1
|
Mahjabeen A, Hasan MZ, Rahman MT, Islam MA, Khan RT, Kaiser MS. Genetic insights into the connection between pulmonary TB and non-communicable diseases: An integrated analysis of shared genes and potential treatment targets. PLoS One 2024; 19:e0312072. [PMID: 39432502 PMCID: PMC11493268 DOI: 10.1371/journal.pone.0312072] [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: 12/12/2023] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Pulmonary Tuberculosis (PTB) is a significant global health issue due to its high incidence, drug resistance, contagious nature, and impact on people with compromised immune systems. As mentioned by the World Health Organization (WHO), TB is responsible for more global fatalities than any other infectious illness. On the other side, WHO also claims that noncommunicable diseases (NCDs) kill 41 million people yearly worldwide. In this regard, several studies suggest that PTB and NCDs are linked in various ways and that people with PTB are more likely to acquire NCDs. At the same time, NCDs can increase susceptibility to active TB infection. Furthermore, because of potential drug interactions and therapeutic challenges, treating individuals with both PTB and NCDs can be difficult. This study focuses on seven NCDs (lung cancer (LC), diabetes mellitus (DM), Parkinson's disease (PD), silicosis (SI), chronic kidney disease (CKD), cardiovascular disease (CVD), and rheumatoid arthritis (RA)) and rigorously presents the genetic relationship with PTB regarding shared genes and outlines possible treatment plans. OBJECTIVES BlueThis study aims to identify the drug components that can regulate abnormal gene expression in NCDs. The study will reveal hub genes, potential biomarkers, and drug components associated with hub genes through statistical measures. This will contribute to targeted therapeutic interventions. METHODS Numerous investigations, including protein-protein interaction (PPI), gene regulatory network (GRN), enrichment analysis, physical interaction, and protein-chemical interaction, have been carried out to demonstrate the genetic correlation between PTB and NCDs. During the study, nine shared genes such as TNF, IL10, NLRP3, IL18, IFNG, HMGB1, CXCL8, IL17A, and NFKB1 were discovered between TB and the above-mentioned NCDs, and five hub genes (NFKB1, TNF, CXCL8, NLRP3, and IL10) were selected based on degree values. RESULTS AND CONCLUSION In this study, we found that all of the hub genes are linked with the 10 drug components, and it was observed that aspirin CTD 00005447 was mostly associated with all the other hub genes. This bio-informatics study may help researchers better understand the cause of PTB and its relationship with NCDs, and eventually, this can lead to exploring effective treatment plans.
Collapse
Affiliation(s)
- Amira Mahjabeen
- Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Dhaka, Bangladesh
| | - Md. Zahid Hasan
- Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Dhaka, Bangladesh
| | - Md. Tanvir Rahman
- Department of Information and Communication Technology, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Md. Aminul Islam
- Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Dhaka, Bangladesh
| | - Risala Tasin Khan
- Institute of Information Technology, Jahangirnagar University, Dhaka, Bangladesh
| | - M. Shamim Kaiser
- Institute of Information Technology, Jahangirnagar University, Dhaka, Bangladesh
| |
Collapse
|
2
|
Thom RE, D’Elia RV. Future applications of host direct therapies for infectious disease treatment. Front Immunol 2024; 15:1436557. [PMID: 39411713 PMCID: PMC11473292 DOI: 10.3389/fimmu.2024.1436557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
New and emerging pathogens, such as SARS-CoV2 have highlighted the requirement for threat agnostic therapies. Some antibiotics or antivirals can demonstrate broad-spectrum activity against pathogens in the same family or genus but efficacy can quickly reduce due to their specific mechanism of action and for the ability of the disease causing agent to evolve. This has led to the generation of antimicrobial resistant strains, making infectious diseases more difficult to treat. Alternative approaches therefore need to be considered, which include exploring the utility of Host-Directed Therapies (HDTs). This is a growing area with huge potential but difficulties arise due to the complexity of disease profiles. For example, a HDT given early during infection may not be appropriate or as effective when the disease has become chronic or when a patient is in intensive care. With the growing understanding of immune function, a new generation of HDT for the treatment of disease could allow targeting specific pathways to augment or diminish the host response, dependent upon disease profile, and allow for bespoke therapeutic management plans. This review highlights promising and approved HDTs that can manipulate the immune system throughout the spectrum of disease, in particular to viral and bacterial pathogens, and demonstrates how the advantages of HDT will soon outweigh the potential side effects.
Collapse
Affiliation(s)
- Ruth E. Thom
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Porton Down, Salisbury, United Kingdom
| | - R V. D’Elia
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
3
|
Nyazema KB, Shey BA, Sei CJ, Peters RP, Maningi NE, Fischer GW, Bernard Fourie P. In vitro evaluation of the binding activity of novel mouse IgG1 opsonic monoclonal antibodies to Mycobacterium tuberculosis and other selected mycobacterial species. J Clin Tuberc Other Mycobact Dis 2024; 35:100435. [PMID: 38601919 PMCID: PMC11004620 DOI: 10.1016/j.jctube.2024.100435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024] Open
Abstract
Antimicrobial resistance alongside other challenges in tuberculosis (TB) therapeutics have stirred renewed interest in host-directed interventions, including the role of antibodies as adjunct therapeutic agents. This study assessed the binding efficacy of two novel IgG1 opsonic monoclonal antibodies (MABs; GG9 & JG7) at 5, 10, and 25 µg/mL to live cultures of Mycobacterium tuberculosis, M. avium, M. bovis, M. fortuitum, M. intracellulare, and M. smegmatis American Type Culture Collection laboratory reference strains, as well as clinical susceptible, multi-drug resistant, and extensively drug resistant M. tuberculosis strains using indirect enzyme-linked immunosorbent assays. These three MAB concentrations were selected from a range of concentrations used in previous optimization (binding and functional) assays. Both MABs bound to all mycobacterial species and sub-types tested, albeit to varying degrees. Statistically significant differences in MAB binding activity were observed when comparing the highest and lowest MAB concentrations (p < 0.05) for both MABs GG9 and JG7, irrespective of the M. tuberculosis resistance profile. Binding affinity increased with an increase in MAB concentration, and optimal binding was observed at 25 µg/mL. JG7 showed better binding activity than GG9. Both MABs also bound to five MOTT species, albeit at varied levels. This non-selective binding to different mycobacterial species suggests a potential role for GG9 and JG7 as adjunctive agents in anti-TB chemotherapy with the aim to enhance bacterial killing.
