1
|
Alrasheed AS, Alqadhibi MA, Khoja RH, Alayyaf AS, Alhumoudi DS, Aldawlan MI, Alghanmi BO, Almutairi FS, Bin-Mahfooz MA, Altalhi LA, Aldanyowi SN, Aleid AM, Alessa AA. Emerging therapies for immunomodulation in traumatic brain injury: A systematic review and meta-analysis. Surg Neurol Int 2024; 15:327. [PMID: 39372991 PMCID: PMC11450791 DOI: 10.25259/sni_502_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 08/12/2024] [Indexed: 10/08/2024] Open
Abstract
Background Traumatic brain injury (TBI) represents a significant global health burden, often leading to significant morbidity and mortality. Mounting evidence underscores the intricate involvement of dysregulated immune responses in TBI pathophysiology, highlighting the potential for immunomodulatory interventions to mitigate secondary injury cascades and enhance patient outcomes. Despite advancements in treatment modalities, optimizing therapeutic strategies remains a critical challenge in TBI management. To address this gap, this systematic review and meta-analysis aimed to rigorously evaluate the efficacy and safety of emerging immunomodulatory therapies in the context of TBI. Methods We searched electronic databases such as PubMed, Scopus, Web of Science and CENTRAL for relevant studies investigating the efficacy of immunomodulatory therapies in TBI that were meticulously selected for inclusion. Two independent reviewers meticulously performed data extraction and quality assessment, adhering to predefined criteria. Both randomized controlled trials (RCTs) and observational studies reporting clinically relevant outcomes, such as mortality rates, the Glasgow coma scale, and adverse events, were meticulously scrutinized. Meta-analysis techniques were employed to assess treatment effects across studies quantitatively and analyzed using the Review Manager software (version 5.2). Results Fourteen studies (n = 1 observational and n = 13 RCTs) were included in our study. Meta-analysis showed no significant overall mortality difference, but erythropoietin (EPO) significantly reduced mortality (odds ratio = 0.49; 95% confidence interval: 0.31-0.78, P = 0.002). The adverse event meta-analysis revealed no significant differences. Conclusion Immunomodulatory therapies did not significantly affect overall mortality, but EPO demonstrated promising results. Adverse events did not significantly differ from controls. Further research is warranted to refine TBI treatment protocols.
Collapse
Affiliation(s)
| | | | - Rammaz Hussam Khoja
- Department of Surgery, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Abdulaziz Saad Alayyaf
- Department of Surgery, College of Medicine, Prince Sattam bin Abdulaziz University, Riyadh, Saudi Arabia
| | - Duaa Saleh Alhumoudi
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mubarak Ibrahim Aldawlan
- Department of Surgery, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | | | | | - Mohammed Ali Bin-Mahfooz
- Department of Surgery, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Lina Abdulrahim Altalhi
- Department of Surgery, College of Medicine and Medical Science, Arabian Gulf University, Manama, Bahrain
| | - Saud Nayef Aldanyowi
- Department of Surgery, College of Medicine, King Faisal University, AlAhsa, Saudi Arabia
| | | | | |
Collapse
|
2
|
Lin CJ, Hsia NY, Hwang DK, Hwang YS, Chang YC, Hsu YR, Yeh PT, Lin CP, Hsu AY, Ho MW, Sheu SJ. Diagnosis and Treatment of Tubercular Uveitis in Taiwan - Consensus of Expert Panels. Ocul Immunol Inflamm 2024; 32:1420-1426. [PMID: 37186887 DOI: 10.1080/09273948.2023.2208660] [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: 02/11/2023] [Revised: 04/12/2023] [Accepted: 04/25/2023] [Indexed: 05/17/2023]
Abstract
There is currently a lack of guidelines with regard to tubercular uveitis (TBU) management in Taiwan. We therefore propose an evidence-based consensus on the management for TBU. The Taiwan Ocular Inflammation Society conducted a meeting that included nine ophthalmologist and one infection disease expert that focused on three broad areas of (1) nomenclature for TBU, (2) assessment and diagnosis for TBU, and (3) treatment of TBU. Brief literature review on TBU diagnosis and management was conducted that informed this panel meeting in order to make decisions on each consensus statements. In terms of our results, a consensus statements and recommendations for the diagnosis and management of TBU were developed. This consensus statement provides an algorithmic approach toward diagnosing and managing TBU. These statements are meant to enhance but not replace individual clinician-patient interactions and to facilitate real-world clinical practice improvement in terms of TBU patients care.
