelli. Tel: +39 049 827 2366; Fax: +39 049 827 2355;
| | | |
Collapse
|
42
|
Qiao M, Li S, Yuan J, Ren W, Shang Y, Wang W, Liu R, Zhang F, Li Q, Wu X, Lu J, Gao M, Pang Y. Delamanid suppresses CXCL10 expression via regulation of JAK/STAT1 signaling and correlates with reduced inflammation in tuberculosis patients. Front Immunol 2022; 13:923492. [DOI: 10.3389/fimmu.2022.923492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
BackgroundApart from bactericidal effects, anti-tuberculosis drugs can interfere with the host’s immune system. In this study, we analyzed the role of delamanid (DLM), an inhibitor of mycolic acid synthesis of mycobacterial cell wall, on human macrophages.MethodsBased on a cohort of multidrug-resistant tuberculosis (MDR-TB) patients treated with DLM, the levels of C-reaction protein (CRP) and cytokines in the plasma were monitored using immunoturbidimetric assay and flow cytometry, respectively. We investigated the role of DLM on CXCL10 expression in U937 cell model using the following methods: cell viability assay, reverse transcription-quantitative polymerase chain reaction, enzyme linked immunosorbent assay, immunoblot, and transwell co-culture assay.ResultsA total of 23 MDR-TB patients were included, comprising of 13 patients treated with optimized background therapeutic regimen (OBR) plus DLM regimen (OBR+DLM) and 10 patients treated with OBR plus placebo. DLM administration was associated with a significant reduce in circulating CRP level. Correspondingly, after treatment, the level of CXCL10 in patients treated with OBR+DLM was significantly lower than that with control. Using cell model, DLM dramatically suppressed CXCL10 expression, which majorly depended on inhibiting the JAK/STAT pathway, and impaired the migration of PBMCs.ConclusionOur data firstly demonstrate that DLM suppresses CXCL10 expression via regulation of JAK2/STAT1 signaling and correlates with reduced inflammation in MDR-TB patients. DLM could be used as a potential drug for immunotherapy of patients with overactive immune response due to CXCL10.
Collapse
|
43
|
Tian J, Chen S, Liu F, Zhu Q, Shen J, Lin W, Zhu K. Equisetin Targets Intracellular Staphylococcus aureus through a Host Acting Strategy. Mar Drugs 2022; 20:656. [PMID: 36354979 PMCID: PMC9694014 DOI: 10.3390/md20110656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/15/2022] [Accepted: 10/16/2022] [Indexed: 09/22/2023] Open
Abstract
Mammalian cells act as reservoirs of internalized bacteria to circumvent extracellular antibacterial compounds, resulting in relapse and reinfection diseases. The intracellular persistence of Staphylococcus aureus renders most traditional antibiotics useless, due to their inadequate subcellular accumulation. To replenish our antibiotic arsenal, we found that a marine-derived compound, equisetin, efficiently eliminates intracellular S. aureus by potentiating the host autophagy and inducing mitochondrial-mediated ROS generation to clear the invading S. aureus. The remarkable anti-infection activity of equisetin was validated in a peritonitis-infected mouse model. The marine product equisetin utilizes a unique dual mechanism to modulate the host-pathogen interaction in the clearance of intracellular bacteria. Thus, equisetin is an inspiring host-acting candidate for overcoming intracellular pathogens.
