1
|
Shaikh SA, Labhade SR, Chobe SS, Gaikwad MV, More RA, Patil BU, Boraste DR. Coumarin-Triazole-Thiazole hybrids: A new avenue in Antitubercular agents. Bioorg Chem 2025; 161:108567. [PMID: 40378733 DOI: 10.1016/j.bioorg.2025.108567] [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: 01/21/2025] [Revised: 05/06/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
The present work reports the design and synthesis of a series of 2-(4-(((2-oxo-2H-chromen-4-yl)oxy)methyl)-1H-1,2,3-triazol-1-yl)-N-(thiazol-2-yl)acetamide derivatives (4a-4k), which were investigated for their potential against tubercular activities. The well-known click cycloaddition reaction of azido derivatives (3a-3k) and 4-(prop-2-yn-1-yloxy)-2H-chromen-2-one (2) in the presence of catalytic amount of CuSO4 in H2O and DMF was employed. Notably, compounds 4c, 4d, and 4g showed the most promising results with MIC values against tuberculosis are 7.81 μg/mL, 2.61 μg/mL, and 7.81 μg/mL, respectively. Compounds (4a-4k) exhibited moderate to excellent antioxidant activities and low cytotoxicities. In addition, molecular docking studies were performed to investigate the binding interaction modes of the compounds (4a-4k) with various proteins and ligand sites.
Collapse
Affiliation(s)
- Samin A Shaikh
- Department of Chemistry, Kr. V. N. Naik Shikshan Prasarak Sanstha's Arts, Commerce and Science College, Canada Corner, Nashik 422002, Savitribai Phule Pune University, Maharashtra, India.
| | - Shivaji R Labhade
- Department of Chemistry, KTHM College, Gangapur Road, Nashik 422002, Savitribai Phule Pune University, Maharashtra, India
| | - Santosh S Chobe
- Research Centre of Chemistry, M.G. V's Loknete Vyankatrao Hiray, Arts, Science and Commerce College, Panchavati, Nashik 422003, Savitribai Phule Pune University, Maharashtra, India
| | - Milind V Gaikwad
- Department of Chemistry, Dr. D.Y. Patil Arts, Commerce and Science College, Pimpri, Savitribai Phule Pune University, Pune, 411018, Maharashtra, India
| | - Rahul A More
- Department of Microbiology, Dayanand Science College, Latur-413 512, Swami Ramanand Teerth Marathwada University, Maharashtra, India
| | - Bhausaheb U Patil
- Gokhale Education Society's Post Graduate Department of Chemistry and Research Centre, HPT Arts and RYK Science College, Nashik 422005, Savitribai Phule Pune University, Maharashtra, India; Gokhale Education Society's Department of Chemistry, Arts, Commerce and Science College, Shreewardhan, Raigad, 402110, Mumbai University, Maharashtra, India
| | - Deepak R Boraste
- Department of Chemistry, Pohang University of Science and Technology, Pohang, South Korea
| |
Collapse
|
2
|
Alyokhin AV, Rosenthal BM, Weber DC, Baker MB. Towards a unified approach in managing resistance to vaccines, drugs, and pesticides. Biol Rev Camb Philos Soc 2025; 100:1067-1082. [PMID: 39807648 DOI: 10.1111/brv.13174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Everywhere, pests and pathogens evolve resistance to our control efforts, impairing human health and welfare. Developing sustainable solutions to this problem requires working with evolved immune and ecological systems, rather than against these evolutionary forces. We advocate a transdisciplinary approach to resistance based on an evolutionary foundation informed by the concepts of integrated pest management and One Health. Diverse, multimodal management approaches create a more challenging environment for the evolution of resistance. Given our permanent evolutionary and ecological relationships with pests and pathogens, responses to most biological threats to health and agriculture should seek sustainable harm reduction rather than eradication.
Collapse
Affiliation(s)
- Andrei V Alyokhin
- School of Biology and Ecology, University of Maine, 5722 Dering Hall, Orono, 04469, Maine, USA
| | - Benjamin M Rosenthal
- Animal Parasitic Diseases Laboratory, Agricultural Research Service, US Department of Agriculture, 10300 Baltimore Avenue, Beltsville, 20705, Maryland, USA
| | - Donald C Weber
- Invasive Insect Biocontrol and Behaviour Laboratory, Agricultural Research Service, US Department of Agriculture, 10300 Baltimore Avenue, Beltsville, 20705, Maryland, USA
| | - Mitchell B Baker
- Biology Department, Queens College, City University of New York, 149th St, Flushing, 11367, New York, USA
| |
Collapse
|
3
|
Xu JT, Li K, Lin Y, Cheng T, Gu J, Chen YK, Yu JF, Deng JY. Diverse impacts of different rpoB mutations on the anti-tuberculosis efficacy of capreomycin. EBioMedicine 2025; 117:105776. [PMID: 40449326 DOI: 10.1016/j.ebiom.2025.105776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 04/06/2025] [Accepted: 05/12/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Since the discovery of streptomycin in the 1940s, more than a dozen drugs have been continuously introduced into tuberculosis (TB) therapy. However, limited attention has been paid to the collateral effects of drug resistance evolution in Mycobacterium tuberculosis (Mtb). Recently, we observed a clear discordance between the capreomycin (CAP) susceptibility of rifampicin-resistant (RR) Mtb clinical isolates and the adverse outcomes associated with CAP treatment, indicating potential collateral effects between rpoB mutations and CAP. To explore this relationship, we integrated clinical isolate data, experimental evolution data, phenotypic data, sequencing data, and genome-wide association studies (GWAS). METHODS We analysed the correlations between CAP resistance and rpoB mutations at various loci based on phenotypic drug susceptibility testing (pDST) profiles and rpoB sequencing data from 565 RR Mtb isolates collected in southwestern China. To validate the clinical observations, we screened RR mutants of Mtb H37Rv and conducted rpoB sequencing to characterise the mutation sites. Additionally, we constructed various rpoB mutants in Mycobacterium smegmatis (Ms). We then examined the impact of these mutations on the efficacy of CAP through minimum inhibitory concentration (MIC) tests and time-kill assays in both Mtb and Ms rpoB mutants. Furthermore, we investigated the influence of three major rpoB mutations on the frequency of occurrence of rrs A1401G-associated with CAP resistance-using a GWAS of 607 Mtb genomes from a global dataset. FINDINGS By analysing 565 clinical isolates from southwestern China, we found that the CAP resistance in isolates with a single mutation at rpoB site 445 was significantly lower than in those with a single mutation at other sites (P < 0.05, Pearson chi-square test and Fisher exact test; odds ratio = 0.272). In contrast, the opposite trend was observed in isolates with a single mutation at rpoB site 435 (P < 0.001, Pearson chi-square test and Fisher exact test; odds ratio = 3.067). Subsequently, using laboratory-evolved RR mutants, we demonstrated that mutations at rpoB site 445 or site 441 enhanced the bactericidal effect of CAP. However, the opposite result was observed in mutants with mutations at rpoB site 435. Furthermore, we found that the occurrence frequency of the rrs A1401G mutation was significantly lower in clinical isolates with rpoB mutations at site 445, but significantly higher in those with mutations at site 435. INTERPRETATION Although rpoB mutations in Mtb did not affect the MIC of CAP, they influenced its bactericidal effect, highlighting the need for time-kill assays when investigating collateral effects. Different rpoB mutations may exert diverse impacts on the bactericidal effect of CAP-or CAP tolerance-underscoring the complexity of collateral effects and supporting the use of targeted sequencing in the molecular diagnosis of RR Mtb. As RNA polymerase plays a central role in bacterial RNA transcription, it regulates most metabolic processes in Mtb. Thus, different rpoB mutations may elicit distinct gene expression profiles upon CAP treatment, a hypothesis warranting further investigation. Additional clinical studies are needed to verify whether the adverse outcomes of CAP treatment are associated with infections caused by strains harbouring rpoB mutations at site 435. If so, such outcomes could be mitigated through rational drug regimens guided by precise molecular diagnosis. This study provides insights into the collateral effects of drug resistance mutations and advances the case for precision medicine in treating infections caused by drug-resistant bacteria. FUNDING Funding for this study was provided by the National Key Research and Development Program of China (Grant no. 2021YFA1300901).
Collapse
Affiliation(s)
- Jin-Tian Xu
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kun Li
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing 400036, China
| | - Yi Lin
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Cheng
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Gu
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yao-Kai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing 400036, China.
| | - Ji-Fang Yu
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Jiao-Yu Deng
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
4
|
Chen W, Luo C, Zhou H, Liu Z, Huang J, Liu Y, You M, Yang G. Mitophagy-mtROS axis contributes to anti-tuberculosis-induced liver injury through activation of the cGAS-STING pathway in rat hepatocytes. Int Immunopharmacol 2025; 160:114984. [PMID: 40449272 DOI: 10.1016/j.intimp.2025.114984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 05/17/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
Tuberculosis (TB) remains a major worldwide healthcare issue, with anti-TB drugs playing a pivotal role in its treatment. However, the emergence of anti-TB drug-induced liver injury (ATB-DILI) poses a considerable challenge, undermining treatment efficacy and patient survival. This study investigates the underlying mechanisms of ATB-DILI, focusing on reactive oxygen species (ROS), mitophagy, lysosomal function, and the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling pathway. A rat hepatocyte model treated with standard anti-TB drugs was established to assess liver inflammation, oxidative stress biomarkers, mitochondrial function, and mitophagy processes. The results indicate that anti-TB drug administration induced significant inflammatory injury, characterized by elevated IL-6 and reduced IL-4 and IL-10 levels. ROS overproduction predominantly originates in the mitochondrial level, consequently resulting in oxidative stress and impaired mitochondrial function. A noticeable decline in both the oxygen consumption rate and ATP production is indicative of this phenomenon. Although mitophagy was activated, impaired lysosomal function hindered mitophagic flux, leading to the buildup of damaged mitochondria and ROS. Pharmacological intervention with mitoTEMPO alleviated mitochondrial dysfunction, while clioquinol restored lysosomal function and improved mitophagy. Additionally, the cGAS-STING signaling pathway was found to regulate inflammation in ATB-DILI, with both mitoTEMPO and clioquinol alleviating its effects. These findings elucidate the crucial impact of lysosome-mediated mitophagy dysfunction and mitochondrial ROS in ATB-DILI, highlighting potential therapeutic targets to enhance liver protection during anti-TB treatment.
Collapse
Affiliation(s)
- Wenyan Chen
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China
| | - Chenjunlei Luo
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China
| | - He Zhou
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China
| | - Zhenhui Liu
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China
| | - Junfei Huang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China
| | - Yining Liu
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou 550004, China
| | - Mingdan You
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, China.
| | - Guanghong Yang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 561113, China; Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou 550004, China.
| |
Collapse
|
5
|
Wu H, Huang MY, Xue ZJ, Zhong SY, Shi JQ, Chen NP, Qian CD. Targeting type II NADH dehydrogenase in tuberculosis treatment: A review. Int J Biol Macromol 2025; 310:143541. [PMID: 40288271 DOI: 10.1016/j.ijbiomac.2025.143541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Tuberculosis (TB), a critical global health issue, continues to impose significant harm on human populations worldwide, and the increasing prevalence of drug-resistant TB strains has exacerbated the challenges in effective treatment, underscoring an urgent need for the development of new therapeutic agents with innovative mechanisms of action. The introduction of bedaquiline highlights targeting Mycobacterium tuberculosis (Mtb) energy metabolism as a novel anti-TB strategy. Among the various targets within Mtb's metabolic pathways, type II NADH dehydrogenase (NDH-2) is of particular significance due to its critical function in bacterial respiration and its absence from human cells, rendering it an appealing candidate for selective inhibition. Recent advances have led to the identification of numerous NDH-2 inhibitors, some of which exhibit potent antibacterial activity at nanomolar concentrations, demonstrating their potential as lead compounds for future drug development. This review explores the biochemical function and molecular structure of NDH-2, underscoring its potential as a target for anti-TB therapies. It also discusses the progress made in discovering and optimizing NDH-2 inhibitors, offering insights into their mechanisms of action, efficacy, and pharmacological properties, with the aim of providing an overview that could inform and guide future research efforts towards developing more effective treatments against TB.
Collapse
Affiliation(s)
- Han Wu
- College of Second Clinical Medical, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ming-Yu Huang
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zheng-Jie Xue
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Si-Yi Zhong
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jia-Qi Shi
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ni-Pi Chen
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Chao-Dong Qian
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
6
|
Liu S, Xiao G, Liang W, Zhang S, Liang J, Pan T, Lin S, Liu X, Zhou Z, Zhang G. Vγ9Vδ2 T cells expanded with vitamin C combined with HMBPP in vitro inhibit intracellular Mycobacterium tuberculosis growth. Front Cell Infect Microbiol 2025; 15:1533277. [PMID: 40370406 PMCID: PMC12075132 DOI: 10.3389/fcimb.2025.1533277] [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: 11/23/2024] [Accepted: 04/04/2025] [Indexed: 05/16/2025] Open
Abstract
Background Mycobacterium tuberculosis (Mtb) presents a significant global health threat, and the existing treatments have notable limitations. Vγ9Vδ2 T cells activated by HMBPP can inhibit the growth of intracellular Mtb. Additionally, vitamin C (VC) promotes the differentiation and proliferation of related T cells. However, it remains uncertain whether VC can enhance the expansion of Vγ9Vδ2 T cells within PBMCs activated by HMBPP and rIL-2, and the underlying mechanism of the inhibitory effect of the expanded T cells on intracellular Mtb has not been elucidated. Methods Venous blood was collected from healthy individuals, and PBMCs were subsequently isolated. In vitro, Vγ9Vδ2 T cells were selectively expanded with HMBPP, rIL-2, and VC. Flow cytometry was utilized to analyze the purities and phenotypes of Vγ9Vδ2 T cells, while cell counts were performed to determine the total number of viable cells. Magnetic bead sorting was employed to purify Vγ9Vδ2 T cells. Mtb strains were cultured, and macrophage infection models derived from THP1 cells were established. Co-culture experiments were conducted with Mtb-infected macrophages and Vγ9Vδ2 T cells, and the number of intracellular bacteria was quantified through CFU counting. The levels of cytokines were measured using the CBA method and flow cytometry. Statistical analysis was carried out using GraphPad Prism and SPSS software. Results VC (70 μM) significantly enhances the expansion of Vγ9Vδ2 T cells within PBMCs during primary HMBPP activation in the presence of rIL-2, with higher induction rates and total cell proliferation. By day 14 of induction, Vγ9Vδ2 T cells expanded with HMBPP, VC, and rIL-2 exhibited the central memory (10-20%) and the effector memory phenotypes (75-90%). Furthermore, these expanded T cells effectively inhibited the growth of intracellular virulent Mtb strain (H37Rv) in a cell-contact-dependent manner. The inhibitory effect was associated with an up-regulated production of TNF-α and IFN-γ, and a down-regulated expression of IL-10 and IL-17A during Mtb infection. Conclusion This study demonstrates that VC enhances the proliferative expansion of Vγ9Vδ2 T cells in PBMCs primarily stimulated with HMBPP and rIL-2. The expanded Vγ9Vδ2 T cells are capable of effectively inhibiting the growth of virulent H37Rv strain, likely through the secretion of TNF-α and IFN-γ. These findings provide a novel direction for tuberculosis treatment research.