Collapse
Affiliation(s)
- Kudzai B. Nyazema
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, South Africa
| | - Bong-Akee Shey
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, South Africa
| | - Clara J. Sei
- Vaccine and Antibody Development, Longhorn Vaccines and Diagnostics, Gaithersburg, MD, USA
| | - Remco P.H. Peters
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, South Africa
- Foundation for Professional Development, Research Unit, East London, South Africa
| | - Nontuthuko E. Maningi
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, South Africa
- Department of Microbiology, School of Life Sciences, University of KwaZulu-Natal, South Africa
| | - Gerald W. Fischer
- Vaccine and Antibody Development, Longhorn Vaccines and Diagnostics, Gaithersburg, MD, USA
| | - P. Bernard Fourie
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, South Africa
| |
Collapse
|
4
|
Wang BY, Song K, Wang HT, Wang SS, Wang WJ, Li ZW, Du WY, Xue FZ, Zhao L, Cao WC. Comorbidity increases the risk of pulmonary tuberculosis: a nested case-control study using multi-source big data. BMC Pulm Med 2024; 24:29. [PMID: 38212743 PMCID: PMC10782630 DOI: 10.1186/s12890-023-02817-6] [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: 05/08/2023] [Accepted: 12/14/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Some medical conditions may increase the risk of developing pulmonary tuberculosis (PTB); however, no systematic study on PTB-associated comorbidities and comorbidity clusters has been undertaken. METHODS A nested case-control study was conducted from 2013 to 2017 using multi-source big data. We defined cases as patients with incident PTB, and we matched each case with four event-free controls using propensity score matching (PSM). Comorbidities diagnosed prior to PTB were defined with the International Classification of Diseases-10 (ICD-10). The longitudinal relationships between multimorbidity burden and PTB were analyzed using a generalized estimating equation. The associations between PTB and 30 comorbidities were examined using conditional logistic regression, and the comorbidity clusters were identified using network analysis. RESULTS A total of 4265 cases and 17,060 controls were enrolled during the study period. A total of 849 (19.91%) cases and 1141 (6.69%) controls were multimorbid before the index date. Having 1, 2, and ≥ 3 comorbidities was associated with an increased risk of PTB (aOR 2.85-5.16). Fourteen out of thirty comorbidities were significantly associated with PTB (aOR 1.28-7.27), and the associations differed by sex and age. Network analysis identified three major clusters, mainly in the respiratory, circulatory, and endocrine/metabolic systems, in PTB cases. CONCLUSIONS Certain comorbidities involving multiple systems may significantly increase the risk of PTB. Enhanced awareness and surveillance of comorbidity are warranted to ensure early prevention and timely control of PTB.
Collapse
Affiliation(s)
- Bao-Yu Wang
- Institute of EcoHealth, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Ke Song
- Institute of EcoHealth, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Hai-Tao Wang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shan-Shan Wang
- Institute of EcoHealth, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Wen-Jing Wang
- Institute of EcoHealth, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Zhen-Wei Li
- Institute of EcoHealth, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Wan-Yu Du
- Institute of EcoHealth, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Fu-Zhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 250012, Jinan, China
- Institute for Medical Dataology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250002, China
| | - Lin Zhao
- Institute of EcoHealth, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Wu-Chun Cao
- Institute of EcoHealth, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 20 Dongda Street, Fengtai District, Beijing, 100071, China.
| |
Collapse
|
5
|
Dwivedi R, Baindara P. Differential Regulation of TFEB-Induced Autophagy during Mtb Infection and Starvation. Microorganisms 2023; 11:2944. [PMID: 38138088 PMCID: PMC10746089 DOI: 10.3390/microorganisms11122944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Through the promotion of phagolysosome formation, autophagy has emerged as a crucial mechanism to eradicate intracellular Mycobacterium tuberculosis (Mtb). A cell-autonomous host defense mechanism called lysosome biogenesis and autophagy transports cytoplasmic cargos and bacterial phagosomes to lysosomes for destruction during infection. Similar occurrences occurred in stressful or starvation circumstances and led to autophagy, which is harmful to the cell. It is interesting to note that under both hunger and infection states, the transcription factor EB (TFEB) acts as a master regulator of lysosomal activities and autophagy. This review highlighted recent research on the multitier regulation of TFEB-induced autophagy by a variety of host effectors and Mtb sulfolipid during Mtb infection and starvation. In general, the research presented here sheds light on how lysosome biogenesis and autophagy are differentially regulated by the TFEB during Mtb infection and starvation.
Collapse
Affiliation(s)
- Richa Dwivedi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Piyush Baindara
- Radiation Oncology, NextGen Precision Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
6
|
Ning B, Shen J, Liu F, Zhang H, Jiang X. Baicalein Suppresses NLRP3 and AIM2 Inflammasome-Mediated Pyroptosis in Macrophages Infected by Mycobacterium tuberculosis via Induced Autophagy. Microbiol Spectr 2023; 11:e0471122. [PMID: 37125940 PMCID: PMC10269511 DOI: 10.1128/spectrum.04711-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/11/2023] [Indexed: 05/02/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) continues to pose a significant threat to global health because it causes granulomas and systemic inflammatory responses during active tuberculosis (TB). Mtb can induce macrophage pyroptosis, which results in the release of IL-1β and causes tissue damage, thereby promoting its spread. In the absence of anti-TB drugs, host-directed therapy (HDT) has been demonstrated to be an effective strategy against TB. In this study, we used an in vitro Mtb-infected macrophage model to assess the effect of baicalein, derived from Scutellariae radix, on pyroptosis induced in Mtb-infected macrophages. Further, we investigated the molecular mechanisms underlying the actions of baicalein. The results of the study suggest that baicalein inhibits pyroptosis in Mtb-infected macrophages by downregulating the assembly of AIM2 and NLRP3 inflammasome and promoting autophagy. Further research has also shown that the mechanism by which baicalein promotes autophagy may involve the inhibition of the activation of the Akt/mTOR pathway and the inhibition of the AIM2 protein, which affects the levels of CHMP2A protein required to promote autophagy. Thus, our data show that baicalein can inhibit Mtb infection-induced macrophage pyroptosis and has the potential to be a new adjunctive HDT drug. IMPORTANCE Current strategies for treating drug-resistant tuberculosis have limited efficacy and undesirable side effects; hence, research on new treatments, including innovative medications, is required. Host-directed therapy (HDT) has emerged as a viable strategy for modulating host cell responses in order to enhance protective immunity against infections. Baicalein, extracted from Scutellariae radix, was shown to inhibit pyroptosis caused by Mycobacterium tuberculosis-infected macrophages and was associated with autophagy. Our findings reveal that baicalein can be used as an adjunctive treatment for tuberculosis or other inflammatory diseases by regulating immune function and enhancing the antibacterial ability of the host. It also provides a new idea for exploring the anti-inflammatory mechanism of baicalein.