Collapse
Affiliation(s)
- Jiayao Tian
- National Center for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Shang Chen
- National Center for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Fei Liu
- National Center for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Qian Zhu
- National Center for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Jianzhong Shen
- National Center for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Wenhan Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Kui Zhu
- National Center for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| |
Collapse
|
44
|
Tang H, Liu Y, Ruan Y, Ge L, Zhang Q. Reconstructed Genome-Scale Metabolic Model Characterizes Adaptive Metabolic Flux Changes in Peripheral Blood Mononuclear Cells in Severe COVID-19 Patients. Int J Mol Sci 2022; 23:12400. [PMID: 36293257 PMCID: PMC9604493 DOI: 10.3390/ijms232012400] [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: 08/31/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) poses a mortal threat to human health. The elucidation of the relationship between peripheral immune cells and the development of inflammation is essential for revealing the pathogenic mechanism of COVID-19 and developing related antiviral drugs. The immune cell metabolism-targeting therapies exhibit a desirable anti-inflammatory effect in some treatment cases. In this study, based on differentially expressed gene (DEG) analysis, a genome-scale metabolic model (GSMM) was reconstructed by integrating transcriptome data to characterize the adaptive metabolic changes in peripheral blood mononuclear cells (PBMCs) in severe COVID-19 patients. Differential flux analysis revealed that metabolic changes such as enhanced aerobic glycolysis, impaired oxidative phosphorylation, fluctuating biogenesis of lipids, vitamins (folate and retinol), and nucleotides played important roles in the inflammation adaptation of PBMCs. Moreover, the main metabolic enzymes such as the solute carrier (SLC) family 2 member 3 (SLC2A3) and fatty acid synthase (FASN), responsible for the reactions with large differential fluxes, were identified as potential therapeutic targets. Our results revealed the inflammation regulation potentials of partial metabolic reactions with differential fluxes and their metabolites. This study provides a reference for developing potential PBMC metabolism-targeting therapy strategies against COVID-19.
Collapse
Affiliation(s)
| | | | | | | | - Qingye Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
45
|
Wang H, Bi J, Zhang Y, Pan M, Guo Q, Xiao G, Cui Y, Hu S, Chan CK, Yuan Y, Kaneko T, Zhang G, Chen S. Human Kinase IGF1R/IR Inhibitor Linsitinib Controls the In Vitro and Intracellular Growth of Mycobacterium tuberculosis. ACS Infect Dis 2022; 8:2019-2027. [PMID: 36048501 PMCID: PMC11807261 DOI: 10.1021/acsinfecdis.2c00278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
ATP provides energy in the biosynthesis of cellular metabolites as well as regulates protein functions through phosphorylation. Many ATP-dependent enzymes are antibacterial and anticancer targets including human kinases acted on by most of the successful drugs. In search of new chemotherapeutics for tuberculosis (TB), we screened repurposing compounds against the essential glutamine synthase (GlnA1) of Mycobacterium tuberculosis (Mtb) and identified linsitinib, a clinical-stage drug originally targeting kinase IGF1R/IR as a potent GlnA1 inhibitor. Linsitinib has direct antimycobacterial activity. Biochemical, molecular modeling, and target engagement analyses revealed the inhibition is ATP-competitive and specific in Mtb. Linsitinib also improves autophagy flux in both Mtb-infected and uninfected THP1 macrophages, as demonstrated by the decreased p-mTOR and p62 and the increased lipid-bound LC3B-II and autophagosome forming puncta. Linsitinib-mediated autophagy reduces intracellular growth of wild-type and isoniazid-resistant Mtb alone or in combination with bedaquiline. We have demonstrated that an IGF-IR/IR inhibitor can potentially be used to treat TB. Our study reinforces the concept of targeting ATP-dependent enzymes for novel anti-TB therapy.