Collapse
Affiliation(s)
- Shuyan Liu
- 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
- Longgang Maternity and Child Institute of Shantou University Medical College, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China
| | - Guohui Xiao
- 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
| | - Wanxin Liang
- 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
| | - Su 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
| | - Juan Liang
- 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
| | - Teng Pan
- Longgang Maternity and Child Institute of Shantou University Medical College, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China
| | - Shaoxiang Lin
- Longgang Maternity and Child Institute of Shantou University Medical College, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China
| | - Xiuju Liu
- Longgang Maternity and Child Institute of Shantou University Medical College, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China
| | - Zhenwen Zhou
- Longgang Maternity and Child Institute of Shantou University Medical College, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, 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
|
7
|
Liyew AM, Clements ACA, Wagnew F, Gilmour B, Alene KA. Geospatial mapping of drug-resistant tuberculosis prevalence in Africa at national and sub-national levels. Int J Infect Dis 2025; 153:107777. [PMID: 39788484 DOI: 10.1016/j.ijid.2025.107777] [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: 09/09/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVES To map subnational and local prevalence of drug-resistant tuberculosis (DR-TB) across Africa. METHODS We assembled a geolocated dataset from 173 sources across 31 African countries, comprising drug susceptibility test results and covariate data from publicly available databases. We used Bayesian model-based geostatistical framework with multivariate Bayesian logistic regression model to estimate DR-TB prevalence at lower administrative levels. RESULTS We estimated 148,239 DR-TB cases (95% uncertainty interval: 17,499-313,683) in Africa, showing significant variation by country. Eswatini and South Africa had highest case numbers, while Algeria and Egypt had the lowest. The highest DR-TB prevalence was estimated in Eswatini (53.26; 95% uncertainty interval 13.13-66.12), Morocco, Tunisia, and South Africa, while the lowest prevalence was found in Gabon, the Republic of Congo, Sierra Leone, and Mali. Marked subnational variation in DR-TB prevalence was noted, where 91 subnational areas across 12 countries had prevalence rates higher than their respective national averages. Factors such as mean temperature (β = 2.01; 95% CrI: 1.21, 3.42), population density (β = 0.41; 95% CrI: 0.19, 0.95), and fine particulate matter (β = 0.66; 95% CrI: 0.20, 0.80) were positively associated with DR-TB prevalence. CONCLUSION The study highlights substantial national and subnational variability in DR-TB prevalence across Africa, aiding policymakers in designing localized TB control interventions.
Collapse
Affiliation(s)
- Alemneh Mekuriaw Liyew
- Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia; School of Population Health, Faculty of Health Sciences, Curtin University, Perth, Australia; Geospatial and Tuberculosis Research Team, Telethon Kids Institute, Perth, Australia.
| | - Archie C A Clements
- Geospatial and Tuberculosis Research Team, Telethon Kids Institute, Perth, Australia; Research and Enterprise, Queen's University Belfast, Belfast, UK
| | - Fasil Wagnew
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; National Centre for Epidemiology and Population Health (NCEPH), College of Health and Medicine, The Australian National University, Canberra, Australia
| | - Beth Gilmour
- School of Population Health, Faculty of Health Sciences, Curtin University, Perth, Australia; Geospatial and Tuberculosis Research Team, Telethon Kids Institute, Perth, Australia
| | - Kefyalew Addis Alene
- School of Population Health, Faculty of Health Sciences, Curtin University, Perth, Australia; Geospatial and Tuberculosis Research Team, Telethon Kids Institute, Perth, Australia
| |
Collapse
|
8
|
Ray B, Roy KK. Deciphering insights into the binding mechanism and plasticity of Telacebec with M. tuberculosis cytochrome bcc-aa3 supercomplex through an unbiased molecular dynamics simulation, free-energy analysis, and DFT study. J Biomol Struct Dyn 2025; 43:2968-2981. [PMID: 38111165 DOI: 10.1080/07391102.2023.2294833] [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: 06/29/2023] [Accepted: 12/02/2023] [Indexed: 12/20/2023]
Abstract
The cytochrome bcc-aa3 supercomplex, a key component in the electron transport chain pathway involved in bacterial energy production and homeostasis, is a clinically validated target for tuberculosis (TB), leading to Telacebec (Q203). Telacebec is a potent candidate drug under Phase II clinical development for the treatment of drug-sensitive and drug-resistant TB. Recently, the cryo-electron microscopy structure of this supercomplex from Mycobacterium tuberculosis (Mtb) complexed with Q203 was resolved at 6.9 Å resolution (PDB ID: 7E1W). To understand the binding site (QP site) flexibility and Q203's stability at the QP site of the Mtb cytochrome bcc complex, we conducted molecular dynamics (MD) simulation and free energy analysis on this complex in an explicit hydrated lipid bilayer environment for 500 ns. Through this study, the persistence of a range of direct and indirect interactions was observed over the course of the simulation. The significance of the interactions with His375, Tyr161, Ala178, Ala179, Ile183, His355, Leu356, and Thr313 is underlined. Electrostatic energy was the primary source of the net binding free energy, regardless of the important interacting residues. The overall binding free energy for Q203 was -112.84 ± 7.73 kcal/mol, of which the electrostatic and lipophilic energy contributions were -116.31 ± 1.14 and -21.32 ± 2.35 kcal/mol, respectively. Meanwhile, DFT calculations were utilized to elucidate Q203's molecular properties. Overall, this study deciphers key insights into the cytochrome bcc-aa3 supercomplex with Q203 on the ground of molecular mechanics and quantum mechanics that may facilitate structure-based drug design and optimization for the discovery of the next-generation antitubercular drug(s).
Collapse
Affiliation(s)
- Bedabrata Ray
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, India
| | - Kuldeep K Roy
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, India
| |
Collapse
|
9
|
Tayal S, Singh V, Bhatnagar S. 3D-QSAR and ADMET studies of morpholino-pyrimidine inhibitors of DprE1 from Mycobacterium tuberculosis. J Biomol Struct Dyn 2025; 43:2948-2967. [PMID: 38112325 DOI: 10.1080/07391102.2023.2294496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023]
Abstract
DprE1 is involved in the synthesis of Mycobacterium tuberculosis cell wall and is a potent drug target for Tuberculosis (TB) treatment. The structure and dynamics of the loops L-I and L-II flanking the inhibitor binding site was studied using molecular dynamics (MD) simulation and MMPBSA in Amber v18. Docking and three-dimensional quantitative structure-activity relationship (3D-QSAR) of 55 Morpholino-pyrimidine (MP) inhibitors was carried out using Autodock v1.2.0 and Forge v10. ADMET analysis was done using SwissADME and pkCSM. All MP inhibitors docked in the DprE1 binding pocket, making contacts with L-II residues. MD studies showed that L-I and L-II unfold in the absence of the inhibitor but fold stably structure with reduced protein motions in the presence of MP-38, the highest affinity inhibitor. This was confirmed by k-means clustering and secondary structure analysis. L-II residues, L317, F320 and R325 contributed most towards the MMPBSA binding free energy of MP-38. A robust field-based 3D-QSAR model showed values of r2train = 0.982, r2test = 0.702 and q2 = 0.516. The MP inhibitor field points were broadly divided into negative electrostatics near the A, B rings and hydrophobic electrostatics near the D, E rings. Addition of negative groups at methanone position and ring B as well as addition of hydrophobic and bulky groups at ring E will improve activity. Highly active compounds 47, 49 and 50 of MP series exhibited highly favourable drug-like properties. SAR and ADMET insights attained from this model will help in the development of active DprE1 inhibitors in future.
Collapse
Affiliation(s)
- Sonali Tayal
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, India
| | - Vasundhara Singh
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, India
| | - Sonika Bhatnagar
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, India
| |
Collapse
|
10
|
Zhang X, Li Z, Tao B, Fu Y, Cui C, Wang F, Li Y, Wang Y, Jiang J, Wang J. Outdoor particulate matter and risk of drug resistance for workers and farmers with pulmonary tuberculosis: a population-based time-series study in Suzhou, China. BMJ Open 2025; 15:e089290. [PMID: 40139714 PMCID: PMC11950948 DOI: 10.1136/bmjopen-2024-089290] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVES The detrimental effects of particulate matter (PM) on human health have been widely corroborated. We aimed to examine the association between outdoor PM and the drug resistance risk among workers and farmers with pulmonary tuberculosis (PTB). DESIGN We performed a population-based time-series study using routinely collected meteorological and TB surveillance data. SETTING We selected Suzhou City, China, as the study area. Data on patients with PTB and meteorological factors were extracted from the National Tuberculosis Online Registration System and the China Meteorological Data Sharing Center. PARTICIPANTS This study included 7868 patients with PTB diagnosed from January 2017 to December 2021 in Suzhou. METHODS The generalised additive model was used to estimate the effects of outdoor PM on the drug resistance risk of TB among workers and farmers who typically work outdoors. Moreover, subgroup analyses were carried out to evaluate the associations in different populations and seasons. RESULTS Although there was no significant association between PM with an aerodynamic diameter≤10 µm (PM10) and drug-resistant risk in the overall analysis, subgroup analysis revealed a significant positive association in the winter season. Similarly, PM with an aerodynamic diameter≤2.5 µm (PM2.5) was significantly associated with drug resistance risk among males with a lag of 0-3 days, people ≤60 years with a lag of 0-7 days and in the winter season with a lag of 0-7 days, 0-15 days, 0-90 days or 0-180 days. CONCLUSIONS Outdoor PM10 and PM2.5 were positively related to the drug resistance risk of workers and farmers with PTB. Reducing ambient PM pollution might reduce the burden of TB. Further research is required to verify the association through in vitro experiments and extensive cohort studies.
Collapse
Affiliation(s)
- Xiaolong Zhang
- Department of Tuberculosis Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, China
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Epidemiology, Gusu School, Nanjing Medical University, Nanjing, China
| | - Zhongqi Li
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Bilin Tao
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ying Fu
- Department of Tuberculosis Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, China
| | - Caiyan Cui
- Department of Tuberculosis Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, China
| | - Feixian Wang
- Department of Tuberculosis Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, China
| | - Yun Li
- Department of Tuberculosis Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, China
| | - Yu Wang
- Department of Tuberculosis Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, China
| | - Jun Jiang
- Department of Tuberculosis Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, China
| | - Jianming Wang
- Department of Epidemiology, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Epidemiology, Gusu School, Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
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
|
12
|
Dunkley ORS, Bell AG, Modi NH, Huang Y, Tseng S, Reiss R, Daivaa N, Davis JL, Vargas DA, Banada P, Xie YL, Myhrvold C. A Streamlined Point-of-Care CRISPR Test for Tuberculosis Detection Directly from Sputum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.19.25322517. [PMID: 40034782 PMCID: PMC11875272 DOI: 10.1101/2025.02.19.25322517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Mycobacterium tuberculosis (Mtb) is a major threat to global health and is responsible for over one million deaths each year. To stem the tide of cases and maximize opportunities for early interventions, there is an urgent need for affordable and simple means of tuberculosis diagnosis in under-resourced areas. We sought to develop a CRISPR-based isothermal assay coupled with a compatible, straightforward sample processing technique for point-of-care use. Here, we combine Recombinase Polymerase Amplification (RPA) with Cas13a and Cas12a, to create two parallelised one-pot assays that detect two conserved elements of Mtb (IS6110 and IS1081) and an internal control targeting human DNA. These assays were shown to be compatible with lateral flow and can be readily lyophilized. Our finalized assay exhibited sensitivity over a wide range of bacterial loads (105 to 102 CFU/mL) in sputum. The limit of detection (LoD) of the assay was determined to be 69.0 (51.0 - 86.9) CFU/mL for Mtb strain H37Rv spiked in sputum and 80.5 (59.4 - 101.6) CFU/mL for M. bovis BCG. Our assay showed no cross reactivity against a wide range of bacterial/fungal isolates. Clinical tests on 13 blinded sputum samples revealed 100% (6/6) sensitivity and 100% (7/7) specificity compared to culture. Our assay exhibited comparable sensitivity in clinical samples to the microbiological gold standard, TB culture, and to the nucleic acid state-of-the-art, GeneXpert MTB/RIF Ultra. This technology streamlines TB diagnosis from sample extraction to assay readout in a rapid and robust format, making it the first test to combine amplification and detection while being compatible with both lateral flow and lyophilization.
Collapse
Affiliation(s)
- Owen R. S. Dunkley
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Alexandra G. Bell
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Nisha H. Modi
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Yujia Huang
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Soleil Tseng
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Robert Reiss
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Naranjargal Daivaa
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - J. Lucian Davis
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Pulmonary, Critical Care, and Sleep Medicine Section, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Deninson Alejandro Vargas
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Padmapriya Banada
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Yingda L. Xie
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, 08544, USA
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, New Jersey, 08544, USA
- Department of Chemistry, Princeton University, Princeton, New Jersey, 08544, USA
| |
Collapse
|
13
|
Sidrônio MGS, Freitas MEG, Magalhães DWA, Carvalho DCM, Gonçalves VAB, Oliveira ACMDQ, Paulino GC, Borges GC, Ribeiro RL, de Sousa NF, Scotti MT, de Araújo DAM, Mendonça-Junior FJB, Freire KRDL, Rodrigues-Mascarenhas S, Santos BVDO, Rodrigues-Junior VS. Host-Mediated Antimicrobial Effects and NLRP3 Inflammasome Modulation by Caulerpin and Its Derivatives in Macrophage Models of Mycobacterial Infections. Microorganisms 2025; 13:561. [PMID: 40142455 PMCID: PMC11944515 DOI: 10.3390/microorganisms13030561] [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: 12/26/2024] [Revised: 01/30/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Caulerpin, a bis-indole alkaloid isolated from Caulerpa racemosa, has several documented pharmacological activities, including antineoplastic and antiviral properties. This study aimed to evaluate the anti-inflammatory and anti-tubercular potentials of caulerpin and its analogues in RAW 264.7 macrophages infected with Mycobacterium spp. Additionally, we evaluated cytokine production and NLRP3 expression in this infection model. Toxicity tests were performed using Vero E6 and HepG2 cell lines and Artemia salina. Pre-incubation of RAW 264.7 cells with caulerpin and its analogues decreased internalized M. smegmatis and M. tuberculosis H37Ra. Furthermore, treatment of M. smegmatis-infected macrophages with caulerpin and its analogues reduced bacterial loads. Caulerpin reduced the CFU count of internalized bacilli in the M. tuberculosis H37Ra infection model. In addition, caulerpin and its diethyl derivative were notably found to modulate IL-1β and TNF-α production in the M. smegmatis infection model after quantifying pro-inflammatory cytokines and NLRP3. Caulerpin and its derivates did not affect the viability of Vero E6 and HepG2 cell lines or nauplii survival in toxicity studies. These findings demonstrate that caulerpin and its analogues exhibit anti-inflammatory activity against Mycobacterium spp. infection in RAW 264.7 macrophages and show promising potential for further efficacy and safety evaluation.