Collapse
Affiliation(s)
- Bangzuo Ning
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingjing Shen
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fanglin Liu
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hemin Zhang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Jiang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
Sun J, Zhang Q, Yang G, Li Y, Fu Y, Zheng Y, Jiang X. The licorice flavonoid isoliquiritigenin attenuates Mycobacterium tuberculosis-induced inflammation through Notch1/NF-κB and MAPK signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115368. [PMID: 35589023 DOI: 10.1016/j.jep.2022.115368] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/24/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The genus Glycyrrhiza is a small perennial herb that has been traditionally used to treat many diseases across the world. Licorice (Gancao in Chinese) is the dried root and rhizome of G. glabra, G. uralensis or G. inflata. Licorice plays an important role in traditional Chinese medicine (TCM), and is the most frequently used in Chinese herbal formulas. Isoliquiritigenin (ISL) is a flavonoid extracted from licorice, and has been evaluated for its various biological activities, including anti-inflammatory, anti-tumor and anti-oxidant activities. Excessive and persistent inflammation in the Mycobacterium tuberculosis (Mtb) infection is not conducive to the elimination of Mtb, but contributes to serious pulmonary dysfunction. AIM OF THE STUDY This study aimed to examine the anti-inflammatory effects of ISL in the Mtb infection. METHODS In vitro models of Mtb-infected macrophages were established. Murine macrophage Raw 264.7 cells and primary peritoneal macrophages were used in this study. Cell viability was determined by the cell counting kit-8 (CCK-8) assay. The effects of ISL on the secretion levels of interleukin -1β (IL-1β), tumor necrosis factor -α (TNF-α), and interleukin -6 (IL-6) were detected by the enzyme-linked immunosorbent assay (ELISA). The expression levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX2) were measured by the real time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and Western blot. Western blot was used to assess the effects of ISL on the activation of NLRP3 inflammasome and Notch1/NF-κB and MAPK signaling pathways. Immunofluorescence assays was used to detected the translocation of phosphorylation of p65 subunit of NF-κB. RESULTS It was revealed that ISL inhibited the secretion of IL-1β and the activation of pore-forming protein (gasdermin D, GSDMD) by suppressing the activation of NLPR3 inflammasome induced by Mtb infection. ISL was also shown to have promising inhibitory effects on inflammatory factors, such as TNF-α, IL-6, iNOS and COX2. Regarding the anti-inflammatory mechanism of ISL, it was found that ISL exerted its anti-inflammatory effects by inhibiting the activation of Notch1/NF-κB and MAPK signaling pathways. CONCLUSION ISL reduced Mtb-induced inflammation through the Notch1/NF-κB and MAPK signaling pathways. ISL might be used as a potential adjuvant drug to treat tuberculosis by adjusting host immune responses.
Collapse
Affiliation(s)
- Jinxia Sun
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China
| | - Qingwen Zhang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China; Department of Inspection and Quarantine, School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Guizhen Yang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China
| | - Yinhong Li
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China
| | - Yan Fu
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China
| | - Yuejuan Zheng
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China.
| | - Xin Jiang
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, PR China.
| |
Collapse
|
8
|
García Clemente M, Olveira C, Girón R, Máiz L, Sibila O, Golpe R, Menéndez R, Rodríguez J, Barreiro E, Rodríguez Hermosa JL, Prados C, De la Rosa D, Carbajal CM, Solís M, Martínez-García MÁ. Impact of Chronic Bronchial Infection by Staphylococcus aureus on Bronchiectasis. J Clin Med 2022; 11:3960. [PMID: 35887723 PMCID: PMC9319377 DOI: 10.3390/jcm11143960] [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: 05/27/2022] [Revised: 06/27/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
The objective of the study was to analyze the factors associated with chronic bronchial infection (CBI) due to methicillin-susceptible Staphylococcus aureus (SA) and assess the clinical impact on severity, exacerbations, hospitalizations, and loss of lung function compared to patients with no isolation of PPMs in a large longitudinal series of patients from the Spanish bronchiectasis registry (RIBRON). Material and methods: A prospective, longitudinal, multicenter study was conducted with patients included in the RIBRON registry between January 2015 and October 2020. The inclusion criteria were an age of 18 years or older and an initial diagnosis of bronchiectasis. Patients recorded in the registry had a situation of clinical stability in the absence of an exacerbation in the four weeks before their inclusion. All patients were encouraged to provide a sputum sample at each visit for microbiological culture. Annual pulmonary function tests were performed according to the national spirometry guidelines. Results: A total of 426 patients were ultimately included in the study: 77 patients (18%) with CBI due to SA and 349 (82%) who did not present any isolation of PPMs in sputum. The mean age was 66.9 years (16.2), and patients 297 (69.7%) were female, with an average BMI of 25.1 (4.7) kg/m2 and an average Charlson index of 1.74 (1.33). The mean baseline value of FEV1 2 L was 0.76, with a mean FEV1% of 78.8% (23.1). One hundred and seventy-two patients (40.4%) had airflow obstruction with FEV1/FVC < 0.7. The mean predictive FACED score was 1.62 (1.41), with a mean value of 2.62 (2.07) for the EFACED score and 7.3 (4.5) for the BSI score. Patients with CBI caused by SA were younger (p < 0.0001), and they had a lower BMI (p = 0.024) and more exacerbations in the previous year (p = 0.019), as well as in the first, second, and third years of follow-up (p = 0.020, p = 0.001, and p = 0.018, respectively). As regards lung function, patients with CBI due to SA had lower levels of FEV1% at the time of inclusion in the registry (p = 0.021), and they presented more frequently with bronchial obstruction (p = 0.042). A lower age (OR: 0.97; 95% CI: 0.94−0.99; p < 0.001), lower FEV1 value% (OR: 0.98; 95% CI: 0.97−0.99; p = 0.035), higher number of affected lobes (OR: 1.53; 95% CI: 1.2−1.95; p < 0.001), and the presence of two or more exacerbations in the previous year (OR: 2.33; 95% CI: 1.15−4.69; p = 0.018) were observed as independent factors associated with CBI due to SA. The reduction in FEv1% in all patients included in the study was −0.31%/year (95% CI: −0.7; −0.07) (p = 0.110). When the reduction in FEv1% is analyzed in the group of patients with CBI due to SA and the group without pathogens, we observed that the reduction in FEV1% was −1.19% (95% CI: −2.09, −0.69) (p < 0.001) in the first group and −0.02% (95% CI: −0.07, −0.01) (p = 0.918) in the second group. According to a linear regression model (mixed effects) applied to determine which factors were associated with a more pronounced reduction in FEv1% in the overall group (including those with CBI due to SA and those with no PPM isolation), age (p = 0.0019), use of inhaled corticosteroids (p = 0.004), presence of CBI due to SA (p = 0.007), female gender (p < 0.001), and the initial value of FEV1 (p < 0.001) were significantly related. Conclusions: Patients with non-CF bronchiectasis with CBI due to SA were younger, with lower FEV1% values, more significant extension of bronchiectasis, and a higher number of exacerbations of mild to moderate symptoms than those with no PPM isolation in respiratory secretions. The reduction in FEV1% was −1.19% (95% CI: −2.09, −0.69) (p < 0.001) in patients with CBI caused by SA.