Collapse
Affiliation(s)
- Chun-Ju Lin
- Department of Ophthalmology, China Medical University Hospital, China Medical University, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Department of Optometry, Asia University, Taichung, Taiwan
| | - Ning-Yi Hsia
- Department of Ophthalmology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - De-Kuang Hwang
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yih-Shiou Hwang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Yo-Chen Chang
- Department of Ophthalmology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yung-Ray Hsu
- Department of Ophthalmology, Far Eastern Memorial Hospital, Taipei, Taiwan
| | - Po-Ting Yeh
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chang-Ping Lin
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Alan Y Hsu
- Department of Ophthalmology, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of General Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Mao-Wang Ho
- Division of Infectious Diseases, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Shwu-Jiuan Sheu
- Department of Ophthalmology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Ophthalomology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
3
|
Vu A, Glassman I, Campbell G, Yeganyan S, Nguyen J, Shin A, Venketaraman V. Host Cell Death and Modulation of Immune Response against Mycobacterium tuberculosis Infection. Int J Mol Sci 2024; 25:6255. [PMID: 38892443 PMCID: PMC11172987 DOI: 10.3390/ijms25116255] [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: 05/01/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a prevalent infectious disease affecting populations worldwide. A classic trait of TB pathology is the formation of granulomas, which wall off the pathogen, via the innate and adaptive immune systems. Some key players involved include tumor necrosis factor-alpha (TNF-α), foamy macrophages, type I interferons (IFNs), and reactive oxygen species, which may also show overlap with cell death pathways. Additionally, host cell death is a primary method for combating and controlling Mtb within the body, a process which is influenced by both host and bacterial factors. These cell death modalities have distinct molecular mechanisms and pathways. Programmed cell death (PCD), encompassing apoptosis and autophagy, typically confers a protective response against Mtb by containing the bacteria within dead macrophages, facilitating their phagocytosis by uninfected or neighboring cells, whereas necrotic cell death benefits the pathogen, leading to the release of bacteria extracellularly. Apoptosis is triggered via intrinsic and extrinsic caspase-dependent pathways as well as caspase-independent pathways. Necrosis is induced via various pathways, including necroptosis, pyroptosis, and ferroptosis. Given the pivotal role of host cell death pathways in host defense against Mtb, therapeutic agents targeting cell death signaling have been investigated for TB treatment. This review provides an overview of the diverse mechanisms underlying Mtb-induced host cell death, examining their implications for host immunity. Furthermore, it discusses the potential of targeting host cell death pathways as therapeutic and preventive strategies against Mtb infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (G.C.); (A.S.)
| |
Collapse
|
4
|
Nore KG, Louet C, Bugge M, Gidon A, Jørgensen MJ, Jenum S, Dyrhol-Riise AM, Tonby K, Flo TH. The Cyclooxygenase 2 Inhibitor Etoricoxib as Adjunctive Therapy in Tuberculosis Impairs Macrophage Control of Mycobacterial Growth. J Infect Dis 2024; 229:888-897. [PMID: 37721470 PMCID: PMC10938220 DOI: 10.1093/infdis/jiad390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/26/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND Current tuberculosis treatment regimens could be improved by adjunct host-directed therapies (HDT) targeting host responses. We investigated the antimycobacterial capacity of macrophages from patients with tuberculosis in a phase 1/2 randomized clinical trial (TBCOX2) of the cyclooxygenase-2 inhibitor etoricoxib. METHODS Peripheral blood mononuclear cells from 15 patients with tuberculosis treated with adjunctive COX-2i and 18 controls (standard therapy) were collected on day 56 after treatment initiation. The ex vivo capacity of macrophages to control mycobacterial infection was assessed by challenge with Mycobacterium avium, using an in vitro culture model. Macrophage inflammatory responses were analyzed by gene expression signatures, and concentrations of cytokines were analyzed in supernatants by multiplex. RESULTS Macrophages from patients receiving adjunctive COX-2i treatment had higher M. avium loads than controls after 6 days, suggesting an impaired capacity to control mycobacterial infection compared to macrophages from the control group. Macrophages from the COX-2i group had lower gene expression of TNF, IL-1B, CCL4, CXCL9, and CXCL10 and lowered production of cytokines IFN-β and S100A8/A9 than controls. CONCLUSIONS Our data suggest potential unfavorable effects with impaired macrophage capacity to control mycobacterial growth in patients with tuberculosis receiving COX-2i treatment. Larger clinical trials are required to analyze the safety of COX-2i as HDT in patients with tuberculosis. CLINICAL TRIALS REGISTRATION NCT02503839.