Collapse
Affiliation(s)
- Maria Gabriella S. Sidrônio
- Postgraduate Program in Development and Technological Innovation in Medicines, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (M.G.S.S.); (B.V.d.O.S.)
| | - Maria Eugênia G. Freitas
- Laboratory of Biotechnology in Microorganisms, Biotechnology Center, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (M.E.G.F.); (G.C.P.); (G.C.B.); (R.L.R.)
| | - Daniel W. A. Magalhães
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (D.W.A.M.); (D.C.M.C.); (S.R.-M.)
| | - Deyse C. M. Carvalho
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (D.W.A.M.); (D.C.M.C.); (S.R.-M.)
| | - Vinícius A. B. Gonçalves
- Department of Cell and Molecular Biology, Biotechnology Center, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (V.A.B.G.); (K.R.d.L.F.)
| | | | - Gisela C. Paulino
- Laboratory of Biotechnology in Microorganisms, Biotechnology Center, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (M.E.G.F.); (G.C.P.); (G.C.B.); (R.L.R.)
| | - Gabriela C. Borges
- Laboratory of Biotechnology in Microorganisms, Biotechnology Center, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (M.E.G.F.); (G.C.P.); (G.C.B.); (R.L.R.)
| | - Rafaelle L. Ribeiro
- Laboratory of Biotechnology in Microorganisms, Biotechnology Center, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (M.E.G.F.); (G.C.P.); (G.C.B.); (R.L.R.)
| | - Natália Ferreira de Sousa
- Postgraduate Program in Natural and Synthetic Bioactive Products, Department of Pharmaceutical Sciences, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (N.F.d.S.); (M.T.S.)
| | - Marcus T. Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Department of Pharmaceutical Sciences, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (N.F.d.S.); (M.T.S.)
| | - Demétrius A. M. de Araújo
- Postgraduate Program in Biotechnology (Renorbio), Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil;
| | - Francisco Jaime B. Mendonça-Junior
- Laboratory of Synthesis and Drug Delivery, Department of Biological Sciences, State University of Paraíba, João Pessoa 58071-160, PB, Brazil;
| | - Kristerson R. de Luna Freire
- Department of Cell and Molecular Biology, Biotechnology Center, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (V.A.B.G.); (K.R.d.L.F.)
| | - Sandra Rodrigues-Mascarenhas
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (D.W.A.M.); (D.C.M.C.); (S.R.-M.)
| | - Bárbara Viviana de O. Santos
- Postgraduate Program in Development and Technological Innovation in Medicines, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (M.G.S.S.); (B.V.d.O.S.)
- Center for Teacher Training, UACEN, University of Campina Grande, Cajazeiras 58900-000, PB, Brazil
| | - Valnês S. Rodrigues-Junior
- Postgraduate Program in Natural and Synthetic Bioactive Products, Department of Pharmaceutical Sciences, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil; (N.F.d.S.); (M.T.S.)
| |
Collapse
|
14
|
Zhu J, Wang H, Chen L. Recent advances in nanomaterials for the detection of mycobacterium tuberculosis (Review). Int J Mol Med 2025; 55:36. [PMID: 39717951 PMCID: PMC11722055 DOI: 10.3892/ijmm.2024.5477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
The world's leading infectious disease killer tuberculosis (TB) has >10 million new cases and ~1.5 million mortalities yearly. Effective TB control and management depends on accurate and timely diagnosis to improve treatment, curb transmission and reduce the burden on the medical system. Current clinical diagnostic methods for tuberculosis face the shortcomings of limited accuracy and sensitivity, time consumption and high cost of equipment and reagents. Nanomaterials have markedly enhanced the sensitivity, specificity and speed of TB detection in recent years, owing to their distinctive physical and chemical features. They offer several biomolecular binding sites, enabling the simultaneous identification of multiple TB biomarkers. Biosensors utilizing nanomaterials are often compact, user‑friendly and well‑suited for detecting TB on location and in settings with limited resources. The present review aimed to review the advances that have occurred during the last five years in the application of nanomaterials for TB diagnostics, focusing on their detection capabilities, structures, working principles and the significance of key nanomaterials. The current review addressed the limitations and challenges of nanomaterials‑based TB diagnostics, along with potential solutions.
Collapse
Affiliation(s)
- Jianmeng Zhu
- Clinical Laboratory of Chun'an First People's Hospital, Zhejiang Provincial People's Hospital Chun'an Branch, Affiliated Chun'an Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 311700, P.R. China
| | - Hongqin Wang
- Clinical Laboratory of Chun'an First People's Hospital, Zhejiang Provincial People's Hospital Chun'an Branch, Affiliated Chun'an Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 311700, P.R. China
| | - Lili Chen
- Orthopedics of Chun'an First People's Hospital, Zhejiang Provincial People's Hospital Chun'an Branch, Affiliated Chun'an Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 311700, P.R. China
| |
Collapse
|
15
|
Fujita DM, Alvarenga RF, de Andrade HF. Trends in tuberculosis and inequality-adjusted Human Development Index in Brazil, 2018-2022. J Public Health Policy 2025; 46:139-148. [PMID: 39710742 DOI: 10.1057/s41271-024-00543-9] [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] [Accepted: 12/08/2024] [Indexed: 12/24/2024]
Abstract
Tuberculosis (TB) remains a significant global health concern, causing 1.5 million deaths annually. We conducted an ecological analysis to examine TB prevalence in Brazil from 2018 to 2022, focusing on its relationship with the Human Development Index (HDI) and its inequality-adjusted version HDI (IHDI). We determined significant correlation between HDI and TB prevalence for the period of 2018-2021 (P = 0.044 in 2018, P = 0.041 in 2019, P = 0.044 in 2020, and P = 0.043 in 2021). However, an unexpected rise in TB cases in 2022, despite an increase in HDI, disrupted this trend (P = 0.12), indicating additional influencing factors. The IHDI showed correlation with TB prevalence, underscoring the importance of addressing socio-economic inequalities. These findings suggest the need for targeted interventions, particularly for vulnerable populations.
Collapse
Affiliation(s)
- Dennis Minoru Fujita
- Anhembi-Morumbi University-Post-Graduation Program in Hospitality, São Paulo, Brazil.
- Department of Pathology, FMUSP, Av. Dr. Enéas de Carvalho Aguiar, 470, São Paulo, Brazil.
- LIM49-HCFMUSP, Av. Dr. Enéas de Carvalho Aguiar, 470, São Paulo, SP, CEP 05403-000, Brazil.
| | | | - Heitor Franco de Andrade
- Department of Pathology, FMUSP, Av. Dr. Enéas de Carvalho Aguiar, 470, São Paulo, Brazil
- LIM49-HCFMUSP, Av. Dr. Enéas de Carvalho Aguiar, 470, São Paulo, SP, CEP 05403-000, Brazil
| |
Collapse
|
16
|
Singh V. The next generation of drug resistant tuberculosis drug design. Future Med Chem 2025; 17:385-387. [PMID: 39814693 PMCID: PMC11834412 DOI: 10.1080/17568919.2025.2453406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Affiliation(s)
- Vinayak Singh
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
17
|
Agnivesh PK, Roy A, Sau S, Kumar S, Kalia NP. Advancements and challenges in tuberculosis drug discovery: A comprehensive overview. Microb Pathog 2025; 198:107074. [PMID: 39521155 DOI: 10.1016/j.micpath.2024.107074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Tuberculosis continues to pose a health challenge causing the loss of millions of lives despite the existence of multiple drugs, for treatment. The emergence of drug-resistant strains has made the situation more complex making it increasingly difficult to fight against this disease. This review outlines the challenges associated with TB drug discovery, the nature of Mycobacterium tuberculosis shedding light on the mechanisms that lead to treatment failure and antibiotic resistance. We explore promising drug targets, encompassing inhibition of mycolyarabinogalactan peptidoglycan (MAGP) assembly, mycolic acid biosynthesis, DNA replication, transcription, translation, protein synthesis, and bioenergetics/metabolism pathways. A comprehensive overview of the global pipeline of anti-tuberculosis drugs at various stages of clinical trials, the diverse strategies being pursued to tackle this complex disease. By gaining an understanding of the mechanisms that contribute to resistance development and identifying suitable targets, we can pave the way for more effective treatments and contribute to global efforts to combat drug-resistant tuberculosis.
Collapse
Affiliation(s)
- Puja Kumari Agnivesh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Arnab Roy
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Shashikanta Sau
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Sunil Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Nitin Pal Kalia
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| |
Collapse
|
18
|
Patel S, Naik L, Das M, Nayak DK, Dandsena PK, Mishra A, Kumar A, Dirisala VR, Mishra A, Das S, Singh R, Behura A, Dhiman R. Furamidine-induced autophagy exerts an anti-mycobacterial effect in a SIRT1-pAMPK-FOXO3a-dependent manner by elevation of intracellular Ca 2+ level expression. Microbiol Res 2025; 290:127976. [PMID: 39591744 DOI: 10.1016/j.micres.2024.127976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024]
Abstract
Mycobacterium tuberculosis (M. tb), the etiological agent of tuberculosis (TB), continues to be a major contributor to global mortality rates. To effectively combat this pandemic, TB control has to be enhanced in several areas, including point-of-care diagnostics, shorter and safer drug regimens, and preventative vaccination. The latest findings have highlighted autophagy as a host-defense mechanism that eradicates many invading bacteria, including M. tb. Thus, novel approaches like the stimulation of autophagy using various pharmaceutical drugs can be undertaken to deal with this noxious pathogen. The present study has been formulated to evaluate the anti-mycobacterial potential of Furamidine, a DNA minor groove binder (MGB). Initially, a non-cytotoxic concentration of Furamidine (10 µM) was used to assess its impact on the intracellular persistence of mycobacteria in differentiated THP-1 (dTHP-1) cells. Furamidine treatment compromised intracellular mycobacterial growth compared to control cells. Autophagy, a well-known host-defensive strategy, was investigated as a possible contributor to revealing the mechanism of action. Multiparametric approaches such as LC3-I to II conversion, protein level expression of different autophagic markers, and MDC staining were employed to study autophagic response that conclusively suggested the autophagy induction potential of Furamidine in dTHP-1 cells. Further, elevated LC3-II expression and increased autophagic vacuole accumulation under Baf-A1 treatment demonstrated the positive regulation of autophagic flux upon Furamidine treatment. Mechanistic investigations showed increased intracellular calcium (Ca2+) level expression, SIRT1, pAMPK, and FOXO3a activation upon its treatment. Inhibition of Ca2+ level expression suppressed Ca2+-mediated-FOXO3a level in Furamidine-treated cells. Furthermore, administering various inhibitors hampered the Furamidine-induced autophagy that impacted intracellular mycobacteria clearance. These results conclude that Furamidine triggered the Ca2+/pAMPK/SIRT1/FOXO3a pathway, causing less mycobacterial load in dTHP-1 cells.
Collapse
Affiliation(s)
- Salina Patel
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Lincoln Naik
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Mousumi Das
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Dev Kiran Nayak
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Pramathesh Kumar Dandsena
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Abtar Mishra
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Ashish Kumar
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Vijaya R Dirisala
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur District, AP-522213, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan 342011, India
| | - Surajit Das
- Laboratory of Environmental Microbiology and Ecology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Ramandeep Singh
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, PO Box # 4, Faridabad, Haryana 121001, India
| | - Assirbad Behura
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India.
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India.
| |
Collapse
|
19
|
da Silva FF, Paz JD, Rambo RS, Gonçalves GA, Muniz MN, de Matos Czeczot A, Perelló MA, Berger A, González LC, Duarte LS, da Silva AB, Ferreira CAS, de Oliveira SD, Moura S, Bizarro CV, Basso LA, Machado P. Unveiling the Antimycobacterial Potential of Novel 4-Alkoxyquinolines: Insights into Selectivity, Mechanism of Action, and In Vivo Exposure. J Med Chem 2024; 67:21781-21794. [PMID: 39630172 DOI: 10.1021/acs.jmedchem.4c01302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
This work presents a comprehensive investigation into the design, synthesis, and evaluation of a novel series of 4-alkoxyquinolines as potential antimycobacterial agents. The design approach, which combined molecular simplification and chain extension, resulted in compounds with potent and selective activity against both drug-susceptible and multidrug-resistant Mycobacterium tuberculosis strains. The lead molecule, targeting the cytochrome bc1 complex, exhibited favorable kinetic solubility and remarkable chemical stability under acidic conditions. Despite in vitro ADME evaluations showing low permeability and high metabolism in rat microsomes, the lead compound exhibited bacteriostatic activity in a murine macrophage model of TB infection and demonstrated promising in vivo exposure following gavage in mice, with an AUC0-t of 127.5 ± 5.7 μM h. To the best of our knowledge, for the first time, a simplified structure from 2-(quinolin-4-yloxy)acetamides has shown such potential. These findings suggest a new avenue for exploring this chemical class as a source of antituberculosis drug candidates.
Collapse
Affiliation(s)
- Fernanda Fries da Silva
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Josiane Delgado Paz
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Raoní Scheibler Rambo
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Guilherme Arraché Gonçalves
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Mauro Neves Muniz
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Alexia de Matos Czeczot
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Marcia Alberton Perelló
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Andresa Berger
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Laura Calle González
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Lovaine Silva Duarte
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Anelise Baptista da Silva
- Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Laboratório de Imunologia e Microbiologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Carlos Alexandre Sanchez Ferreira
- Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Laboratório de Imunologia e Microbiologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Sílvia Dias de Oliveira
- Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Laboratório de Imunologia e Microbiologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Sidnei Moura
- Laboratório de Biotecnologia de Produtos Naturais e Sintéticos, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Rio Grande do Sul 95070-560, Brazil
| | - Cristiano Valim Bizarro
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Luiz Augusto Basso
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| | - Pablo Machado
- Instituto Nacional de Ciência e Tecnologia em Tuberculose, Centro de Pesquisas em Biologia Molecular e Funcional, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90616-900, Brazil
| |
Collapse
|
20
|
Wang Z, Shen W, Li Y, Wang X, Zhong X, Wang X. Multi-omics Analysis of Klebsiella pneumoniae Revealed Opposing Effects of Rutin and Luteolin on Strain Growth. Curr Microbiol 2024; 82:9. [PMID: 39585437 DOI: 10.1007/s00284-024-03982-5] [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: 04/23/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
The emergence of pathogenic bacteria resistant to conventional antibiotics is becoming increasingly challenging. Plant-derived flavonoids are potential alternatives to antibiotics, owing to their antimicrobial properties. However, the molecular mechanisms through which they inhibit the growth of pathogenic microorganisms remain unclear. Therefore, Klebsiella pneumoniae ATCC700603 was separately incubated in two flavonoids to elucidate their inhibitory mechanism. Metabolomic and transcriptomic analyses were performed after 4-h incubation. In total, 5483 genes and 882 metabolites were identified. Compared to the untreated control, rutin and luteolin activated 507 and 374 differentially expressed genes (DEGs), respectively. However, the number of differential abundant metabolites (DAMs) remained the same. The top 10 correlated DEGs and DAMs were identified within each comparative group after a correlation analysis. Rutin induced the accumulation of unique metabolites and suppressed gene expression whereas luteolin did not. Our results explain the disparate effects of these two flavonoids and demonstrate the inhibitory mechanism of rutin on strain growth.