Collapse
Affiliation(s)
- Marta García Clemente
- Respiratory Department, Central University Hospital, 33011 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Casilda Olveira
- Respiratory Department, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain;
- Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29003 Málaga, Spain
| | - Rosa Girón
- Respiratory Department, Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, 28015 Madrid, Spain; (R.G.); (M.S.)
| | - Luis Máiz
- Respiratory Department, Hospital Ramón y Cajal, 28015 Madrid, Spain;
| | - Oriol Sibila
- Respiratory Department, Hospital Clinic, 08035 Barcelona, Spain;
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 28015 Madrid, Spain;
| | - Rafael Golpe
- Respiratory Department, Hospital Lucus Augusti, 27080 Lugo, Spain;
| | - Rosario Menéndez
- Respiratory Department, Hospital Universitario y Politécnico La Fe, 46003 Valencia, Spain;
| | - Juan Rodríguez
- Respiratory Department, Hospital San Agustín, 33401 Avilés, Spain;
| | - Esther Barreiro
- Respiratory Department, Hospital del Mar-IMIM, Parc de Salut Mar, 08003 Barcelona, Spain;
| | - Juan Luis Rodríguez Hermosa
- Pulmonary Department, Research Institute of Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain;
- Respiratory Department, Hospital Clinico San Carlos, 28015 Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, 28015 Madrid, Spain
| | | | - David De la Rosa
- Respiratory Department, Hospital Santa Creu I Sant Pau, 08041 Barcelona, Spain;
| | | | - Marta Solís
- Respiratory Department, Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, 28015 Madrid, Spain; (R.G.); (M.S.)
| | - Miguel Ángel Martínez-García
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 28015 Madrid, Spain;
- Respiratory Department, Hospital Universitario y Politécnico La Fe, 46003 Valencia, Spain;
| |
Collapse
|
9
|
Bhandari V, Suresh A. Next-Generation Approaches Needed to Tackle Antimicrobial Resistance for the Development of Novel Therapies Against the Deadly Pathogens. Front Pharmacol 2022; 13:838092. [PMID: 35721221 PMCID: PMC9205218 DOI: 10.3389/fphar.2022.838092] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
The advent of antimicrobials was a miracle that saved millions of lives across the globe. With the discovery of penicillin, varieties of other antimicrobials came into play one after another. However, the injudicious use of antimicrobials for therapeutics and prophylactics and overuse in agriculture and animal husbandry industries resulted in its gloominess and rise of antimicrobial resistance. The microbes have slowly outsmarted the human race with diverse mechanisms to evade the antimicrobial effects of the drugs in use. The review aims to discuss the emergence of resistance in bacterial species with time and the various means by which bacterial cells had safeguarded themselves. In addition to that, we have also highlighted new approaches currently used to tackle antimicrobial resistance or practices that could be useful in identifying new treatment options.
Collapse
Affiliation(s)
- Vasundhra Bhandari
- National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad, Hyderabad, India
| | - Akash Suresh
- National Institute of Animal Biotechnology, Hyderabad, India.,Manipal Academy of Higher Education (MAHE), Hyderabad, India
| |
Collapse
|
10
|
Use of Inhaled Corticosteroids and Risk of Acquiring Haemophilus influenzae in Patients with Chronic Obstructive Pulmonary Disease. J Clin Med 2022; 11:jcm11123539. [PMID: 35743610 PMCID: PMC9225538 DOI: 10.3390/jcm11123539] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Inhaled corticosteroids (ICS) are widely used in chronic obstructive pulmonary disease (COPD), despite the known risk of severe adverse effects including pulmonary infections. Research Question: Our study investigates the risk of acquiring a positive Haemophilus influenzae airway culture with use of ICS in outpatients with COPD. Study Design and Methods: We conducted an epidemiological cohort study using data from 1 January 2010 to 19 February 2018, including 21,218 outpatients with COPD in Denmark. ICS use 365 days prior to cohort entry was categorised into low, moderate, and high, based on cumulated ICS dose extracted from a national registry on reimbursed prescriptions. A Cox proportional hazards regression model was used to assess the future risk of acquiring H. Influenzae within 365 days from cohort entry, and sensitivity analyses were performed using propensity score matched models. Results: In total, 801 (3.8%) patients acquired H. Influenzae during follow-up. Use of ICS was associated with a dose-dependent increased risk of acquiring H. Influenzae with hazard ratio (HR) 1.2 (95% confidence interval (CI) 0.9−1.5, p value = 0.1) for low-dose ICS; HR 1.7 (95% CI 1.3−2.1, p value < 0.0001) for moderate dose; and HR 1.9 (95% CI 1.5−2.4, p value < 0.0001) for high-dose ICS compared to no ICS use. Results were confirmed in the propensity-matched model using the same categories. Conclusions: ICS use in outpatients with COPD was associated with a dose-dependent increase in risk of isolating H. Influenzae. This observation supports that high dose ICS should be used with caution.