Collapse
Affiliation(s)
- Kristin G Nore
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Claire Louet
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Bugge
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Alexandre Gidon
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Trude Helen Flo
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Infection, St Olav's Hospital, Trondheim, Norway
| |
Collapse
|
5
|
Saini S, Gangwar A, Sharma R. Harnessing host-pathogen interactions for innovative drug discovery and host-directed therapeutics to tackle tuberculosis. Microbiol Res 2023; 275:127466. [PMID: 37531813 DOI: 10.1016/j.micres.2023.127466] [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: 06/20/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/04/2023]
Abstract
Tuberculosis (TB) is a highly contagious bacterial infection caused by Mycobacterium tuberculosis (Mtb), which has been ranked as the second leading cause of death worldwide from a single infectious agent. As an intracellular pathogen, Mtb has well adapted to the phagocytic host microenvironment, influencing diverse host processes such as gene expression, trafficking, metabolism, and signaling pathways of the host to its advantage. These responses are the result of dynamic interactions of the bacteria with the host cell signaling pathways, whereby the bacteria attenuate the host cellular processes for their survival. Specific host genes and the mechanisms involved in the entry and subsequent stabilization of M. tuberculosis intracellularly have been identified in various genetic and chemical screens recently. The present understanding of the co-evolution of Mtb and macrophage system presented us the new possibilities for exploring host-directed therapeutics (HDT). Here, we discuss the host-pathogen interaction for Mtb, including the pathways adapted by Mtb to escape immunity. The review sheds light on different host-directed therapies (HDTs) such as repurposed drugs and vitamins, along with their targets such as granuloma, autophagy, extracellular matrix, lipids, and cytokines, among others. The article also examines the available clinical data on these drug molecules. In conclusion, the review presents a perspective on the current knowledge in the field of HDTs and the need for additional research to overcome the challenges associated HDTs.
Collapse
Affiliation(s)
- Sapna Saini
- Infectious Diseases Division, CSIR, Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anjali Gangwar
- Infectious Diseases Division, CSIR, Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rashmi Sharma
- Infectious Diseases Division, CSIR, Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
6
|
Sachan RK, Mistry V, Dholaria M, Rana A, Devgon I, Ali I, Iqbal J, Eldin SM, Mohammad Said Al-Tawaha AR, Bawazeer S, Dutta J, Karnwal A. Overcoming Mycobacterium tuberculosis Drug Resistance: Novel Medications and Repositioning Strategies. ACS OMEGA 2023; 8:32244-32257. [PMID: 37720746 PMCID: PMC10500578 DOI: 10.1021/acsomega.3c02563] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/10/2023] [Indexed: 09/19/2023]
Abstract
Mycobacterium tuberculosis, the bacterium responsible for tuberculosis, is a global health concern, affecting millions worldwide. This bacterium has earned a reputation as a formidable adversary due to its multidrug-resistant nature, allowing it to withstand many antibiotics. The development of this drug resistance in Mycobacterium tuberculosis is attributed to innate and acquired mechanisms. In the past, rifampin was considered a potent medication for treating tuberculosis infections. However, the rapid development of resistance to this drug by the bacterium underscores the pressing need for new therapeutic agents. Fortunately, several other medications previously overlooked for tuberculosis treatment are already available in the market. Moreover, several innovative drugs are under clinical investigation, offering hope for more effective treatments. To enhance the effectiveness of these drugs, it is recommended that researchers concentrate on identifying unique target sites within the bacterium during the drug development process. This strategy could potentially circumvent the issues presented by Mycobacterium drug resistance. This review primarily focuses on the characteristics of novel drug resistance mechanisms in Mycobacterium tuberculosis. It also discusses potential medications being repositioned or sourced from novel origins. The ultimate objective of this review is to discover efficacious treatments for tuberculosis that can successfully tackle the hurdles posed by Mycobacterium drug resistance.