Collapse
Affiliation(s)
- Zhibin Wang
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wanxia Shen
- Citrus Research Institute, Southwest University, Beibei, Chongqing, 400715, China
| | - Yuejiao Li
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaoyun Wang
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaolin Zhong
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xing Wang
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
21
|
Aono T, Tamura S, Suzuki Y, Imanara T, Niwa R, Yamane Y, Kobayashi T, Kikuyama S, Hasunuma I, Iwamuro S. Cloning and Functional Analysis of Skin Host Defense Peptides from Yakushima Tago's Brown Frog ( Rana tagoi yakushimensis) and Development of Serum Endotoxin Detection System. Antibiotics (Basel) 2024; 13:1127. [PMID: 39766517 PMCID: PMC11672578 DOI: 10.3390/antibiotics13121127] [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: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objective: Amphibian skin is a valuable source of host defense peptides (HDPs). This study aimed to identify HDPs with novel amino acid sequences from the skin of Rana tagoi yakushimensis and analyze their functions. Methods: cDNAs encoding HDP precursors were cloned and sequenced using RT-PCR and 3'-RACE. The novel HDPs were synthesized to evaluate their antimicrobial activity, antioxidant activity, and cytotoxicity. Antimicrobial activity was evaluated by way of broth microdilution and endotoxin- and β-glucan-binding capacity using an enzyme-linked endotoxin binding assay (ELEBA) and a modified ELEBA, respectively. Results: Nine cDNAs encoding precursors for various HDP families, including temporin, ranatuerin-2, brevinin-1, amurin-9, and a novel yakushimin peptide, were identified. Brevinin-1TYa exhibited antibacterial activity against Staphylococcus aureus, and brevinin-1TYa and amurin-9TYa induced morphological changes in Escherichia coli and S. aureus. Yakushimin-TYa, amurin-9TYa, and brevinin-1TYa showed concentration-dependent antibacterial effects against the plant pathogens Xanthomonas oryzae pv. oryzae and Clavibacter michiganensis subsp. michiganensis. Amurin-9TYa demonstrated strong binding affinity to lipopolysaccharide, lipoteichoic acid, and β-glucan, exhibited antioxidant activity, and lacked cytotoxicity, making it a promising therapeutic candidate. Moreover, brevinin-1TYa showed strong cytotoxicity, whereas yakushimin-TYa exhibited weak cytotoxicity. Conclusions: These findings highlight the potential of these peptides, particularly amurin-9TYa, for future applications as antimicrobial and therapeutic agents.
Collapse
Affiliation(s)
- Taichi Aono
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi-shi 274-8510, Chiba, Japan
| | - Saki Tamura
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi-shi 274-8510, Chiba, Japan
| | - Yua Suzuki
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi-shi 274-8510, Chiba, Japan
| | - Taichi Imanara
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi-shi 274-8510, Chiba, Japan
| | - Ryosei Niwa
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi-shi 274-8510, Chiba, Japan
| | - Yoshie Yamane
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi-shi 274-8510, Chiba, Japan
| | - Tetsuya Kobayashi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-okubo, Sakura-ku, Saitama-shi 338-8570, Saitama, Japan
| | - Sakae Kikuyama
- Department of Biology, Faculty of Education and Integrated Arts and Sciences, Center for Advanced Biomedical Sciences, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku 162-8480, Tokyo, Japan
| | - Itaru Hasunuma
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi-shi 274-8510, Chiba, Japan
| | - Shawichi Iwamuro
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi-shi 274-8510, Chiba, Japan
| |
Collapse
|
22
|
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
|
23
|
Tóth ZS, Leveles I, Nyíri K, Nagy GN, Harmat V, Jaroentomeechai T, Ozohanics O, Miller RL, Álvarez MB, Vértessy BG, Benedek A. The homodimerization domain of the Stl repressor is crucial for efficient inhibition of mycobacterial dUTPase. Sci Rep 2024; 14:27171. [PMID: 39511242 PMCID: PMC11544220 DOI: 10.1038/s41598-024-76349-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
The dUTPase is a key DNA repair enzyme in Mycobacterium tuberculosis, and it may serve as a novel promising anti-tuberculosis target. Stl repressor from Staphylococcus aureus was shown to bind to and inhibit dUTPases from various sources, and its expression in mycobacterial cells interfered with cell growth. To fine-tune and optimize Stl-induced inhibition of mycobacterial dUTPase, we aimed to decipher the molecular details of this interaction. Structural background of the complex between dUTPase and a truncated Stl lacking the repressor C-terminal homodimerization domain has been described, however, the effects of this truncation of Stl on enzyme binding and inhibition are still not known. Using several independent biophysical, structural and enzyme kinetic methods, here we show that lack of the repressor homodimerization domain strongly perturbs both enzyme binding and inhibition. We also investigated the role of a mycobacteria-specific loop in the Stl-interaction. Our results show that removal of this loop leads to a ten-fold increase in the apparent inhibition constant of Stl. We present a high-resolution three-dimensional structure of mycobacterial dUTPase lacking the genus-specific loop for structural insight. Our present data suggest that potent inhibition of mycobacterial dUTPase by Stl requires the wild-type full-length protein context.
Collapse
Affiliation(s)
- Zoé S Tóth
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary.
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary.
| | - Ibolya Leveles
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Kinga Nyíri
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Gergely N Nagy
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Veronika Harmat
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, ELTE Eötvös Loránd University, Budapest, Hungary
- HUN-REN-ELTE Protein Modelling Research Group, Hungarian Research Network, Budapest, Hungary
| | - Thapakorn Jaroentomeechai
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Oliver Ozohanics
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Rebecca L Miller
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Marina Ballesteros Álvarez
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Beáta G Vértessy
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary.
| | - András Benedek
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
- Department of Applied Biotechnology and Food Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary.
| |
Collapse
|
24
|
Qin L, Xu J, Chen J, Wang S, Zheng R, Cui Z, Liu Z, Wu X, Wang J, Huang X, Wang Z, Wang M, Pan R, Kaufmann SHE, Meng X, Zhang L, Sha W, Liu H. Cell-autonomous targeting of arabinogalactan by host immune factors inhibits mycobacterial growth. eLife 2024; 13:RP92737. [PMID: 39495223 PMCID: PMC11534329 DOI: 10.7554/elife.92737] [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] [Indexed: 11/05/2024] Open
Abstract
Deeper understanding of the crosstalk between host cells and Mycobacterium tuberculosis (Mtb) provides crucial guidelines for the rational design of novel intervention strategies against tuberculosis (TB). Mycobacteria possess a unique complex cell wall with arabinogalactan (AG) as a critical component. AG has been identified as a virulence factor of Mtb which is recognized by host galectin-9. Here, we demonstrate that galectin-9 directly inhibited mycobacterial growth through AG-binding property of carbohydrate-recognition domain 2. Furthermore, IgG antibodies with AG specificity were detected in the serum of TB patients. Based on the interaction between galectin-9 and AG, we developed a monoclonal antibody (mAb) screening assay and identified AG-specific mAbs which profoundly inhibit Mtb growth. Mechanistically, proteomic profiling and morphological characterizations revealed that AG-specific mAbs regulate AG biosynthesis, thereby inducing cell wall swelling. Thus, direct AG-binding by galectin-9 or antibodies contributes to protection against TB. Our findings pave the way for the rational design of novel immunotherapeutic strategies for TB control.
Collapse
Affiliation(s)
- Lianhua Qin
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Junfang Xu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Jianxia Chen
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Sen Wang
- Department of Infectious Diseases, National Medical Centre for Infectious Diseases, National Clinical Research Centre for Aging and Medicine, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Ruijuan Zheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Zhenling Cui
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Zhonghua Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiangyang Wu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Jie Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiaochen Huang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | | | | | | | - Stefan HE Kaufmann
- Max Planck Institute for Infection BiologyBerlinGermany
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Hagler Institute for Advanced Study, Texas A&M UniversityCollege StationUnited States
| | - Xun Meng
- Abmart IncShanghaiChina
- Multitude TherapeuticsShanghaiChina
| | - Lu Zhang
- School of Life Science, Fudan UniversityShanghaiChina
| | - Wei Sha
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
- Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Haipeng Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| |
Collapse
|
25
|
Zhou CY, Yang YL, Han ZY, Chen YX, Liu HL, Fan K, Li MC, Tu SH, Wen Q, Zhou XY, Ma L. Peripheral blood MR1 tetramer-positive mucosal-associated invariant T-cell function is modulated by mammalian target of rapamycin complex 1 in patients with active tuberculosis. Immunology 2024; 173:497-510. [PMID: 39022997 DOI: 10.1111/imm.13834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Tuberculosis (TB) is still an urgent global public health problem. Notably, mucosal-associated invariant T (MAIT) cells play an important role in early anti-TB immune response. Targeted control of them may be an effective method to improve vaccine efficacy and TB treatment. However, the biology and signal regulation mechanisms of MAIT cells in TB patients are still poorly understood. Previous studies have been limited by the lack of reagents to specifically identify MAIT cells. In addition, the use of alternative markers may subsume non-MAIT cell into MAIT cell populations. In this study, the human MR1 tetramer which can specifically identify MAIT cells was used to further explore the effect and mechanism of MAIT cells in anti-TB immune response. Our results showed that the tetramer+ MAIT cells in peripheral blood of TB patients were mainly CD8+ or CD4-CD8- cells, and very few were CD4+ cells. After BCG infecting autologous antigen-presenting cells, MAIT cells in patients produced significantly higher levels of cytokines, lysis and proliferation compared with healthy controls. After suppression of mTORC1 by the mTORC1-specific inhibitor rapamycin, the immune response of MAIT cells in patients was significantly reduced. This study demonstrates that peripheral blood tetramer+ MAIT cells from TB patients have significant anti-TB immune effect, which is regulated by mTORC1. This could provide ideas and potential therapeutic targets for the development of novel anti-TB immunotherapy.
Collapse
Affiliation(s)
- Chao-Ying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Ya-Long Yang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Zhen-Yu Han
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Yao-Xin Chen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Hong-Lin Liu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Ke Fan
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Ming-Chong Li
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Si-Hang Tu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Qian Wen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Xin-Ying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Dhivya LS, Manoharadas S, Pandiaraj S, Thiruvengadam M, Viswanathan D, Govindasamy R. Halogenated chalcones against Mycobacterium tuberculosis targeting InhA: Rational design, in silico and in vitro evaluation. Microb Pathog 2024; 196:106945. [PMID: 39284520 DOI: 10.1016/j.micpath.2024.106945] [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: 09/02/2023] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024]
Abstract
A library of 25-series compounds was designed against Mycobacterium Tuberculosis (M.tb) to identify novel antitubercular drugs. In silico inhibition of InhA, an essential component of FAS-II, was successfully achieved. The drug ability, lead-likeness, and toxicity of the compounds were assessed using Swiss ADME, pkCSM, and Osiris Property Explorer, which revealed the potential for drug development of chalcone compounds. Through in silico research, it was confirmed that toxic-free compounds could bind to InhA. It was found that all of the compounds bind to InhA with binding affinities ranging from -7.78 to -10.29 kcal/mol-1 which is higher than the reference standard Isoniazid and Pyrazinamide. The top five compounds were synthesized from 15 toxic-free compounds. The structural characteristics of the compounds were determined using IR, NMR, and mass spectrometry techniques. These findings indicate that these substances are competitive, reversible, and specific InhA inhibitors of InhA. using the Alamar Blue assay method (H37RV, ATCC No. 27294), the in vitro anti-mycobacterial activity of each of the synthesized compounds against M.tb was evaluated. The two most powerful compounds were (2E)-3-[4-(benzyloxy)-3,5-dimethylphenyl] and (2E)-1-(3,5-dibromophenyl)-3-(3-phenoxyphenyl) prop-2-en-1-one. In the MABA Assay, the MIC for 1-(3,5-dibromophenyl) prop-2-en-1-one was 6.25 μg/ml.
Collapse
Affiliation(s)
- L S Dhivya
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Dr MGR Educational and Research Institute, Velappanchavadi, Chennai, 77, Tamil Nadu, India
| | - Salim Manoharadas
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2454, Riyadh, Saudi Arabia
| | - Saravanan Pandiaraj
- Department of Self-Development Skills, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, 05029, South Korea
| | - Dhivya Viswanathan
- Department of Orthodontics, Saveetha Dental College, and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamilnadu, India
| | - Rajakumar Govindasamy
- Department of Orthodontics, Saveetha Dental College, and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamilnadu, India.
| |
Collapse
|
27
|
Wotale TW, Lelisho ME, Negasa BW, Tareke SA, Gobena WE, Amesa EG. Identifying risk factors for recurrent multidrug resistant tuberculosis based on patient's record data from 2016 to 2021: retrospective study. Sci Rep 2024; 14:23912. [PMID: 39397064 PMCID: PMC11471762 DOI: 10.1038/s41598-024-73209-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Globally, the prevalence of multidrug-resistant tuberculosis (MDR-TB) has been increasing recently. This is a major public health concern, as MDR-TB is more difficult to treat and has poorer outcomes compared to drug-sensitive tuberculosis. The main objective of the study was to identify risk factors for recurrent multidrug-resistant tuberculosis, at Alert Specialized Hospital, Addis Ababa, by using different parametric shared frailty models. From January 2016 to December 2021, a retrospective study was conducted on MDR-TB patients at Alert Specialized Hospital in Addis Ababa. The data for the study were collected from the medical records of MDR-TB patients at the hospital during this time period. Gamma and inverse-Gaussian shared frailty models were used to analyze the dataset, with the exponential, Weibull, and lognormal distributions included as baseline hazard functions. The data were analyzed using R statistical software. The median recurrence time of the patients was 12 months, and 149 (34.3%) had recurrences. The clustering effect was statistically significant for multiple drug-resistant tuberculosis patients' recurrence. According to the Weibull-Inverse-Gaussian model, factors that reduced time to MDR-TB recurrence included lower weight (ɸ = 0.944), smoking (ɸ = 0.045), alcohol use (ɸ = 0.631), hemoptysis (ɸ = 0.041), pneumonia (ɸ = 0.564), previous anti-TB treatment (ɸ = 0.106), rural residence (ɸ = 0.163), and chronic diseases like diabetes (ɸ = 0.442) were associated with faster recurrence. While, higher education (ɸ = 3.525) and age (ɸ = 1.021) extended time to recurrence. For weight increment, smokers and alcohol users, clinical complications of hemoptysis and pneumonia, patients with pulmonary disease who had a history of previous anti-TB treatment, and being rural residents are prognostic factors. There was a significant clustering effect at the Alert Specialized Hospital in Addis Ababa, Ethiopia. The Weibull-Inverse Gaussian Shared Frailty Model was chosen as the best model for predicting the time to recurrence of MDR-TB.