Collapse
|
11
|
Yao Y, Liu H, Yuan L, Du X, Yang Y, Zhou K, Wu X, Qin L, Yang M, Xiang Y, Qu X, Qin X, Liu C. Integrins are double-edged swords in pulmonary infectious diseases. Biomed Pharmacother 2022; 153:113300. [PMID: 35728353 DOI: 10.1016/j.biopha.2022.113300] [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: 04/20/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Integrins are an important family of adhesion molecules that are widely distributed on immune cells in the lungs. Of note, accumulating evidences have shown that integrins are double-edged swords in pulmonary infectious diseases. On one hand, integrins promote the migration of immune cells to remove the invaded pathogens in the infected lungs. However, on the other hand, integrins also act as the targets for pathogens to escape from host immune system, which is a potential factor leading to further tissue damage. Thus, the innovative therapeutic strategies based on integrins has inspired well-founded hopes to treat pulmonary infectious diseases. In this review, we illustrate the involvement of integrins in pulmonary infectious diseases, and further discuss the innovative therapeutic targets based on integrins.
Collapse
Affiliation(s)
- Ye Yao
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Yu Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Kai Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xinyu Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, Australia
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Chi Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Research Center of China-Africa Infectious Diseases, Xiangya School of Medicine Central South University, Changsha, Hunan, China.
| |
Collapse
|
12
|
Baindara P, Agrawal S, Franco OL. Host-directed therapies for malaria and tuberculosis: common infection strategies and repurposed drugs. Expert Rev Anti Infect Ther 2022; 20:849-869. [DOI: 10.1080/14787210.2022.2044794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Piyush Baindara
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Missouri, Columbia, MO, USA
| | - Sonali Agrawal
- Immunology Division, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, Uttar Pradesh, India
| | - O. L. Franco
- Proteomics Analysis and Biochemical Center, Catholic University of Brasilia, Brasilia, Brazil; S-Inova Biotech, Catholic University Dom Bosco, Campo Grande, MS, Brazil
| |
Collapse
|
13
|
Abstract
Host-directed therapy (HDT) is gaining traction as a strategy to combat infectious diseases caused by viruses and intracellular bacteria, but its implementation in the context of parasitic diseases has received less attention. Here, we provide a brief overview of this field and advocate HDT as a promising strategy for antimalarial intervention based on untapped targets. HDT provides a basis from which repurposed drugs could be rapidly deployed and is likely to strongly limit the emergence of resistance. This strategy can be applied to any intracellular pathogen and is particularly well placed in situations in which rapid identification of treatments is needed, such as emerging infections and pandemics, as starkly illustrated by the current COVID-19 crisis.
Collapse
|
14
|
Borah P, Deb PK, Venugopala KN, Al-Shar'i NA, Singh V, Deka S, Srivastava A, Tiwari V, Mailavaram RP. Tuberculosis: An Update on Pathophysiology, Molecular Mechanisms of Drug Resistance, Newer Anti-TB Drugs, Treatment Regimens and Host- Directed Therapies. Curr Top Med Chem 2021; 21:547-570. [PMID: 33319660 DOI: 10.2174/1568026621999201211200447] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/16/2020] [Accepted: 11/19/2020] [Indexed: 11/22/2022]
Abstract
Human tuberculosis (TB) is primarily caused by Mycobacterium tuberculosis (Mtb) that inhabits inside and amidst immune cells of the host with adapted physiology to regulate interdependent cellular functions with intact pathogenic potential. The complexity of this disease is attributed to various factors such as the reactivation of latent TB form after prolonged persistence, disease progression specifically in immunocompromised patients, advent of multi- and extensivelydrug resistant (MDR and XDR) Mtb strains, adverse effects of tailor-made regimens, and drug-drug interactions among anti-TB drugs and anti-HIV therapies. Thus, there is a compelling demand for newer anti-TB drugs or regimens to overcome these obstacles. Considerable multifaceted transformations in the current TB methodologies and molecular interventions underpinning hostpathogen interactions and drug resistance mechanisms may assist to overcome the emerging drug resistance. Evidently, recent scientific and clinical advances have revolutionised the diagnosis, prevention, and treatment of all forms of the disease. This review sheds light on the current understanding of the pathogenesis of TB disease, molecular mechanisms of drug-resistance, progress on the development of novel or repurposed anti-TB drugs and regimens, host-directed therapies, with particular emphasis on underlying knowledge gaps and prospective for futuristic TB control programs.
Collapse
Affiliation(s)
- Pobitra Borah
- Pratiksha Institute of Pharmaceutical Sciences, Chandrapur Road, Panikhaiti, Guwahati-26, Assam, India
| | - Pran K Deb
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Philadelphia University, PO Box 1, Amman 19392, Jordan
| | - Katharigatta N Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Nizar A Al-Shar'i
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Vinayak Singh
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, 7701, South Africa
| | - Satyendra Deka
- Pratiksha Institute of Pharmaceutical Sciences, Chandrapur Road, Panikhaiti, Guwahati-26, Assam, India
| | - Amavya Srivastava
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221 005, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Lab, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221 005, India
| | - Raghu P Mailavaram
- Department of Pharmaceutical Chemistry, Shri Vishnu College of Pharmacy, Vishnupur, Bhimavaram - 534 202, West Godavari Dist., Andhra Pradesh, India
| |
Collapse
|
15
|
Stephanie F, Saragih M, Tambunan USF. Recent Progress and Challenges for Drug-Resistant Tuberculosis Treatment. Pharmaceutics 2021; 13:pharmaceutics13050592. [PMID: 33919204 PMCID: PMC8143172 DOI: 10.3390/pharmaceutics13050592] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 11/23/2022] Open
Abstract
Control of Mycobacterium tuberculosis infection continues to be an issue, particularly in countries with a high tuberculosis (TB) burden in the tropical and sub-tropical regions. The effort to reduce the catastrophic cost of TB with the WHO’s End TB Strategy in 2035 is still obstructed by the emergence of drug-resistant TB (DR-TB) cases as result of various mutations of the MTB strain. In the approach to combat DR-TB, several potential antitubercular agents were discovered as inhibitors for various existing and novel targets. Host-directed therapy and immunotherapy also gained attention as the drug-susceptibility level of the pathogen can be reduced due to the pathogen’s evolutionary dynamics. This review is focused on the current progress and challenges in DR-TB treatment. We briefly summarized antitubercular compounds that are under development and trials for both DR-TB drug candidates and host-directed therapy. We also highlighted several problems in DR-TB diagnosis, the treatment regimen, and drug discovery that have an impact on treatment adherence and treatment failure.