Collapse
Affiliation(s)
| | - Vyoma Mistry
- C.
G. Bhakta Institute of Biotechnology, Uka
Tarsadia University, Bardoli 394350, Surat, India
| | - Mayuri Dholaria
- Naran
Lala College of Professional and Applied Sciences, Navsari 396450, Gujarat, India
| | - Abhishek Rana
- Jindal
Global Law School, OP Jindal Global University, Sonepat 131001, Haryana, India
| | - Inderpal Devgon
- Lovely
Professional University, Phagwara 144411, Punjab, India
| | - Iftikhar Ali
- Center
for Plant Science and Biodiversity, University
of Swat, Charbagh 19120, Pakistan
- Department
of Genetics and Development, Columbia University
Irving Medical Center, New York, New York 10032, United States
| | - Javed Iqbal
- Department
of Botany, Bacha Khan University, Charsadda, 24420 Khyber Pakhtunkhwa, Pakistan
| | - Sayed M. Eldin
- Center
of Research, Faculty of Engineering, Future
University in Egypt, New Cairo 11835, Egypt
| | | | - Sami Bawazeer
- Faculty
of Pharmacy, Department of Pharmacognosy, Umm Al-Qura University, Makkah 4041-4152, Kingdom of Saudi Arabia
| | - Joydeep Dutta
- Lovely
Professional University, Phagwara 144411, Punjab, India
| | - Arun Karnwal
- Lovely
Professional University, Phagwara 144411, Punjab, India
| |
Collapse
|
7
|
Arias L, Otwombe K, Waja Z, Tukvadze N, Korinteli T, Moloantoa T, Fonseca KL, Pillay N, Seiphetlo T, Ouchi-Vernet D, Siles A, Carabias L, Quiñones C, Vashakidze S, Martinson N, Vilaplana C. SMA-TB: study protocol for the phase 2b randomized double-blind, placebo-controlled trial to estimate the potential efficacy and safety of two repurposed drugs, acetylsalicylic acid and ibuprofen, for use as adjunct therapy added to, and compared with, the standard WHO recommended TB regimen. Trials 2023; 24:435. [PMID: 37370174 PMCID: PMC10304643 DOI: 10.1186/s13063-023-07448-0] [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: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The duration and regimen of tuberculosis (TB) treatment is currently based predominantly on whether the M. tuberculosis (Mtb) strain is drug-sensitive (DS) or multidrug-resistant (MDR) with doses adjusted by patients' weight only. The systematic stratification of patients for personalized treatment does not exist for TB. As each TB case is different, individualized treatment regimens should be applied to obtain better outcomes. In this scenario, novel therapeutic approaches are urgently needed to (1) improve outcomes and (2) shorten treatment duration, and host-directed therapies (HDT) might be the best solution. Within HDT, repurposed drugs represent a shortcut in drug development and can be implemented at the short term. As hyperinflammation is associated with worse outcomes, HDT with an anti-inflammatory effect might improve outcomes by reducing tissue damage and thus the risk of permanent sequelae. METHODS SMA-TB is a multicentre randomized, phase IIB, placebo-controlled, three-arm, double-blinded clinical trial (CT) that has been designed in the context of the EC-funded SMA-TB Project ( www.smatb.eu ) in which we propose to use 2 common non-steroidal anti-inflammatory drugs (NSAID), acetylsalicylic acid (ASA) and ibuprofen (Ibu), as an HDT for use as adjunct therapy added to, and compared with, the standard of care (SoC) World Health Organization (WHO)-recommended TB regimen in TB patients. A total of 354 South African and Georgian adults diagnosed with confirmed pulmonary TB will be randomized into SoC TB treatment + placebo, SoC + acetylsalicylic acid or SoC + ibuprofen. DISCUSSION SMA-TB will provide proof of concept of the HDT as a co-adjuvant treatment and identify the suitability of the intervention for different population groups (different epidemiological settings and drug susceptibility) in the reduction of tissue damage and risk of bad outcomes for TB patients. This regimen potentially will be more effective and targeted: organ saving, reducing tissue damage and thereby decreasing the length of treatment and sequelae, increasing cure rates and pathogen clearance and decreasing transmission rates. It will result in better clinical practice, care management and increased well-being of TB patients. TRIAL REGISTRATION Clinicaltrials.gov NCT04575519. Registered on October 5, 2020.