Collapse
Affiliation(s)
- Teramaj Wongel Wotale
- Department of Statistics, College of Natural and Computational Sciences, Dilla University, Dilla, Ethiopia.
- Department of Statistics, College of Natural and Computational Sciences, Mattu University, Mattu, Ethiopia.
| | - Mesfin Esayas Lelisho
- Department of Statistics, College of Natural and Computational Sciences, Mizan-Tepi University, Tepi, Ethiopia.
| | - Bikiltu Wakuma Negasa
- Department of Statistics, College of Natural and Computational Sciences, Mattu University, Mattu, Ethiopia
| | - Seid Ali Tareke
- Department of Statistics, College of Natural and Computational Sciences, Mizan-Tepi University, Tepi, Ethiopia
| | - Woldemariam Erkalo Gobena
- Department of Statistics, College of Natural and Computational Sciences, Mattu University, Mattu, Ethiopia
| | - Ebsa Gelan Amesa
- Department of Statistics, College of Natural and Computational Sciences, Mattu University, Mattu, Ethiopia
| |
Collapse
|
28
|
Yang J, Ma Y, Yu J, Liu Y, Xia J, Kong X, Jin X, Li J, Lin S, Ruan Y, Yang F, Pi J. Advancing Roles and Therapeutic Potentials of Pyroptosis in Host Immune Defenses against Tuberculosis. Biomolecules 2024; 14:1255. [PMID: 39456188 PMCID: PMC11505957 DOI: 10.3390/biom14101255] [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: 07/25/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (Mtb) infection, remains a deadly global public health burden. The use of recommended drug combinations in clinic has seen an increasing prevalence of drug-resistant TB, adding to the impediments to global control of TB. Therefore, control of TB and drug-resistant TB has become one of the most pressing issues in global public health, which urges the exploration of potential therapeutic targets in TB and drug-resistant TB. Pyroptosis, a form of programmed cell death characterized by cell swelling and rupture, release of cellular contents and inflammatory responses, has been found to promote pathogen clearance and adopt crucial roles in the control of bacterial infections. It has been demonstrated that Mtb can cause host cell pyroptosis, and these host cells, which are infected by Mtb, can kill Mtb accompanied by pyroptosis, while, at the same time, pyroptosis can also release intracellular Mtb, which may potentially worsen the infection by exacerbating the inflammation. Here, we describe the main pathways of pyroptosis during Mtb infection and summarize the identified effectors of Mtb that regulate pyroptosis to achieve immune evasion. Moreover, we also discuss the potentials of pyroptosis to serve as an anti-TB therapeutic target, with the aim of providing new ideas for the development of TB treatments.
Collapse
Affiliation(s)
- Jiayi Yang
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Yuhe Ma
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiaqi Yu
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Yilin Liu
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiaojiao Xia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China;
| | - Xinen Kong
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Xiaoying Jin
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiaxiang Li
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Siqi Lin
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Yongdui Ruan
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Fen Yang
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiang Pi
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| |
Collapse
|
29
|
Chauhan M, Barot R, Yadav R, Joshi K, Mirza S, Chikhale R, Srivastava VK, Yadav MR, Murumkar PR. The Mycobacterium tuberculosis Cell Wall: An Alluring Drug Target for Developing Newer Anti-TB Drugs-A Perspective. Chem Biol Drug Des 2024; 104:e14612. [PMID: 39237482 DOI: 10.1111/cbdd.14612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/26/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024]
Abstract
The Mycobacterium cell wall is a capsule-like structure comprising of various layers of biomolecules such as mycolic acid, peptidoglycans, and arabinogalactans, which provide the Mycobacteria a sort of cellular shield. Drugs like isoniazid, ethambutol, cycloserine, delamanid, and pretomanid inhibit cell wall synthesis by inhibiting one or the other enzymes involved in cell wall synthesis. Many enzymes present across these layers serve as potential targets for the design and development of newer anti-TB drugs. Some of these targets are currently being exploited as the most druggable targets like DprE1, InhA, and MmpL3. Many of the anti-TB agents present in clinical trials inhibit cell wall synthesis. The present article covers a systematic perspective of developing cell wall inhibitors targeting various enzymes involved in cell wall biosynthesis as potential drug candidates for treating Mtb infection.
Collapse
Affiliation(s)
- Monica Chauhan
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Rahul Barot
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Rasana Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Karan Joshi
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Sadaf Mirza
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Rupesh Chikhale
- The Cambridge Crystallography Data Center, Cambridge, UK
- School of Pharmacy, University College London, London, UK
| | | | - Mange Ram Yadav
- Centre of Research for Development, Parul University, Vadodara, Gujarat, India
| | - Prashant R Murumkar
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| |
Collapse
|
30
|
Chatterjee C, Mohan GR, Chinnasamy HV, Biswas B, Sundaram V, Srivastava A, Matheshwaran S. Anti-mutagenic agent targeting LexA to combat antimicrobial resistance in mycobacteria. J Biol Chem 2024; 300:107650. [PMID: 39122002 PMCID: PMC11408154 DOI: 10.1016/j.jbc.2024.107650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Antimicrobial resistance (AMR) is a serious global threat demanding innovations for effective control of pathogens. The bacterial SOS response, regulated by the master regulators, LexA and RecA, contributes to AMR through advantageous mutations. Targeting the LexA/RecA system with a novel inhibitor could suppress the SOS response and potentially reduce the occurrence of AMR. RecA presents a challenge as a therapeutic target due to its conserved structure and function across species, including humans. Conversely, LexA which is absent in eukaryotes, can be potentially targeted, due to its involvement in SOS response which is majorly responsible for adaptive mutagenesis and AMR. Our studies combining bioinformatic, biochemical, biophysical, molecular, and cell-based assays present a unique inhibitor of mycobacterial LexA, wherein we show that the inhibitor interacts directly with the catalytic site residues of LexA of Mycobacterium tuberculosis (Mtb), consequently hindering its cleavage, suppressing SOS response thereby reducing mutation frequency and AMR.
Collapse
Affiliation(s)
- Chitral Chatterjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Gokul Raj Mohan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Hariharan V Chinnasamy
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Bhumika Biswas
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Vidya Sundaram
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Gujarat, India
| | - Ashutosh Srivastava
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, Gujarat, India
| | - Saravanan Matheshwaran
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India; Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India; Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology, Kanpur, Uttar Pradesh, India; Kotak School of Sustainability, Indian Institute of Technology, Kanpur, Uttar Pradesh, India.
| |
Collapse
|
31
|
Xu JT, Lin Y, Cheng T, Deng JY. The rv2820c K114N mutation is related with capreomycin tolerance. Tuberculosis (Edinb) 2024; 148:102551. [PMID: 39084000 DOI: 10.1016/j.tube.2024.102551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/17/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
As one of the factors affecting the treatment outcomes, drug tolerance in mycobacteriosis has not been paid due attention. Genome-wide association studies on 607 Mycobacterium tuberculosis clinical isolates with phenotypic drug susceptibility test data revealed that a K114N mutation on the rv2820c gene was highly enriched in capreomycin-resistant isolates (32/213, 15.02%). However, the mutation was also observed in capreomycin-sensitive isolates (10/394, 2.53%). In most cases (31/42, 73.81%), the rv2820c K114N mutation occurred in isolates with the known capreomycin resistance conferring mutation rrs A1401G. In contrast, the general frequency of the rv2820c K114N mutation was low in 7061 genomes downloaded from the National Center for Biotechnology Information database. To determine the impact of this mutation on the antimycobacterial activity of capreomycin, the intact rv2820c gene and the rv2820c K114N mutant were over-expressed in Mycobacterium smegmatis (Ms), and the results of susceptibility tests showed that the rv2820c K114N mutation did not affect the minimum inhibition concentration (MIC) of capreomycin. Subsequently, the data of time-kill assays showed that, it took only 2 h of capreomycin treatment (40 μg/ml, 5 × MIC) to kill 99.9% bacterial cells of Ms MC2155 pMV261::rv2820cH37Rv, while it took 6 h to achieve that for Ms MC2155 pMV261::rv2820cK114N. Taken together, these data suggested that the rv2820c K114N mutation is related with capreomycin tolerance, which merits further investigation.
Collapse
Affiliation(s)
- Jin-Tian Xu
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi Lin
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tao Cheng
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiao-Yu Deng
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
32
|
Nguyen DTC, Nguyen NTT, Nguyen TTT, Tran TV. Recent advances in the biosynthesis of ZnO nanoparticles using floral waste extract for water treatment, agriculture and biomedical engineering. NANOSCALE ADVANCES 2024; 6:4047-4061. [PMID: 39114141 PMCID: PMC11302053 DOI: 10.1039/d4na00133h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/29/2024] [Indexed: 08/10/2024]
Abstract
Flowers are often discarded after cultural and religious events, making it worthwhile to explore the utilization of this floral waste for material production. Floral extracts contain a diverse array of phytochemicals such as polyphenols, flavonoids, and reducing sugars, which play a significant role in the formation and influencing the properties of zinc oxide (ZnO) nanoparticles. In this review, we delve into the importance of floral extract, methodology, mechanism, and influencing factors in the production of ZnO nanoparticles. Additionally, the role of green ZnO nanoparticles as an adsorbent and photocatalyst for water treatment is discussed. These floral extract-mediated ZnO nanoparticles exhibit advantages in agricultural and biomedical applications, including promoting seed germination and demonstrating antibacterial, anticancer, and antifungal properties. Cost analysis reveals that while various expenses are associated with ZnO production, scaling up processes can help reduce these costs. This review underscores the potential of floral waste extract for the synthesis of green ZnO nanoparticles, thereby contributing to waste-to-wealth strategies and adhering to green chemistry principles.
Collapse
Affiliation(s)
- Duyen Thi Cam Nguyen
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University 298-300A Nguyen Tat Thanh, District 4 Ho Chi Minh City 755414 Vietnam
| | - Ngoan Thi Thao Nguyen
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University 298-300A Nguyen Tat Thanh, District 4 Ho Chi Minh City 755414 Vietnam
- Nong Lam University - Ho Chi Minh City Ho Chi Minh City 700000 Vietnam
| | | | - Thuan Van Tran
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University 298-300A Nguyen Tat Thanh, District 4 Ho Chi Minh City 755414 Vietnam
| |
Collapse
|
33
|
Kangongwe MH, Mwanza W, Mwamba M, Mwenya J, Muzyamba J, Mzyece J, Hamukale A, Tembo E, Nsama D, Chimzizi R, Mubanga A, Tambatamba B, Mudenda S, Lishimpi K. Drug resistance profiles of Mycobacterium tuberculosis clinical isolates by genotype MTBDRplus line probe assay in Zambia: findings and implications. JAC Antimicrob Resist 2024; 6:dlae122. [PMID: 39055721 PMCID: PMC11271804 DOI: 10.1093/jacamr/dlae122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Background The emergence of drug resistance is a threat to global tuberculosis (TB) elimination goals. This study investigated the drug resistance profiles of Mycobacterium tuberculosis (M. tuberculosis) using the Genotype MTBDRplus Line Probe Assay at the National Tuberculosis Reference Laboratory (NTRL) in Zambia. Methods A cross-sectional study was conducted between January 2019 and December 2020. GenoType MTBDRplus line probe assay records for patients at the NTRL were reviewed to investigate drug susceptibility profiles of M. tuberculosis isolates to rifampicin and isoniazid. Data analysis was done using Stata version 16.1. Results Of the 241 patient records reviewed, 77% were for females. Overall, 44% of patients were newly diagnosed with TB, 29% had TB relapse, 10% treatment after failure and 8.3% treatment after loss to follow-up. This study found that 65% of M. tuberculosis isolates were susceptible to rifampicin and isoniazid. Consequently, 35% of the isolates were resistant to rifampicin and/or isoniazid and 21.2% were multidrug-resistant (MDR). Treatment after failure [relative risk ratios (RRR) = 6.1, 95% CI: 1.691-22.011] and treatment after loss to follow-up (RRR = 7.115, 95% CI: 1.995-25.378) were significantly associated with MDR-TB. Unknown HIV status was significantly associated with isoniazid mono-resistance (RRR = 5.449, 95% CI: 1.054-28.184). Conclusions This study found that 65% of M. tuberculosis isolates were susceptible to rifampicin and isoniazid while 35% were resistant. Consequently, a high prevalence of MDR-TB is of public health concern. There is a need to heighten laboratory surveillance and early detection of drug-resistant TB to prevent the associated morbidity and mortality.
Collapse
Affiliation(s)
- Mundia Hendrix Kangongwe
- Ministry of Health, Chest Diseases Laboratory, Lusaka, Zambia
- Institute for Basic and Biomedical Sciences, Levy Mwanawasa Medical University, Lusaka, Zambia
| | - Winnie Mwanza
- Ministry of Health, National Tuberculosis and Leprosy Programme, Lusaka, Zambia
- Public Health, USAID-STAR Project
| | - Mutende Mwamba
- Ministry of Health, Chest Diseases Laboratory, Lusaka, Zambia
| | - Jonathan Mwenya
- Ministry of Health, Chest Diseases Laboratory, Lusaka, Zambia
| | - John Muzyamba
- Ministry of Health, Chest Diseases Laboratory, Lusaka, Zambia
| | - Judith Mzyece
- Ministry of Health, Laboratory and Pathological Services, Lusaka, Zambia
| | - Amos Hamukale
- Epidemiology and Surveillance, Zambia National Public Health Institute, Lusaka, Zambia
| | - Emmanuel Tembo
- Ministry of Health, National Tuberculosis and Leprosy Programme, Lusaka, Zambia
| | - Davy Nsama
- Ministry of Health, Laboratory and Pathological Services, Lusaka, Zambia
| | - Rehab Chimzizi
- Ministry of Health, National Tuberculosis and Leprosy Programme, Lusaka, Zambia
- Public Health, USAID-STAR Project
| | - Angel Mubanga
- Ministry of Health, National Tuberculosis and Leprosy Programme, Lusaka, Zambia
| | | | - Steward Mudenda
- Department of Pharmacy, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | - Kennedy Lishimpi
- Technical Services, Ministry of Health Headquarters, Lusaka, Zambia
| |
Collapse
|
34
|
Banerjee A, Sharma A, Kamble P, Garg P. Prediction of Mycobacterium tuberculosis cell wall permeability using machine learning methods. Mol Divers 2024; 28:2317-2329. [PMID: 39133353 DOI: 10.1007/s11030-024-10952-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024]
Abstract
Tuberculosis (TB) caused by the bacteria Mycobacterium tuberculosis (M. tb), continues to pose a significant worldwide health threat. The advent of drug-resistant strains of the disease highlights the critical need for novel treatments. The unique cell wall of M. tb provides an extra layer of protection for the bacteria and hence only compounds that can penetrate this barrier can reach their targets within the bacterial cell wall. The creation of a reliable machine learning (ML) model to predict the mycobacterial cell wall permeability of small molecules is presented in this work and four ML algorithms, including Random Forest, Support Vector Machines (SVM), k-nearest Neighbour (k-NN) and Logistic Regression were trained on a dataset of 5368 compounds. RDKit and Mordred toolkits were used to calculate features. To determine the most effective model, various performance metrics were used such as accuracy, precision, recall, F1 score, and area under the receiver operating characteristic curve. The best-performing model was further refined with hyperparameter tuning and tenfold cross-validation. The SVM model with filtering outperformed the other machine learning models and demonstrated 80.26% and 81.13% accuracy on the test and validation datasets, respectively. The study also provided insights into the molecular descriptors that play the most important role in predicting the ability of a molecule to pass the M. tb cell wall, which could guide future compound design. The model is available at https://github.com/PGlab-NIPER/MTB_Permeability .