Collapse
Affiliation(s)
- Heng Wang
- Global Health Drug Discovery Institute, Haidian, Beijing 100192, China
| | - Jing Bi
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Yuan Zhang
- Global Health Drug Discovery Institute, Haidian, Beijing 100192, China
| | - Miaomiao Pan
- Global Health Drug Discovery Institute, Haidian, Beijing 100192, China
| | - Qinglong Guo
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Genhui Xiao
- Global Health Drug Discovery Institute, Haidian, Beijing 100192, China
| | - Yumeng Cui
- Global Health Drug Discovery Institute, Haidian, Beijing 100192, China
| | - Song Hu
- Global Health Drug Discovery Institute, Haidian, Beijing 100192, China
| | - Chi Kin Chan
- Global Health Drug Discovery Institute, Haidian, Beijing 100192, China
| | - Ying Yuan
- Global Health Drug Discovery Institute, Haidian, Beijing 100192, China
| | - Takushi Kaneko
- Global Alliance for TB Drug Development, New York, New York 10005, United States
| | - Guoliang Zhang
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Shawn Chen
- Global Health Drug Discovery Institute, Haidian, Beijing 100192, China
| |
Collapse
|
46
|
Singh M, Kumar S, Singh B, Jain P, Kumari A, Pahuja I, Chaturvedi S, Prasad DVR, Dwivedi VP, Das G. The 1, 2-ethylenediamine SQ109 protects against tuberculosis by promoting M1 macrophage polarization through the p38 MAPK pathway. Commun Biol 2022; 5:759. [PMID: 35902694 PMCID: PMC9334294 DOI: 10.1038/s42003-022-03693-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/10/2022] [Indexed: 11/23/2022] Open
Abstract
Directly Observed Treatment Short-course (DOTs), is an effective and widely recommended treatment for tuberculosis (TB). The antibiotics used in DOTs, are immunotoxic and impair effector T cells, increasing the risk of re-infections and reactivation. Multiple reports suggest that addition of immune-modulators along with antibiotics improves the effectiveness of TB treatment. Therefore, drugs with both antimicrobial and immunomodulatory properties are desirable. N1-(Adamantan-2-yl)-N2-[(2E)-3,7-dimethylocta-2,6-dien-1-yl]ethane-1,2-diamine (SQ109) is an asymmetric diamine derivative of adamantane, that targets Mycobacterial membrane protein Large 3 (MmpL3). SQ109 dissipates the transmembrane electrochemical proton-gradient necessary for cell-wall biosynthesis and bacterial activity. Here, we examined the effects of SQ109 on host-immune responses using a murine TB model. Our results suggest the pro-inflammatory nature of SQ109, which instigates M1-macrophage polarization and induces protective pro-inflammatory cytokines through the p38-MAPK pathway. SQ109 also promotes Th1 and Th17-immune responses that inhibit the bacillary burden in a murine model of TB. These findings put forth SQ109 as a potential-adjunct to TB antibiotic therapy. The adamantine derivative SQ109 induces protective pro-inflammatory cytokines and promotes Th1 and Th17-immune responses that inhibit bacterial burden in a tuberculosis mouse model.
Collapse
Affiliation(s)
- Mona Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India.,Deshbandhu College, University of Delhi, Kalkaji, New Delhi, 110 019, India
| | - Santosh Kumar
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Baldeep Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Preeti Jain
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Anjna Kumari
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Isha Pahuja
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Shivam Chaturvedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | | | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.
| | - Gobardhan Das
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
47
|
Basak P, Maitra P, Khan U, Saha K, Bhattacharya SS, Dutta M, Bhattacharya S. Capsaicin Inhibits Shigella flexneri Intracellular Growth by Inducing Autophagy. Front Pharmacol 2022; 13:903438. [PMID: 35873583 PMCID: PMC9298657 DOI: 10.3389/fphar.2022.903438] [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: 03/24/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Antibiotic treatment plays an essential role in preventing Shigella infection. However, incidences of global rise in antibiotic resistance create a major challenge to treat bacterial infection. In this context, there is an urgent need for newer approaches to reduce S. flexneri burden. This study largely focuses on the role of the herbal compound capsaicin (Caps) in inhibiting S. flexneri growth and evaluating the molecular mechanism behind bacterial clearance. Here, we show for the first time that Caps inhibits intracellular S. flexneri growth by inducing autophagy. Activation of autophagy by Caps is mediated through transcription factor TFEB, a master regulator of autophagosome biogenesis. Caps induced the nuclear localization of TFEB. Activation of TFEB further induces the gene transcription of autophagosomal genes. Our findings revealed that the inhibition of autophagy by silencing TFEB and Atg5 induces bacterial growth. Hence, Caps-induced autophagy is one of the key factors responsible for bacterial clearance. Moreover, Caps restricted the intracellular proliferation of S. flexneri-resistant strain. The efficacy of Caps in reducing S. flexneri growth was confirmed by an animal model. This study showed for the first time that S. flexneri infection can be inhibited by inducing autophagy. Overall observations suggest that Caps activates TFEB to induce autophagy and thereby combat S. flexneri infection.