Collapse
Affiliation(s)
- Lilibeth Arias
- Unitat de Tuberculosi Experimental, Germans Trias I Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Kennedy Otwombe
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
| | - Ziyaad Waja
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
| | - Nestani Tukvadze
- National Center for Tuberculosis and Lung Diseases (NCTLD), Tbilisi, Georgia
| | - Tamta Korinteli
- National Center for Tuberculosis and Lung Diseases (NCTLD), Tbilisi, Georgia
| | - Tumelo Moloantoa
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
| | - Kaori L Fonseca
- Unitat de Tuberculosi Experimental, Germans Trias I Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Natasha Pillay
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
| | - Thabiso Seiphetlo
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
| | - Dan Ouchi-Vernet
- Institut d'Investigació en Atenció Primària de Salut Jordi Gol (IDIAPJgol), Barcelona, Spain
| | - Adrian Siles
- Pharmacy department, Germans Trias I Pujol Hospital and Research Institute (HUGTIP-IGTP), Badalona, Catalonia, Spain
| | - Lidia Carabias
- Pharmacy department, Germans Trias I Pujol Hospital and Research Institute (HUGTIP-IGTP), Badalona, Catalonia, Spain
| | - Carles Quiñones
- Pharmacy department, Germans Trias I Pujol Hospital and Research Institute (HUGTIP-IGTP), Badalona, Catalonia, Spain
| | - Sergo Vashakidze
- National Center for Tuberculosis and Lung Diseases (NCTLD), Tbilisi, Georgia
- The University of Georgia, Tbilisi, Georgia
| | - Neil Martinson
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
- Centre for TB Research, Johns Hopkins University, Baltimore, USA
| | - Cristina Vilaplana
- Unitat de Tuberculosi Experimental, Germans Trias I Pujol Research Institute (IGTP), Badalona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.
- Microbiology Department, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias I Pujol, Badalona, Catalonia, Spain.
- Direcció Clínica Territorial de Malalties Infeccioses, Salut Internacional de Gerència Territorial Metropolitana Nord, Badalona, Catalonia, Spain.
| |
Collapse
|
8
|
Putera I, Schrijver B, Ten Berge JCEM, Gupta V, La Distia Nora R, Agrawal R, van Hagen PM, Rombach SM, Dik WA. The immune response in tubercular uveitis and its implications for treatment: From anti-tubercular treatment to host-directed therapies. Prog Retin Eye Res 2023:101189. [PMID: 37236420 DOI: 10.1016/j.preteyeres.2023.101189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 05/28/2023]
Abstract
Tubercular uveitis (TB-uveitis) remains a conundrum in the uveitis field, which is mainly related to the diverse clinical phenotypes of TB-uveitis. Moreover, it remains difficult to differentiate whether Mycobacterium tuberculosis (Mtb) is present in the ocular tissues, elicits a heightened immune response without Mtb invasion in ocular tissues, or even induces an anti-retinal autoimmune response. Gaps in the immuno-pathological knowledge of TB-uveitis likely delay timely diagnosis and appropriate management. In the last decade, the immunopathophysiology of TB-uveitis and its clinical management, including experts' consensus to treat or not to treat certain conditions with anti-tubercular treatment (ATT), have been extensively investigated. In the meantime, research on TB treatment, in general, is shifting more toward host-directed therapies (HDT). Given the complexities of the host-Mtb interaction, enhancement of the host immune response is expected to boost the effectiveness of ATT and help overcome the rising burden of drug-resistant Mtb strains in the population. This review will summarize the current knowledge on the immunopathophysiology of TB-uveitis and recent advances in treatment modalities and outcomes of TB-uveitis, capturing results gathered from high- and low-burden TB countries with ATT as the mainstay of treatment. Moreover, we outline the recent progress of HDT development in the pulmonary TB field and discuss the possibility of its applicability to TB-uveitis. The concept of HDT might help direct future development of efficacious therapy for TB-uveitis, although more in-depth research on the immunoregulation of this disease is still necessary.