Collapse
Affiliation(s)
- Aritra Banerjee
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab, 160 062, India
| | - Anju Sharma
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab, 160 062, India
| | - Pradnya Kamble
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab, 160 062, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab, 160 062, India.
| |
Collapse
|
35
|
Ruiz-Santana S, Dearriba-Reyes J, Saavedra P, Iglesias-Llorente L, Alonso-Acero L, Hernández-Socorro CR, Sánchez-Ramírez C. Prediction of Concomitant Nosocomial Infection in Patients Previously Colonized Colorectally by Multidrug-Resistant Bacteria in an SDD Setting. Antibiotics (Basel) 2024; 13:717. [PMID: 39200017 PMCID: PMC11350885 DOI: 10.3390/antibiotics13080717] [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: 06/21/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Antibiotic resistance is a worldwide concern. This study retrospectively analyzed patients admitted to the ICU of a tertiary hospital over a period of 7 months who were rectally colonized by multidrug-resistant microorganisms. The incidence of concomitant nosocomial infections was estimated, thus providing the risk of a colonizing microorganism producing a nosocomial infection. METHODS Infections with the same microorganism (concomitant) or different microorganisms (non-concomitant) were analyzed in order to adjust the empirical antibiotic treatment. Patients with rectal colonization by at least one multidrug-resistant bacterium (MDRB) on admission or after ICU admission were included. All patients had complete selective digestive decontamination (SDD) prophylaxis. For univariate analysis, categorical variables are expressed as frequencies and percentages and continuous variables as means and standard deviations, or as medians and interquartile ranges. For multivariate analysis, the model is summarized with p-values and hazard ratios with 95% confidence intervals. Survival analysis was conducted using the Kaplan-Meier method, which was performed to evaluate the time elapsed from colonization to infection by the same bacteria. Statistical significance was considered at p < 0.05. RESULTS Of the 130 patients with MDRB bacterial colonization analyzed, 98 remained free of infection, while 22 developed non-concomitant infections and 10 had infections concomitant to rectal colonizing bacteria. OXA-48-producing bacteria and MDR-Pseudomonas spp. incidences were 18.9% (95% CI: 7.96-35.2) and 44.4% (CI: 13.7-78.8), respectively. CONCLUSIONS OXA-48-producing bacteria and MDR-Pseudomonas spp. were the only bacteria associated with the development of infections concomitant to rectal colonization in an SDD setting. The incidence of MDRB infections was low.
Collapse
Affiliation(s)
- Sergio Ruiz-Santana
- Intensive Care Unit, Hospital Universitario de Gran Canaria Dr. Negrín, University de Las Palmas de Gran Canaria, E-35010 Las Palmas de Gran Canaria, Spain;
| | - José Dearriba-Reyes
- Department of Medical and Surgical Sciences, University de Las Palmas de Gran Canaria, E-35010 Las Palmas de Gran Canaria, Spain;
| | - Pedro Saavedra
- Department of Mathematics, University de Las Palmas de Gran Canaria, E-35010 Las Palmas de Gran Canaria, Spain;
| | - Laura Iglesias-Llorente
- Department of Microbiology, Hospital Universitario de Gran Canaria Dr. Negrín, E-35010 Las Palmas de Gran Canaria, Spain; (L.I.-L.); (L.A.-A.)
| | - Laura Alonso-Acero
- Department of Microbiology, Hospital Universitario de Gran Canaria Dr. Negrín, E-35010 Las Palmas de Gran Canaria, Spain; (L.I.-L.); (L.A.-A.)
| | - Carmen-Rosa Hernández-Socorro
- Department of Radiology, Hospital Universitario de Gran Canaria Dr. Negrín, University de Las Palmas de Gran Canaria, E-35010 Las Palmas de Gran Canaria, Spain;
| | - Catalina Sánchez-Ramírez
- Intensive Care Unit, Hospital Universitario de Gran Canaria Dr. Negrín, University de Las Palmas de Gran Canaria, E-35010 Las Palmas de Gran Canaria, Spain;
| |
Collapse
|
36
|
Bague D, Wang R, Hodge D, Mikati MO, Roma JS, Boshoff HI, Dailey AL, Girma M, Couch RD, Odom John AR, Dowd CS. Inhibition of DXR in the MEP pathway with lipophilic N-alkoxyaryl FR900098 analogs. RSC Med Chem 2024; 15:2422-2439. [PMID: 39026652 PMCID: PMC11253873 DOI: 10.1039/d3md00642e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/22/2024] [Indexed: 07/20/2024] Open
Abstract
In Mycobacterium tuberculosis (Mtb) and Plasmodium falciparum (Pf), the methylerythritol phosphate (MEP) pathway is responsible for isoprene synthesis. This pathway and its products are vital to bacterial/parasitic metabolism and survival, and represent an attractive set of drug targets due to their essentiality in these pathogens but absence in humans. The second step in the MEP pathway is the conversion of 1-deoxy-d-xylulose-5-phosphate (DXP) to MEP and is catalyzed by 1-deoxy-d-xylulose-5-phosphate reductoisomerase (DXR). Natural products fosmidomycin and FR900098 inhibit DXR, but are too polar to reach the desired target inside some cells, such as Mtb. Synthesized FR900098 analogs with lipophilic substitution in the position α to the phosphorous atom showed promise, resulting in increased activity against Mtb and Pf. Here, an α substitution, consisting of a 3,4-dichlorophenyl substituent, in combination with various O-linked alkylaryl substituents on the hydroxamate moiety is utilized in the synthesis of a novel series of FR900098 analogs. The purpose of the O-linked alkylaryl substituents is to further enhance DXR inhibition by extending the structure into the adjacent NADPH binding pocket, blocking the binding of both DXP and NADPH. Of the initial O-linked alkylaryl substituted analogs, compound 6e showed most potent activity against Pf parasites at 3.60 μM. Additional compounds varying the phenyl ring of 6e were synthesized. The most potent phosphonic acids, 6l and 6n, display nM activity against PfDXR and low μM activity against Pf parasites. Prodrugs of these compounds were less effective against Pf parasites but showed modest activity against Mtb cells. Data from this series of compounds suggests that this combination of substituents can be advantageous in designing a new generation of antimicrobials.
Collapse
Affiliation(s)
- Darean Bague
- Department of Chemistry, George Washington University Washington D.C. 20052 USA
| | - Ruiqin Wang
- Department of Chemistry, George Washington University Washington D.C. 20052 USA
| | - Dana Hodge
- Division of Infectious Diseases, Children's Hospital of Philadelphia Philadelphia PA 19104 USA
| | - Marwa O Mikati
- Department of Molecular Microbiology, Washington University School of Medicine St. Louis MO 63110 USA
| | - Jose S Roma
- Tuberculosis Research Section, LCIM, NIAID/NIH Bethesda MD 20892 USA
| | - Helena I Boshoff
- Tuberculosis Research Section, LCIM, NIAID/NIH Bethesda MD 20892 USA
| | - Allyson L Dailey
- Department of Chemistry and Biochemistry, George Mason University Fairfax VA 22030 USA
| | - Misgina Girma
- Department of Chemistry and Biochemistry, George Mason University Fairfax VA 22030 USA
| | - Robin D Couch
- Department of Chemistry and Biochemistry, George Mason University Fairfax VA 22030 USA
| | - Audrey R Odom John
- Division of Infectious Diseases, Children's Hospital of Philadelphia Philadelphia PA 19104 USA
- Department of Molecular Microbiology, Washington University School of Medicine St. Louis MO 63110 USA
| | - Cynthia S Dowd
- Department of Chemistry, George Washington University Washington D.C. 20052 USA
| |
Collapse
|
37
|
Rahim MA, Seo H, Kim S, Barman I, Ghorbanian F, Hossain MS, Shuvo MSH, Lee S, Song HY. Exploring the potential of Lactocaseibacillus rhamnosus PMC203 in inducing autophagy to reduce the burden of Mycobacterium tuberculosis. Med Microbiol Immunol 2024; 213:14. [PMID: 38977511 PMCID: PMC11231020 DOI: 10.1007/s00430-024-00794-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/15/2024] [Indexed: 07/10/2024]
Abstract
Mycobacterium tuberculosis, a lethal pathogen in human history, causes millions of deaths annually, which demands the development of new concepts of drugs. Considering this fact, earlier research has explored the anti-tuberculosis potential of a probiotic strain, Lactocaseibacillus rhamnosus PMC203, leading to a subsequent focus on the molecular mechanism involved in its effect, particularly on autophagy. In this current study, immunoblotting-based assay exhibited a remarkable expression of autophagy marker LC3-II in the PMC203 treated group compared to an untreated group. A remarkable degradation of p62 was also noticed within treated cells compared to control. Furthermore, the immunofluorescence-based assay showed significant fold change in fluorescence intensity for alexa-647-LC3 and alexa-488-LC3, whereas p62 was degraded noticeably. Moreover, lysosomal biogenesis generation was elevated significantly in terms of LAMP1 and acidic vesicular organelles. As a result, PMC203-induced autophagy played a vital role in reducing M. tuberculosis burden within the macrophages in treated groups compared to untreated group. A colony -forming unit assay also revealed a significant reduction in M. tuberculosis in the treated cells over time. Additionally, the candidate strain significantly upregulated the expression of autophagy induction and lysosomal biogenesis genes. Together, these results could enrich our current knowledge of probiotics-mediated autophagy in tuberculosis and suggest its implications for innovatively managing tuberculosis.
Collapse
Affiliation(s)
- Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
- Human Microbiome Medical Research Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Hoonhee Seo
- Human Microbiome Medical Research Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Sukyung Kim
- Human Microbiome Medical Research Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
- Human Microbiome Medical Research Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Fatemeh Ghorbanian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
- Human Microbiome Medical Research Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
- Human Microbiome Medical Research Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Md Sarower Hossen Shuvo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
- Human Microbiome Medical Research Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Saebim Lee
- Human Microbiome Medical Research Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
- Human Microbiome Medical Research Center, Soonchunhyang University, Asan, 31538, Republic of Korea.
| |
Collapse
|
38
|
Zhang SY, Qiu L, Zhang SX, Xiao HP, Chu NH, Zhang X, Zhang HQ, Zheng PY, Zhang HY, Lu ZH. Efficacy and Safety of Bufei Jiedu Granules in Treating Multidrug-Resistant Pulmonary Tuberculosis: A Multi-center, Double-Blinded and Randomized Controlled Trial. Chin J Integr Med 2024; 30:579-587. [PMID: 38733454 DOI: 10.1007/s11655-024-3812-7] [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] [Accepted: 09/06/2023] [Indexed: 05/13/2024]
Abstract
OBJECTIVE To assess the efficacy and safety of Bufei Jiedu (BFJD) ranules as adjuvant therapy for patients with multidrug-resistant pulmonary tuberculosis (MDR-PTB). METHODS A large-scale, multi-center, double-blinded, and randomized controlled trial was conducted in 18 sentinel hospitals in China from December 2012 to December 2016. A total of 312 MDR-PTB patients were randomly assigned to BFJD Granules or placebo groups (1:1) using a stratified randomization method, which both received the long-course chemotherapy regimen for 18 months (6 Am-Lfx-P-Z-Pto, 12 Lfx-P-Z-Pto). Meanwhile, patients in both groups also received BFJD Granules or placebo twice a day for a total of 18 months, respectively. The primary outcome was cure rate. The secondary outcomes included time to sputum-culture conversion, changes in lung cavities and quality of life (QoL) of patients. Adverse reactions were monitored during and after the trial. RESULTS A total of 216 cases completed the trial, 111 in the BFJD Granules group and 105 in the placebo group. BFJD Granules, as an adjuvant treatment, increased the cure rate by 13.6% at the end of treatment, compared with the placebo (58.4% vs. 44.8%, P=0.02), and accelerated the median time to sputum-culture conversion (5 months vs. 11 months). The cavity closure rate of the BFJD Granules group (50.6%, 43/85) was higher than that of the placebo group (32.1%, 26/81; P=0.02) in patients who completed the treatment. At the end of the intensive treatment, according to the 36-item Short Form, the BFJD Granules significantly improved physical functioning, general health, and vitality of patients relative to the placebo group (all P<0.01). Overall, the death rates in the two groups were not significantly different; 5.1% (8/156) in the BFJD Granules group and 2.6% (4/156) in the placebo group. CONCLUSIONS Supplementing BFJD Granules with the long-course chemotherapy regimen significantly increased the cure rate and cavity closure rates, and rapidly improved QoL of patients with MDR-PTB (Registration No. ChiCTR-TRC-12002850).
Collapse
Affiliation(s)
- Shao-Yan Zhang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lei Qiu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shun-Xian Zhang
- Clinical Research Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - He-Ping Xiao
- Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University, Shanghai, 200433, China
| | - Nai-Hui Chu
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing, 101100, China
| | - Xia Zhang
- Department of Tuberculosis, the Second Hospital of Nanjing, Nanjing, 210003, China
| | - Hui-Qiang Zhang
- Department of Tuberculosis, the First Hospital Affiliated to Xinxiang Medical College, Xinxiang, Henan Province, 453100, China
| | - Pei-Yong Zheng
- Clinical Research Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Hui-Yong Zhang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhen-Hui Lu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
39
|
Komarova K, Vinogradova L, Lukin A, Zhuravlev M, Deniskin D, Chudinov M, Gureev M, Dogonadze M, Zabolotnykh N, Vinogradova T, Lavrova A, Yablonskiy P. The Nitrofuran-Warhead-Equipped Spirocyclic Azetidines Show Excellent Activity against Mycobacterium tuberculosis. Molecules 2024; 29:3071. [PMID: 38999023 PMCID: PMC11243650 DOI: 10.3390/molecules29133071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
A series of 21 new 7'H-spiro[azetidine-3,5'-furo [3,4-d]pyrimidine]s substituted at the pyrimidine ring second position were synthesized. The compounds showed high antibacterial in vitro activity against M. tuberculosis. Two compounds had lower minimum inhibitory concentrations against Mtb (H37Rv strain) compared with isoniazid. The novel spirocyclic scaffold shows excellent properties for anti-tuberculosis drug development.