Collapse
|
16
|
Etna MP, Severa M, Licursi V, Pardini M, Cruciani M, Rizzo F, Giacomini E, Macchia G, Palumbo O, Stallone R, Carella M, Livingstone M, Negri R, Pellegrini S, Coccia EM. Genome-Wide Gene Expression Analysis of Mtb-Infected DC Highlights the Rapamycin-Driven Modulation of Regulatory Cytokines via the mTOR/GSK-3β Axis. Front Immunol 2021; 12:649475. [PMID: 33936070 PMCID: PMC8086600 DOI: 10.3389/fimmu.2021.649475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
In human primary dendritic cells (DC) rapamycin-an autophagy inducer and protein synthesis inhibitor-overcomes the autophagy block induced by Mycobacterium tuberculosis (Mtb) and promotes a Th1 response via IL-12 secretion. Here, the immunostimulatory activity of rapamycin in Mtb-infected DC was further investigated by analyzing both transcriptome and translatome gene profiles. Hundreds of differentially expressed genes (DEGs) were identified by transcriptome and translatome analyses of Mtb-infected DC, and some of these genes were found further modulated by rapamycin. The majority of transcriptome-associated DEGs overlapped with those present in the translatome, suggesting that transcriptionally stimulated mRNAs are also actively translated. In silico analysis of DEGs revealed significant changes in intracellular cascades related to cytokine production, cytokine-induced signaling and immune response to pathogens. In particular, rapamycin treatment of Mtb-infected DC caused an enrichment of IFN-β, IFN-λ and IFN-stimulated gene transcripts in the polysome-associated RNA fraction. In addition, rapamycin led to an increase of IL-12, IL-23, IL-1β, IL-6, and TNF-α but to a reduction of IL-10. Interestingly, upon silencing or pharmacological inhibition of GSK-3β, the rapamycin-driven modulation of the pro- and anti-inflammatory cytokine balance was lost, indicating that, in Mtb-infected DC, GSK-3β acts as molecular switch for the regulation of the cytokine milieu. In conclusion, our study sheds light on the molecular mechanism by which autophagy induction contributes to DC activation during Mtb infection and points to rapamycin and GSK-3β modulators as promising compounds for host-directed therapy in the control of Mtb infection.
Collapse
Affiliation(s)
- Marilena P Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Valerio Licursi
- Department of Biology and Biotechnology, Sapienza University, Rome, Italy
| | - Manuela Pardini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Melania Cruciani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Giacomini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Orazio Palumbo
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Raffaella Stallone
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Massimo Carella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Mark Livingstone
- Cytokine Signaling Unit, Inserm, Institut Pasteur, Paris, France
| | - Rodolfo Negri
- Department of Biology and Biotechnology, Sapienza University, Rome, Italy
| | | | - Eliana M Coccia
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
17
|
Nienaber A, Hayford FEA, Variava E, Martinson N, Malan L. The Manipulation of the Lipid Mediator Metabolism as Adjunct Host-Directed Therapy in Tuberculosis. Front Immunol 2021; 12:623941. [PMID: 33777003 PMCID: PMC7994275 DOI: 10.3389/fimmu.2021.623941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Host-directed therapies (HDTs) enhance the host response to tuberculosis (TB) infection to reduce disease severity. For instance, the manipulation of lipid mediator production diminishes the hyperactive immune response which is a known pathological feature of TB that generates lung tissue damage. Non-steroidal anti-inflammatory drugs (NSAIDs) and omega-3 long-chain polyunsaturated fatty acids (n-3 LCPUFA) are examples of such HDTs. In this mini-review, we recapitulate the literature available on the effects of NSAIDs and n-3 LCPUFA in TB as well as the immunological pathways underpinning these effects. Many NSAIDs have a great deal of data describing their effects and safety and in many jurisdictions are inexpensive, and sold over the counter in neighborhood convenience stores and supermarkets. The potential benefits of NSAIDs in TB are well-documented in pre-clinical studies. The reduction of pro-inflammatory lipid mediator production by inhibiting cyclooxygenase (COX) pathways with NSAIDs has been found to improve lung histopathology, bacterial control, and survival. Additionally, n-3 LCPUFA and its novel bioactive metabolites produced by COX and lipoxygenase (LOX) have been identified as safe and effective pro-resolving and antibacterial pharmaconutrients. Nevertheless, heterogeneous results have been reported in pre-clinical TB studies. Recently, the importance of the correct timing of NSAIDs and n-3 LCPUFA administration in TB has also been highlighted. This mini-review will provide a better understanding of the potential contribution of these therapies toward reducing inflammatory lung damage and improving bactericidal activity, especially during later stages of TB infection. It further highlights that clinical trials are required to confirm benefit and safety in TB patients.
Collapse
Affiliation(s)
- Arista Nienaber
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Frank E A Hayford
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa.,Department of Nutrition and Dietetics, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Ebrahim Variava
- Perinatal HIV Research Unit, University of Witwatersrand, Soweto, South Africa.,Department of Internal Medicine, Klerksdorp Tshepong Hospital Complex, North West Department of Health, Klerksdorp, South Africa
| | - Neil Martinson
- Perinatal HIV Research Unit, University of Witwatersrand, Soweto, South Africa
| | - Linda Malan
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| |
Collapse
|
18
|
Mishra R, Krishnamoorthy P, Kumar H. MicroRNA-30e-5p Regulates SOCS1 and SOCS3 During Bacterial Infection. Front Cell Infect Microbiol 2021; 10:604016. [PMID: 33585275 PMCID: PMC7873556 DOI: 10.3389/fcimb.2020.604016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Host innate immunity is the major player against continuous microbial infection. Various pathogenic bacteria adopt the strategies to evade the immunity and show resistance toward the various established therapies. Despite the advent of many antibiotics for bacterial infections, there is a substantial need for the host-directed therapies (HDTs) to combat the infection. HDTs are recently being adopted to be useful in eradicating intracellular bacterial infection. Changing the innate immune responses of the host cells alters pathogen's ability to reside inside the cell. MicroRNAs are the small non-coding endogenous molecules and post-transcriptional regulators to target the 3'UTR of the messenger RNA. They are reported to modulate the host's immune responses during bacterial infections. Exploiting microRNAs as a therapeutic candidate in HDTs upon bacterial infection is still in its infancy. Here, initially, we re-analyzed the publicly available transcriptomic dataset of macrophages, infected with different pathogenic bacteria and identified significant genes and microRNAs common to the differential infections. We thus identified and miR-30e-5p, to be upregulated in different bacterial infections which enhances innate immunity to combat bacterial replication by targeting key negative regulators such as SOCS1 and SOCS3 of innate immune signaling pathways. Therefore, we propose miR-30e-5p as one of the potential candidates to be considered for additional clinical validation toward HDTs.