Collapse
Affiliation(s)
- Ikhwanuliman Putera
- Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Internal Medicine, Section Allergy and Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands; Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia.
| | - Benjamin Schrijver
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Vishali Gupta
- Retina and Uvea Services, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Rina La Distia Nora
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Rupesh Agrawal
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke NUS University, Singapore; Singapore Eye Research Institute, Singapore; Moorfields Eye Hospital, London, United Kingdom
| | - P Martin van Hagen
- Department of Internal Medicine, Section Allergy and Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands; Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - S M Rombach
- Department of Internal Medicine, Section Allergy and Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Willem A Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
9
|
Zhang MN, Yuan YL, Ao SH. Advances in the study of myeloid-derived suppressor cells in infectious lung diseases. Front Immunol 2023; 14:1125737. [PMID: 37063919 PMCID: PMC10090681 DOI: 10.3389/fimmu.2023.1125737] [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: 12/16/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature cells capable of inhibiting T-cell responses. MDSCs have a crucial role in the regulation of the immune response of the body to pathogens, especially in inflammatory response and pathogenesis during anti-infection. Pathogens such as bacteria and viruses use MDSCs as their infectious targets, and even some pathogens may exploit the inhibitory activity of MDSCs to enhance pathogen persistence and chronic infection of the host. Recent researches have revealed the pathogenic significance of MDSCs in pathogens such as bacteria and viruses, despite the fact that the majority of studies on MDSCs have focused on tumor immune evasion. With the increased prevalence of viral respiratory infections, the resurgence of classical tuberculosis, and the advent of medication resistance in common bacterial pneumonia, research on MDSCs in these illnesses is intensifying. The purpose of this work is to provide new avenues for treatment approaches to pulmonary infectious disorders by outlining the mechanism of action of MDSCs as a biomarker and therapeutic target in pulmonary infectious diseases.
Collapse
Affiliation(s)
- Meng-Nan Zhang
- College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu-Lai Yuan
- The Department of Respirology of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Su-Hua Ao
- The Department of Respirology of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- *Correspondence: Su-Hua Ao,
| |
Collapse
|
10
|
Qahtan MQM, Bakhite EA, Kumari J, M Sayed A, Kandeel M, Sriram D, Abdu-Allah HHM. Synthesis, biological evaluation and molecular docking study of some new 4-aminosalicylic acid derivatives as anti-inflammatory and antimycobacterial agents. Bioorg Chem 2023; 132:106344. [PMID: 36669356 DOI: 10.1016/j.bioorg.2023.106344] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/27/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023]
Abstract
In this study, new derivatives of the antitubercular and anti-inflammatory drug, 4-aminosaliclic acids (4-ASA) were synthesized, characterized, and evaluated for these activities. In vivo and in viro evaluation of anti-inflammatory activity revealed that compounds 10, 19 and 20 are the most active with potent cyclooxygenase-2 (COX-2) and 5-lipooxgenase (5-LOX) inhibition and without causing gasric lesions. The minimum inhibitory concentrations (MIC) of the newly synthesized compound were, also, measured against Mycobacterium tuberculosis H37RV. Among the tested compounds 17, 19 and 20 exhibited significant activities against the growth of M. tuberculosis. 20 is the most potent with (MIC 1.04 µM) 2.5 folds more potent than the parent drug 4-ASA. 20 displayed low cytotoxicity against normal cell providing a high therapeutic index. Important structure features were analyzed by docking and structure-activity relationship analysis to give better insights into the structural determinants for predicting the anti-inflammatory and anti-TB activities. Our results indicated that compounds 19 and 20 are potential lead compounds for the discovery of dual anti-inflammatory and anti-TB drug candidates.