Collapse
Affiliation(s)
- Kristina Komarova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119454 Moscow, Russia
| | - Lyubov Vinogradova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119454 Moscow, Russia
| | - Alexey Lukin
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119454 Moscow, Russia
| | - Maxim Zhuravlev
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119454 Moscow, Russia
| | - Dmitry Deniskin
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119454 Moscow, Russia
| | - Mikhail Chudinov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 119454 Moscow, Russia
| | - Maxim Gureev
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave 4, 194064 Saint Petersburg, Russia
| | - Marine Dogonadze
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, 191036 Saint Petersburg, Russia
| | - Natalia Zabolotnykh
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, 191036 Saint Petersburg, Russia
| | - Tatiana Vinogradova
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, 191036 Saint Petersburg, Russia
| | - Anastasia Lavrova
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, 191036 Saint Petersburg, Russia
- Sophya Kovalevskaya North-West Mathematical Research Center, Immanuel Kant Baltic Federal University, 236041 Kaliningrad, Russia
| | - Petr Yablonskiy
- Department of Hospital Surgery, Faculty of Medicine, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| |
Collapse
|
40
|
Shurygina APS, Zabolotnykh NV, Vinogradova TI, Vitovskaya ML, Dogonadze MZ, Vasilyev KA, Buzitskaya ZV, Yablonskiy PK, Lioznov DA, Stukova MA. TB/FLU-06E Influenza Vector-Based Vaccine in the Complex Therapy of Drug-Susceptible and Drug-Resistant Experimental Tuberculosis. Pharmaceutics 2024; 16:857. [PMID: 39065554 PMCID: PMC11279844 DOI: 10.3390/pharmaceutics16070857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
The steady rise of drug-resistant tuberculosis (TB), which renders standard therapy regimens ineffective, necessitates the development of innovative treatment approaches. Immunotherapeutic vaccines have the potential to effectively regulate the anti-TB immune response and enhance the efficacy of anti-TB treatment. In the present study, we aimed to evaluate the potency of the mucosal vector vaccine TB/FLU-06E as part of a complex treatment regimen for drug-susceptible (DS) or drug-resistant (DR) tuberculosis in C57BL/6 mice. Incorporating TB/FLU-06E into the treatment protocol significantly increased the effectiveness of therapy for both forms of tuberculosis. It was evidenced by higher survival rates and reduced pulmonary bacterial load (1.83 lg CFU for DS tuberculosis and 0.93 lg CFU for DR tuberculosis). Furthermore, the treatment reduced pathomorphological lesions in the lungs and stimulated the local and systemic T-helper 1 (Th1) and cytotoxic T-lymphocyte (CTL) anti-TB immune responses. Thus, therapeutic immunization with the TB/FLU-06E vaccine significantly enhances the efficacy of tuberculosis treatment, which is particularly important in DR tuberculosis.
Collapse
Affiliation(s)
- Anna-Polina S. Shurygina
- Smorodintsev Research Institute of Influenza, The Ministry of Health of the Russian Federation, 197022 Saint-Petersburg, Russia
| | - Natalia V. Zabolotnykh
- Saint-Petersburg State Research Institute of Phthisiopulmonology, The Ministry of Health of the Russian Federation, 194064 Saint-Petersburg, Russia
| | - Tatiana I. Vinogradova
- Saint-Petersburg State Research Institute of Phthisiopulmonology, The Ministry of Health of the Russian Federation, 194064 Saint-Petersburg, Russia
| | - Maria L. Vitovskaya
- Saint-Petersburg State Research Institute of Phthisiopulmonology, The Ministry of Health of the Russian Federation, 194064 Saint-Petersburg, Russia
| | - Marine Z. Dogonadze
- Saint-Petersburg State Research Institute of Phthisiopulmonology, The Ministry of Health of the Russian Federation, 194064 Saint-Petersburg, Russia
| | - Kirill A. Vasilyev
- Smorodintsev Research Institute of Influenza, The Ministry of Health of the Russian Federation, 197022 Saint-Petersburg, Russia
| | - Zhanna V. Buzitskaya
- Smorodintsev Research Institute of Influenza, The Ministry of Health of the Russian Federation, 197022 Saint-Petersburg, Russia
| | - Petr K. Yablonskiy
- Saint-Petersburg State Research Institute of Phthisiopulmonology, The Ministry of Health of the Russian Federation, 194064 Saint-Petersburg, Russia
| | - Dmitriy A. Lioznov
- Smorodintsev Research Institute of Influenza, The Ministry of Health of the Russian Federation, 197022 Saint-Petersburg, Russia
| | - Marina A. Stukova
- Smorodintsev Research Institute of Influenza, The Ministry of Health of the Russian Federation, 197022 Saint-Petersburg, Russia
| |
Collapse
|
41
|
Santos-Júnior PFDS, Batista VDM, Nascimento IJDS, Nunes IC, Silva LR, Costa CACB, Freitas JDD, Quintans-Júnior LJ, Araújo-Júnior JXD, Freitas MEGD, Zhan P, Green KD, Garneau-Tsodikova S, Mendonça-Júnior FJB, Rodrigues-Junior VS, Silva-Júnior EFD. A consensus reverse docking approach for identification of a competitive inhibitor of acetyltransferase enhanced intracellular survival protein from Mycobacterium tuberculosis. Bioorg Med Chem 2024; 108:117774. [PMID: 38833750 DOI: 10.1016/j.bmc.2024.117774] [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: 01/24/2024] [Revised: 05/08/2024] [Accepted: 05/24/2024] [Indexed: 06/06/2024]
Abstract
Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb), which remains a significant global health challenge. The emergence of multidrug-resistant (MDR) Mtb strains imposes the development of new therapeutic strategies. This study focuses on the identification and evaluation of potential inhibitors against Mtb H37Ra through a comprehensive screening of an in-house chemolibrary. Subsequently, a promising pyrimidine derivative (LQM495) was identified as promising and then further investigated by experimental and in silico approaches. In this context, computational techniques were used to elucidate the potential molecular target underlying the inhibitory action of LQM495. Then, a consensus reverse docking (CRD) protocol was used to investigate the interactions between this compound and several Mtb targets. Out of 98 Mtb targets investigated, the enhanced intracellular survival (Eis) protein emerged as a target for LQM495. To gain insights into the stability of the LQM495-Eis complex, molecular dynamics (MD) simulations were conducted over a 400 ns trajectory. Further insights into its binding modes within the Eis binding site were obtained through a Quantum mechanics (QM) approach, using density functional theory (DFT), with B3LYP/D3 basis set. These calculations shed light on the electronic properties and reactivity of LQM495. Subsequently, inhibition assays and kinetic studies of the Eis activity were used to investigate the activity of LQM495. Then, an IC50 value of 11.0 ± 1.4 µM was found for LQM495 upon Eis protein. Additionally, its Vmax, Km, and Ki parameters indicated that it is a competitive inhibitor. Lastly, this study presents LQM495 as a promising inhibitor of Mtb Eis protein, which could be further explored for developing novel anti-TB drugs in the future.
Collapse
Affiliation(s)
| | - Vitoria de Melo Batista
- Research Group of Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil
| | - Igor José Dos Santos Nascimento
- Post-Graduation Program of Pharmaceutical Sciences, Pharmacy Department, State University of Paraíba, Campina Grande, Brazil
| | - Isabelle Cavalcante Nunes
- Research Group of Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil
| | - Leandro Rocha Silva
- Research Group of Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil
| | | | - Johnnatan Duarte de Freitas
- Department of Chemistry, Federal Institute of Alagoas, Maceió campus, Mizael Domingues Street, 57020-600 Maceió, Alagoas, Brazil
| | - Lucindo José Quintans-Júnior
- Pharmaceutical Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, Sergipe 49100-001, Brazil
| | - João Xavier de Araújo-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil
| | | | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Keith D Green
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, 40536-0596, United States
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, 40536-0596, United States
| | | | - Valnês S Rodrigues-Junior
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa, Brazil; Post-Graduation Program in Natural Products and Bioactive Synthetics, Federal University of Paraíba, João Pessoa, Brazil
| | - Edeildo Ferreira da Silva-Júnior
- Research Group of Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, AC. Simões campus, 57072-970 Alagoas, Maceió, Brazil.
| |
Collapse
|
42
|
Zheng J, Dong Z, Jin X, Li J, Zou Y, Bai G, Wu Q, Xu S, Wang Z, Sun X, Liu D, Guo L. In vitro Antibacterial Effect Study of Plasma-Activated Saline on Mycobacterium Tuberculosis. Infect Drug Resist 2024; 17:2315-2328. [PMID: 38882657 PMCID: PMC11179663 DOI: 10.2147/idr.s456181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/22/2024] [Indexed: 06/18/2024] Open
Abstract
Purpose This study aimed to investigate the antibacterial effects of plasma-activated saline (PAS) on My-cobacterium tuberculosis (Mtb). Methods We conducted a growth assay on 3 strains of Mtb and an antibiotic sensitivity test on 4 strains of Mtb. Both tests included groups treated with normal saline (NS), PAS, and hydrochloric acid (HCl). The test of antibiotic sensitivity consisted of parallel tests with two concentrations of bacteria suspension: 10-2 and 10-4. The selected antibiotics were rifampicin (RIF), isoniazid (INH), ethambutol (EMB), and streptomycin (SM). The number of bacteria was determined after one month of culture under different conditions. The Kruskal-Wallis test was used to analyze the differences in grouping factors at representative time points. Results The growth assay indicated that PAS significantly inhibited the growth of 3 strains of Mtb compared with NS and HCl treatment groups. Furthermore, except for the initial observation time point, the remaining three observation time points consistently demonstrate no significant differences between the NS group and the HCl group. The antibiotic sensitivity test of INH, SM, and RIF indicated that PAS could inhibit the growth of antibiotic-resistant Mtb, and the antibiotic sensitivity test of INH and SM with bacterial suspension concentration of 10-2 and SM with bacterial suspension concentration of 10-4 showed statistically different results. The antibiotic sensitivity test of EMB indicated that the growth of Mtb in PAS was slower than that in NS and HCl in both antibiotic-resistant and sensitive Mtb, but there was no statistical difference. Conclusion The study indicates that PAS contains a significant amount of active substances and exhibits high oxidizability and an acidic pH state. The unique physicochemical properties of PAS significantly delayed the growth of Mtb, compared to the NS and the HCl. PAS not only inhibited the growth of drug-sensitive strains but also significantly enhanced the sensitivity of drug-resistant strains to anti-tuberculosis drugs, which may provide a new therapeutic strategy for the treatment of tuberculosis.
Collapse
Affiliation(s)
- Jianbao Zheng
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
- Tuberculosis Prevention and Treatment Hospital in Shaanxi Province, Xi'an, 710100, People's Republic of China
| | - Zepeng Dong
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Xianzhen Jin
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Jing Li
- Tuberculosis Prevention and Treatment Hospital in Shaanxi Province, Xi'an, 710100, People's Republic of China
| | - Yuanwu Zou
- Tuberculosis Prevention and Treatment Hospital in Shaanxi Province, Xi'an, 710100, People's Republic of China
| | - Guanghong Bai
- Tuberculosis Prevention and Treatment Hospital in Shaanxi Province, Xi'an, 710100, People's Republic of China
| | - Qianhong Wu
- Tuberculosis Prevention and Treatment Hospital in Shaanxi Province, Xi'an, 710100, People's Republic of China
| | - Shenghang Xu
- Center for Plasma Biomedicine, State Key Laboratory of Electrical Insulation and Power Equipment, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Zifeng Wang
- Center for Plasma Biomedicine, State Key Laboratory of Electrical Insulation and Power Equipment, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Xuejun Sun
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Dingxin Liu
- Center for Plasma Biomedicine, State Key Laboratory of Electrical Insulation and Power Equipment, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Li Guo
- Center for Plasma Biomedicine, State Key Laboratory of Electrical Insulation and Power Equipment, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| |
Collapse
|
43
|
Glajzner P, Bernat A, Jasińska-Stroschein M. Improving the treatment of bacterial infections caused by multidrug-resistant bacteria through drug repositioning. Front Pharmacol 2024; 15:1397602. [PMID: 38910882 PMCID: PMC11193365 DOI: 10.3389/fphar.2024.1397602] [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: 03/07/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Drug repurposing (repositioning) is a dynamically-developing area in the search for effective therapy of infectious diseases. Repositioning existing drugs with a well-known pharmacological and toxicological profile is an attractive method for quickly discovering new therapeutic indications. The off-label use of drugs for infectious diseases requires much less capital and time, and can hasten progress in the development of new antimicrobial drugs, including antibiotics. The use of drug repositioning in searching for new therapeutic options has brought promising results for many viral infectious diseases, such as Ebola, ZIKA, Dengue, and HCV. This review describes the most favorable results for repositioned drugs for the treatment of bacterial infections. It comprises publications from various databases including PubMed and Web of Science published from 2015 to 2023. The following search keywords/strings were used: drug repositioning and/or repurposing and/or antibacterial activity and/or infectious diseases. Treatment options for infections caused by multidrug-resistant bacteria were taken into account, including methicillin-resistant staphylococci, multidrug-resistant Mycobacterium tuberculosis, or carbapenem-resistant bacteria from the Enterobacteriaceae family. It analyses the safety profiles of the included drugs and their synergistic combinations with antibiotics and discusses the potential of antibacterial drugs with antiparasitic, anticancer, antipsychotic effects, and those used in metabolic diseases. Drug repositioning may be an effective response to public health threats related to the spread of multidrug-resistant bacterial strains and the growing antibiotic resistance of microorganisms.
Collapse
Affiliation(s)
- Paulina Glajzner
- Department of Biopharmacy, Faculty of Pharmacy, Medical University of Lodz, Łódź, Poland
| | | | | |
Collapse
|
44
|
Wulandari DA, Hartati YW, Ibrahim AU, Pitaloka DAE, Irkham. Multidrug-resistant tuberculosis. Clin Chim Acta 2024; 559:119701. [PMID: 38697459 DOI: 10.1016/j.cca.2024.119701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/05/2024]
Abstract
One of predominant contributors to global mortality is tuberculosis (TB), an infection caused by Mycobacterium tuberculosis (MTB). Inappropriate and ineffectual treatment can lead to the development of drug-resistant TB. One of the most common forms of drug-resistant TB is multidrug-resistant tuberculosis (MDR-TB), caused by mutations in the rpoB and katG genes that lead to resistance to anti-TB drugs, rifampicin (RIF) and isoniazid (INH), respectively. Although culturing remains the gold standard, it is not rapid thereby delaying potential treatment and potentially increasing the incidence of MDR-TB. In contrast, molecular techniques provide a highly sensitive and specific alternative. This review discusses the classification of biomarkers used to detect MDR-TB, some of the commonly used anti-TB drugs, and DNA mutations in MTB that lead to anti-TB resistance. The objective of this review is to increase awareness of the need for rapid and precise detection of MDR-TB cases to decrease morbidity and mortality of this infectious disease worldwide.