Collapse
Affiliation(s)
- Richa Mishra
- Laboratory of Immunology and Infectious Disease Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India
| | - Pandikannan Krishnamoorthy
- Laboratory of Immunology and Infectious Disease Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India
| | - Himanshu Kumar
- Laboratory of Immunology and Infectious Disease Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India.,WPI Immunology, Frontier Research Centre, Osaka University, Osaka, Japan
| |
Collapse
|
19
|
Dalberto PF, de Souza EV, Abbadi BL, Neves CE, Rambo RS, Ramos AS, Macchi FS, Machado P, Bizarro CV, Basso LA. Handling the Hurdles on the Way to Anti-tuberculosis Drug Development. Front Chem 2020; 8:586294. [PMID: 33330374 PMCID: PMC7710551 DOI: 10.3389/fchem.2020.586294] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/25/2020] [Indexed: 12/17/2022] Open
Abstract
The global epidemic of tuberculosis (TB) imposes a sustained epidemiologic vigilance and investments in research by governments. Mycobacterium tuberculosis, the main causative agent of TB in human beings, is a very successful pathogen, being the main cause of death in the population among infectious agents. In 2018, ~10 million individuals were contaminated with this bacillus and became ill with TB, and about 1.2 million succumbed to the disease. Most of the success of the M. tuberculosis to linger in the population comes from its ability to persist in an asymptomatic latent state into the host and, in fact, the majority of the individuals are unaware of being contaminated. Even though TB is a treatable disease and is curable in most cases, the treatment is lengthy and laborious. In addition, the rise of resistance to first-line anti-TB drugs elicits a response from TB research groups to discover new chemical entities, preferably with novel mechanisms of action. The pathway to find a new TB drug, however, is arduous and has many barriers that are difficult to overcome. Fortunately, several approaches are available today to be pursued by scientists interested in anti-TB drug development, which goes from massively testing chemical compounds against mycobacteria, to discovering new molecular targets by genetic manipulation. This review presents some difficulties found along the TB drug development process and illustrates different approaches that might be used to try to identify new molecules or targets that are able to impair M. tuberculosis survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Luiz A. Basso
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
20
|
Baindara P, Mandal SM. Bacteria and bacterial anticancer agents as a promising alternative for cancer therapeutics. Biochimie 2020; 177:164-189. [PMID: 32827604 DOI: 10.1016/j.biochi.2020.07.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/04/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022]
Abstract
Cancer is the leading cause of deaths worldwide, though significant advances have occurred in its diagnosis and treatment. The development of resistance against chemotherapeutic agents, their side effects, and non-specific toxicity urge to screen for the novel anticancer agent. Hence, the development of novel anticancer agents with a new mechanism of action has become a major scientific challenge. Bacteria and bacterially produced bioactive compounds have recently emerged as a promising alternative for cancer therapeutics. Bacterial anticancer agents such as antibiotics, bacteriocins, non-ribosomal peptides, polyketides, toxins, etc. These are adopted different mechanisms of actions such as apoptosis, necrosis, reduced angiogenesis, inhibition of translation and splicing, and obstructing essential signaling pathways to kill cancer cells. Also, live tumor-targeting bacteria provided a unique therapeutic alternative for cancer treatment. This review summarizes the anticancer properties and mechanism of actions of the anticancer agents of bacterial origin and antitumor bacteria along with their possible future applications in cancer therapeutics.
Collapse
Affiliation(s)
- Piyush Baindara
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA.
| | - Santi M Mandal
- Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur, 721302, WB, India.
| |
Collapse
|
21
|
Abstract
Coronavirus disease 2019 (COVID-19) characterized by immuno-pathological host responses including pneumonia, lymphopenia, and cytokine storm that leads to severe lung inflammation, developed in acute respiratory distress syndrome (ARDS). In the absence of an effective vaccine or any definitive cure, the use of host-directed therapies is an effective alternative and demanding treatment option in the current pandemic outbreak of COVID-19.
Collapse
Affiliation(s)
- Piyush Baindara
- Department of Molecular Microbiology and Immunology, University of Missouri , Columbia, MO, USA
| | - Sonali Agrawal
- Immunology Division, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases , Agra, India
| | - Santi M Mandal
- Central Research Facility, Indian Institute of Technology Kharagpur , Kharagpur, India
| |
Collapse
|
22
|
Gopalaswamy R, Shanmugam S, Mondal R, Subbian S. Of tuberculosis and non-tuberculous mycobacterial infections - a comparative analysis of epidemiology, diagnosis and treatment. J Biomed Sci 2020; 27:74. [PMID: 32552732 PMCID: PMC7297667 DOI: 10.1186/s12929-020-00667-6] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/05/2020] [Indexed: 12/26/2022] Open
Abstract
Pulmonary diseases due to mycobacteria cause significant morbidity and mortality to human health. In addition to tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), recent epidemiological studies have shown the emergence of non-tuberculous mycobacteria (NTM) species in causing lung diseases in humans. Although more than 170 NTM species are present in various environmental niches, only a handful, primarily Mycobacterium avium complex and M. abscessus, have been implicated in pulmonary disease. While TB is transmitted through inhalation of aerosol droplets containing Mtb, generated by patients with symptomatic disease, NTM disease is mostly disseminated through aerosols originated from the environment. However, following inhalation, both Mtb and NTM are phagocytosed by alveolar macrophages in the lungs. Subsequently, various immune cells are recruited from the circulation to the site of infection, which leads to granuloma formation. Although the pathophysiology of TB and NTM diseases share several fundamental cellular and molecular events, the host-susceptibility to Mtb and NTM infections are different. Striking differences also exist in the disease presentation between TB and NTM cases. While NTM disease is primarily associated with bronchiectasis, this condition is rarely a predisposing factor for TB. Similarly, in Human Immunodeficiency Virus (HIV)-infected individuals, NTM disease presents as disseminated, extrapulmonary form rather than as a miliary, pulmonary disease, which is seen in Mtb infection. The diagnostic modalities for TB, including molecular diagnosis and drug-susceptibility testing (DST), are more advanced and possess a higher rate of sensitivity and specificity, compared to the tools available for NTM infections. In general, drug-sensitive TB is effectively treated with a standard multi-drug regimen containing well-defined first- and second-line antibiotics. However, the treatment of drug-resistant TB requires the additional, newer class of antibiotics in combination with or without the first and second-line drugs. In contrast, the NTM species display significant heterogeneity in their susceptibility to standard anti-TB drugs. Thus, the treatment for NTM diseases usually involves the use of macrolides and injectable aminoglycosides. Although well-established international guidelines are available, treatment of NTM disease is mostly empirical and not entirely successful. In general, the treatment duration is much longer for NTM diseases, compared to TB, and resection surgery of affected organ(s) is part of treatment for patients with NTM diseases that do not respond to the antibiotics treatment. Here, we discuss the epidemiology, diagnosis, and treatment modalities available for TB and NTM diseases of humans.