Collapse
Affiliation(s)
- Priyanka Basak
- Division of Biochemistry, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Priyanka Maitra
- Division of Biochemistry, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Uzma Khan
- Division of Biochemistry, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Kalyani Saha
- Division of Biochemistry, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | | | - Moumita Dutta
- Division of Electron Microscopy, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Sushmita Bhattacharya
- Division of Biochemistry, National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
48
|
Deretic V, Lazarou M. A guide to membrane atg8ylation and autophagy with reflections on immunity. J Cell Biol 2022; 221:e202203083. [PMID: 35699692 PMCID: PMC9202678 DOI: 10.1083/jcb.202203083] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 12/11/2022] Open
Abstract
The process of membrane atg8ylation, defined herein as the conjugation of the ATG8 family of ubiquitin-like proteins to membrane lipids, is beginning to be appreciated in its broader manifestations, mechanisms, and functions. Classically, membrane atg8ylation with LC3B, one of six mammalian ATG8 family proteins, has been viewed as the hallmark of canonical autophagy, entailing the formation of characteristic double membranes in the cytoplasm. However, ATG8s are now well described as being conjugated to single membranes and, most recently, proteins. Here we propose that the atg8ylation is coopted by multiple downstream processes, one of which is canonical autophagy. We elaborate on these biological outputs, which impact metabolism, quality control, and immunity, emphasizing the context of inflammation and immunological effects. In conclusion, we propose that atg8ylation is a modification akin to ubiquitylation, and that it is utilized by different systems participating in membrane stress responses and membrane remodeling activities encompassing autophagy and beyond.
Collapse
Affiliation(s)
- Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Michael Lazarou
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
49
|
Song L, Hu X, Ren X, Liu J, Liu X. Antibacterial Modes of Herbal Flavonoids Combat Resistant Bacteria. Front Pharmacol 2022; 13:873374. [PMID: 35847042 PMCID: PMC9278433 DOI: 10.3389/fphar.2022.873374] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/17/2022] [Indexed: 12/21/2022] Open
Abstract
The increasing dissemination of multidrug resistant (MDR) bacterial infections endangers global public health. How to develop effective antibacterial agents against resistant bacteria is becoming one of the most urgent demands to solve the drug resistance crisis. Traditional Chinese medicine (TCM) with multi-target antibacterial actions are emerging as an effective way to combat the antibacterial resistance. Based on the innovative concept of organic wholeness and syndrome differentiation, TCM use in antibacterial therapies is encouraging. Herein, advances on flavonoid compounds of heat-clearing Chinese medicine exhibit their potential for the therapy of resistant bacteria. In this review, we focus on the antibacterial modes of herbal flavonoids. Additionally, we overview the targets of flavonoid compounds and divide them into direct-acting antibacterial compounds (DACs) and host-acting antibacterial compounds (HACs) based on their modes of action. We also discuss the associated functional groups of flavonoid compounds and highlight recent pharmacological activities against diverse resistant bacteria to provide the candidate drugs for the clinical infection.
Collapse
Affiliation(s)
- Lianyu Song
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Changping, China
| | - Xin Hu
- Animal Science and Technology College, Beijing University of Agriculture, Changping, China
| | - Xiaomin Ren
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Changping, China
| | - Jing Liu
- Animal Science and Technology College, Beijing University of Agriculture, Changping, China
| | - Xiaoye Liu
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Changping, China
- Animal Science and Technology College, Beijing University of Agriculture, Changping, China
- *Correspondence: Xiaoye Liu,
| |
Collapse
|
50
|
Activating transcription factor 3 protects alveolar epithelial type II cells from Mycobacterium tuberculosis infection-induced inflammation. Tuberculosis (Edinb) 2022; 135:102227. [DOI: 10.1016/j.tube.2022.102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/07/2022] [Accepted: 06/19/2022] [Indexed: 11/22/2022]
|