Collapse
Affiliation(s)
- Dika Apriliana Wulandari
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km 21, 45363, Indonesia
| | - Yeni Wahyuni Hartati
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km 21, 45363, Indonesia
| | - Abdullahi Umar Ibrahim
- Department of Biomedical Engineering, Near East University, Mersin 10, Nicosia 99010, Turkey; Research Center for Science, Technology and Engineering (BILTEM), Near East University, 99138 Nicosia, TRNC, Mersin 10, Turkey
| | - Dian Ayu Eka Pitaloka
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Irkham
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km 21, 45363, Indonesia.
| |
Collapse
|
45
|
Alavidze I, Shubitidze M, Khodeli G, Dvali S, Tskitishvili A. Numerous Asymptomatic Brain Tuberculomas Complicated by Fatal Tuberculous Meningitis. Cureus 2024; 16:e63090. [PMID: 39055453 PMCID: PMC11270632 DOI: 10.7759/cureus.63090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Tuberculosis (TB) is still one of the most challenging infectious diseases worldwide. Coinfection with HIV increases the likelihood of extrapulmonary involvement, including the tuberculosis of the central nervous system (CNS-TB). CNS-TB often presents as tuberculomas or tuberculous meningitis. Although tuberculomas can be single or multiple, asymptomatic carriage of numerous tuberculomas is seldom reported. We present a case of a 55-year-old man who carried at least 34 tuberculomas of different sizes asymptomatically before developing and succumbing to tuberculous meningitis. Furthermore, we highlight several possible public health challenges that might have complicated his clinical course, suggesting that future studies also focus on these variables alongside more traditional clinical issues.
Collapse
Affiliation(s)
- Irakli Alavidze
- Aieti Medical School, David Tvildiani Medical University, Tbilisi, GEO
| | - Mariam Shubitidze
- Aieti Medical School, David Tvildiani Medical University, Tbilisi, GEO
| | | | - Shorena Dvali
- HIV/AIDS Department, Infectious Diseases, AIDS and Clinical Immunology Research Center, Tbilisi, GEO
| | | |
Collapse
|
46
|
Tang C, Wu L, Li M, Dai J, Shi Y, Wang Q, Xu F, Zheng L, Xiao X, Cai J, Zhang Y, Yang Y, Zheng X, Xiang G. High-throughput nanopore targeted sequencing for efficient drug resistance assay of Mycobacterium tuberculosis. Front Microbiol 2024; 15:1331656. [PMID: 38841074 PMCID: PMC11152171 DOI: 10.3389/fmicb.2024.1331656] [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: 11/01/2023] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
Drug-resistant tuberculosis (TB), especially multidrug-resistant tuberculosis (MDR-TB) and extensively drug-resistant tuberculosis (XDR-TB), is one of the urgent clinical problems and public health challenges. Culture-based phenotypic drug susceptibility testing (pDST) is time-consuming, and PCR-based assays are limited to hotspot mutations. In this study, we developed and validated a convenient and efficient approach based on high-throughput nanopore sequencing technology combined with multiplex PCR, namely nanopore targeted sequencing (NTS), to simultaneously sequence 18 genes associated with antibiotic resistance in Mycobacterium tuberculosis (MTB). The analytical performance of NTS was evaluated, and 99 clinical samples were collected to assess its clinical performance. The NTS results showed that MTB and its drug resistance were successfully identified in approximately 7.5 h. Furthermore, compared to the pDST and Xpert MTB/RIF assays, NTS provided much more drug resistance information, covering 14 anti-TB drugs, and it identified 20 clinical cases of drug-resistant MTB. The mutations underlying these drug-resistant cases were all verified using Sanger sequencing. Our approach for this TB drug resistance assay offers several advantages, including being culture-free, efficient, high-throughput, and highly accurate, which would be very helpful for clinical patient management and TB infection control.
Collapse
Affiliation(s)
- Chen Tang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lianpeng Wu
- Department of Clinical Laboratory, Wenzhou Central Hospital, Wenzhou, Zhejiang, China
| | - Machao Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jianyi Dai
- Department of Infectious Diseases, Wenzhou Central Hospital, Wenzhou, Zhejiang, China
| | - Ye Shi
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiongdan Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Feng Xu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Laibao Zheng
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xingxing Xiao
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junwen Cai
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanjun Zhang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuting Yang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoqun Zheng
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guangxin Xiang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
47
|
Singh V. Tuberculosis treatment-shortening. Drug Discov Today 2024; 29:103955. [PMID: 38548262 DOI: 10.1016/j.drudis.2024.103955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/06/2024] [Accepted: 03/21/2024] [Indexed: 04/06/2024]
Abstract
Tuberculosis (TB) presents a significant global health concern, with ∼10 million people developing TB and 1.3 million people dying from the disease each year. The standard treatment regimen for drug-susceptible TB was between 6 and 9 months until recently, presenting a prolonged therapeutic duration compared with other infectious diseases. This is a long time for patients to adhere to the medication, consequently increasing the risk of developing drug-resistant Mycobacterium tuberculosis - a significant challenge in TB management globally. Therefore, the primary objective of contemporary TB drug development research is to shorten the treatment duration. This review comprehensively explores the strategies aimed at shortening TB treatment.
Collapse
Affiliation(s)
- Vinayak Singh
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch 7701, South Africa; South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Observatory 7925, South Africa.
| |
Collapse
|
48
|
Niculescu AG, Mük GR, Avram S, Vlad IM, Limban C, Nuta D, Grumezescu AM, Chifiriuc MC. Novel strategies based on natural products and synthetic derivatives to overcome resistance in Mycobacterium tuberculosis. Eur J Med Chem 2024; 269:116268. [PMID: 38460268 DOI: 10.1016/j.ejmech.2024.116268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/09/2024] [Accepted: 02/18/2024] [Indexed: 03/11/2024]
Abstract
One of the biggest health challenges of today's world is the emergence of antimicrobial resistance (AMR), which renders conventional therapeutics insufficient and urgently demands the generation of novel antimicrobial strategies. Mycobacterium tuberculosis (M. tuberculosis), the pathogen causing tuberculosis (TB), is among the most successful bacteria producing drug-resistant infections. The versatility of M. tuberculosis allows it to evade traditional anti-TB agents through various acquired and intrinsic mechanisms, rendering TB among the leading causes of infectious disease-related mortality. In this context, researchers worldwide focused on establishing novel approaches to address drug resistance in M. tuberculosis, developing diverse alternative treatments with varying effectiveness and in different testing phases. Overviewing the current progress, this paper aims to briefly present the mechanisms involved in M. tuberculosis drug-resistance, further reviewing in more detail the under-development antibiotics, nanotechnological approaches, and natural therapeutic solutions that promise to overcome current treatment limitations.
Collapse
Affiliation(s)
- Adelina-Gabriela Niculescu
- Research Institute of the University of Bucharest, University of Bucharest, 90 Panduri Road, Bucharest, Romania; Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061, Bucharest, Romania.
| | - Georgiana Ramona Mük
- Faculty of Biology, University of Bucharest, Splaiul Independenței 91-95, Bucharest, R-050095, Romania; St. Stephen's Pneumoftiziology Hospital, Șoseaua Ștefan cel Mare 11, Bucharest, 020122, Romania.
| | - Speranta Avram
- Faculty of Biology, University of Bucharest, Splaiul Independenței 91-95, Bucharest, R-050095, Romania.
| | - Ilinca Margareta Vlad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia, 020956, Bucharest, Romania.
| | - Carmen Limban
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia, 020956, Bucharest, Romania.
| | - Diana Nuta
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia, 020956, Bucharest, Romania.
| | - Alexandru Mihai Grumezescu
- Research Institute of the University of Bucharest, University of Bucharest, 90 Panduri Road, Bucharest, Romania; Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061, Bucharest, Romania.
| | - Mariana-Carmen Chifiriuc
- Research Institute of the University of Bucharest, University of Bucharest, 90 Panduri Road, Bucharest, Romania; Faculty of Biology, University of Bucharest, Splaiul Independenței 91-95, Bucharest, R-050095, Romania.
| |
Collapse
|
49
|
O'Connell LM, Coffey A, O'Mahony JM. Genomic analysis of seven mycobacteriophages identifies three novel species with differing phenotypic stabilities. Heliyon 2024; 10:e27932. [PMID: 38515691 PMCID: PMC10955285 DOI: 10.1016/j.heliyon.2024.e27932] [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: 08/31/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
Recently, case studies have been published regarding the application of mycobacteriophage (MP) therapy (MPT) in patients with multi-antibiotic-resistant infections. A major limitation in the development of MPT is the paucity of therapeutically useful MP. As there are approximately 10,000 MP that have yet to be sequenced, it is possible that characterization of this cohort would increase the repertoire of useful MP. This study aims to contribute to such a strategy, by characterizing a cohort of 7 mycobacteriophages. Sequencing analyses revealed that the MP have unique sequences, and subsequent gene annotation revealed differences in gene organization. Notably, MP LOCARD has the largest genome and operons encoding for glycosyltransferases. Taxonomic analysis executed with VIRIDIC, Gegenees and VICTOR revealed that LOCARD belongs to a different genus than the other phages and is the foundational member of one of three novel species identified in this study. LOCARD, LOCV2, and LOCV5 were selected as representative members of their species and subjected to phenotypic analyses to compare their stability under biologically and industrially relevant conditions. Again LOCARD stood out, as it was unaffected by the typical temperatures (37 °C) and salinity (0.9%) experienced in mammals, while the viability of LOCV2 and LOCV5 was significantly reduced. LOCARD was also tolerant to pH 10, low levels of antiviral detergent and was the least impacted by a single freeze-thaw cycle. When all these results are considered, it indicates that LOCARD in particular, has potential therapeutic and/or diagnostics applications, given its resilience towards physiological and storage conditions.
Collapse
Affiliation(s)
- Laura M. O'Connell
- Munster Technological University, Rossa Avenue, Bishopstown, Cork, T12 P928, Ireland
| | - Aidan Coffey
- Munster Technological University, Rossa Avenue, Bishopstown, Cork, T12 P928, Ireland
| | - Jim M. O'Mahony
- Munster Technological University, Rossa Avenue, Bishopstown, Cork, T12 P928, Ireland
| |
Collapse
|
50
|
Nyambo K, Tapfuma KI, Adu-Amankwaah F, Julius L, Baatjies L, Niang IS, Smith L, Govender KK, Ngxande M, Watson DJ, Wiesner L, Mavumengwana V. Molecular docking, molecular dynamics simulations and binding free energy studies of interactions between Mycobacterium tuberculosis Pks13, PknG and bioactive constituents of extremophilic bacteria. Sci Rep 2024; 14:6794. [PMID: 38514663 PMCID: PMC10957976 DOI: 10.1038/s41598-024-57124-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
Mycobacterial pathogens present a significant challenge to disease control efforts globally due to their inherent resistance to multiple antibiotics. The rise of drug-resistant strains of Mycobacterium tuberculosis has prompted an urgent need for innovative therapeutic solutions. One promising way to discover new tuberculosis drugs is by utilizing natural products from the vast biochemical space. Multidisciplinary methods can used to harness the bioactivity of these natural products. This study aimed to evaluate the antimycobacterial efficacy of functional crude extracts from bacteria isolated from gold mine tailings in South Africa. Bacterial strains were identified using 16S rRNA sequencing. The crude extracts obtained from the bacteria were tested against Mycobacterium tuberculosis H37Rv, Mycobacterium smegmatis mc2155, and Mycobacterium aurum A+. Untargeted HPLC-qTOF and molecular networking were used to identify the functional constituents present in extracts that exhibited inhibitory activity. A virtual screening workflow (VSW) was used to filter compounds that were strong binders to Mycobacterium tuberculosis Pks13 and PknG. The ligands returned from the VSW were subjected to optimization using density functional theory (DFT) at M06-2X/6-311++ (d,p) level of theory and basis set implemented in Gaussian16 Rev.C01. The optimized ligands were re-docked against Mycobacterium tuberculosis Pks13 and PknG. Molecular dynamics simulation and molecular mechanics generalized born surface area were used to evaluate the stability of the protein-ligand complexes formed by the identified hits. The hit that showed promising binding characteristics was virtually modified through multiple synthetic routes using reaction-driven enumeration. Three bacterial isolates showed significant activity against the two strains of Mycobacterium, while only two, Bacillus subtilis and Bacillus licheniformis, exhibited activity against both Mycobacterium tuberculosis H37Rv, Mycobacterium smegmatis mc2155, and Mycobacterium aurum A+. The tentatively identified compounds from the bacterial crude extracts belonged to various classes of natural compounds associated with antimicrobial activity. Two compounds, cyclo-(L-Pro-4-OH-L-Leu) and vazabitide A, showed strong binding against PknG and Pks13, with pre-MD MM-GBSA values of - 42.8 kcal/mol and - 47.6 kcal/mol, respectively. The DFT-optimized compounds exhibited the same docking scores as the ligands optimized using the OPSL-4 force field. After modifying vazabitide A, its affinity to the Pks13 binding site increased to - 85.8 kcal/mol, as revealed by the post-MD MM-GBSA analysis. This study highlights the potential of bacteria isolates from gold mine tailings as a source of new scaffolds for designing and optimizing anti-Mycobacterium agents. These agents synthesized in-silico can be further tested in-vitro to evaluate their efficacy.
Collapse
Affiliation(s)
- Kudakwashe Nyambo
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, Cape Town, South Africa
| | - Kudzanai Ian Tapfuma
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, Cape Town, South Africa
| | - Francis Adu-Amankwaah
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, Cape Town, South Africa
| | - Lauren Julius
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, Cape Town, South Africa
| | - Lucinda Baatjies
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, Cape Town, South Africa
| | - Idah Sithole Niang
- Department of Biotechnology and Biochemistry, University of Zimbabwe, B064, Mount Pleasant, Harare, Zimbabwe
| | - Liezel Smith
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, Cape Town, South Africa
| | - Krishna Kuben Govender
- Department of Chemical Sciences, University of Johannesburg, Doornfontein Campus, P.O. Box 17011, Johannesburg, 2028, South Africa
- National Institute for Theoretical and Computational Sciences (NITheCS), Cape Town, South Africa
| | - Mkhuseli Ngxande
- Computer Science Division, Department of Mathematical Sciences, Faculty of Science, University of Stellenbosch, Matieland, South Africa
| | - Daniel J Watson
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Vuyo Mavumengwana
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, 7505, Cape Town, South Africa.
| |
Collapse
|