Collapse
Affiliation(s)
- Radha Gopalaswamy
- Department of Bacteriology, National Institute for Research in Tuberculosis, Chennai, India
| | - Sivakumar Shanmugam
- Department of Bacteriology, National Institute for Research in Tuberculosis, Chennai, India
| | - Rajesh Mondal
- Department of Bacteriology, National Institute for Research in Tuberculosis, Chennai, India
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States.
| |
Collapse
|
23
|
Winchell CG, Mishra BB, Phuah JY, Saqib M, Nelson SJ, Maiello P, Causgrove CM, Ameel CL, Stein B, Borish HJ, White AG, Klein EC, Zimmerman MD, Dartois V, Lin PL, Sassetti CM, Flynn JL. Evaluation of IL-1 Blockade as an Adjunct to Linezolid Therapy for Tuberculosis in Mice and Macaques. Front Immunol 2020; 11:891. [PMID: 32477361 PMCID: PMC7235418 DOI: 10.3389/fimmu.2020.00891] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/17/2020] [Indexed: 12/13/2022] Open
Abstract
In 2017 over 550,000 estimated new cases of multi-drug/rifampicin resistant tuberculosis (MDR/RR-TB) occurred, emphasizing a need for new treatment strategies. Linezolid (LZD) is a potent antibiotic for drug-resistant Gram-positive infections and is an effective treatment for TB. However, extended LZD use can lead to LZD-associated host toxicities, most commonly bone marrow suppression. LZD toxicities may be mediated by IL-1, an inflammatory pathway important for early immunity during M. tuberculosis infection. However, IL-1 can contribute to pathology and disease severity late in TB progression. Since IL-1 may contribute to LZD toxicity and does influence TB pathology, we targeted this pathway with a potential host-directed therapy (HDT). We hypothesized LZD efficacy could be enhanced by modulation of IL-1 pathway to reduce bone marrow toxicity and TB associated-inflammation. We used two animal models of TB to test our hypothesis, a TB-susceptible mouse model and clinically relevant cynomolgus macaques. Antagonizing IL-1 in mice with established infection reduced lung neutrophil numbers and partially restored the erythroid progenitor populations that are depleted by LZD. In macaques, we found no conclusive evidence of bone marrow suppression associated with LZD, indicating our treatment time may have been short enough to avoid the toxicities observed in humans. Though treatment was only 4 weeks (the FDA approved regimen at the time of study), we observed sterilization of the majority of granulomas regardless of co-administration of the FDA-approved IL-1 receptor antagonist (IL-1Rn), also known as Anakinra. However, total lung inflammation was significantly reduced in macaques treated with IL-1Rn and LZD compared to LZD alone. Importantly, IL-1Rn administration did not impair the host response against Mtb or LZD efficacy in either animal model. Together, our data support that inhibition of IL-1 in combination with LZD has potential to be an effective HDT for TB and the need for further research in this area.
Collapse
Affiliation(s)
- Caylin G. Winchell
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Bibhuti B. Mishra
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Jia Yao Phuah
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Mohd Saqib
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Samantha J. Nelson
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Chelsea M. Causgrove
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Cassaundra L. Ameel
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Brianne Stein
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - H. Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Alexander G. White
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Edwin C. Klein
- Division of Laboratory Animal Research, University of Pittsburgh, Pittsburgh, PA, United States
| | - Matthew D. Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Philana Ling Lin
- Department of Pediatrics, UPMC Children's Hospital of the University of Pittsburgh, Pittsburgh, PA, United States
| | - Christopher M. Sassetti
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
24
|
Mycobacterium tuberculosis Shikimate Pathway Enzymes as Targets for the Rational Design of Anti-Tuberculosis Drugs. Molecules 2020; 25:molecules25061259. [PMID: 32168746 PMCID: PMC7144000 DOI: 10.3390/molecules25061259] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 12/17/2022] Open
Abstract
Roughly a third of the world’s population is estimated to have latent Mycobacterium tuberculosis infection, being at risk of developing active tuberculosis (TB) during their lifetime. Given the inefficacy of prophylactic measures and the increase of drug-resistant M. tuberculosis strains, there is a clear and urgent need for the development of new and more efficient chemotherapeutic agents, with selective toxicity, to be implemented on patient treatment. The component enzymes of the shikimate pathway, which is essential in mycobacteria and absent in humans, stand as attractive and potential targets for the development of new drugs to treat TB. This review gives an update on published work on the enzymes of the shikimate pathway and some insight on what can be potentially explored towards selective drug development.
Collapse
|
25
|
Baindara P, Ghosh AK, Mandal SM. Coevolution of Resistance Against Antimicrobial Peptides. Microb Drug Resist 2020; 26:880-899. [PMID: 32119634 DOI: 10.1089/mdr.2019.0291] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Antimicrobial peptides (AMPs) are produced by all forms of life, ranging from eukaryotes to prokaryotes, and they are a crucial component of innate immunity, involved in clearing infection by inhibiting pathogen colonization. In the recent past, AMPs received high attention due to the increase of extensive antibiotic resistance by these pathogens. AMPs exhibit a diverse spectrum of activity against bacteria, fungi, parasites, and various types of cancer. AMPs are active against various bacterial pathogens that cause disease in animals and plants. However, because of the coevolution of host and pathogen interaction, bacteria have developed the mechanisms to sense and exhibit an adaptive response against AMPs. These resistance mechanisms are playing an important role in bacterial virulence within the host. Here, we have discussed the different resistance mechanisms used by gram-positive and gram-negative bacteria to sense and combat AMP actions. Understanding the mechanism of AMP resistance may provide directions toward the development of novel therapeutic strategies to control multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Piyush Baindara
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ananta K Ghosh
- Department of Biotechnology, Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Santi M Mandal
- Department of Biotechnology, Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|