Collapse
Affiliation(s)
- Maha Q M Qahtan
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt; Chemistry Department, Faculty of Science, Taiz University, Taiz, Yemen
| | - Etify A Bakhite
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Jyothi Kumari
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad 500 078, India
| | - Ahmed M Sayed
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Dharmarajan Sriram
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad 500 078, India
| | - Hajjaj H M Abdu-Allah
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| |
Collapse
|
11
|
Dobrzeniecka W, Daca M, Nowakowska B, Sobiesiak M, Szewczyk-Golec K, Woźniak A, Hołyńska-Iwan I. The Impact of Diclofenac Gel on Ion Transport in the Rabbit ( Oryctolagus cuniculus) Skin: An In Vitro Study. Molecules 2023; 28:molecules28031332. [PMID: 36770998 PMCID: PMC9920221 DOI: 10.3390/molecules28031332] [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: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Diclofenac belongs to the non-steroidal anti-inflammatory drugs with analgesic, antipyretic and anti-inflammatory effects. Diclofenac administration on the skin may be associated with the appearance of side effects. The study aimed to evaluate the impact of diclofenac gel on transepithelial electrophysiological parameters of the 55 rabbit abdomen skin specimens. The electric parameters were analyzed in a modified Ussing chamber. The resistance (R) of the skin specimens treated with diclofenac gel significantly increased, which could be related to the reduction in the water content in intercellular spaces and, consequently, tighter adhesion of the cells. Increased electric potential (PD) was also observed in the skin specimens treated with diclofenac gel. The increase in both R and PD measured under stationary conditions was most likely caused by a transient and reversible increase in sodium ion transport, as the R and PD values decreased after the diclofenac gel was washed away. However, diclofenac gel did not affect the maximum and minimum PDs measured during stimulations. Therefore, it seems that diclofenac gel does not affect the perception of stimuli in the model system used.
Collapse
Affiliation(s)
- Wioletta Dobrzeniecka
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Małgorzata Daca
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Barbara Nowakowska
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Marta Sobiesiak
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Karolina Szewczyk-Golec
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
- Correspondence: (A.W.); (I.H.-I.)
| | - Iga Hołyńska-Iwan
- Department of Pathobiochemistry and Clinical Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
- Correspondence: (A.W.); (I.H.-I.)
| |
Collapse
|
12
|
Restrepo BI, Scordo JM, Aguillón-Durán GP, Ayala D, Quirino-Cerrillo AP, Loera-Salazar R, Cruz-González A, Caso JA, Joya-Ayala M, García-Oropesa EM, Salinas AB, Martinez L, Schlesinger LS, Torrelles JB, Turner J. Differential Role of Type 2 Diabetes as a Risk Factor for Tuberculosis in the Elderly versus Younger Adults. Pathogens 2022; 11:1551. [PMID: 36558885 PMCID: PMC9784720 DOI: 10.3390/pathogens11121551] [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: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
The elderly are understudied despite their high risk of tuberculosis (TB). We sought to identify factors underlying the lack of an association between TB and type 2 diabetes (T2D) in the elderly, but not adults. We conducted a case-control study in elderly (≥65 years old; ELD) vs. younger adults (young/middle-aged adults (18-44/45-64 years old; YA|MAA) stratified by TB and T2D, using a research study population (n = 1160) and TB surveillance data (n = 8783). In the research study population the adjusted odds ratio (AOR) of TB in T2D was highest in young adults (AOR 6.48) but waned with age becoming non-significant in the elderly. Findings were validated using TB surveillance data. T2D in the elderly (vs. T2D in younger individuals) was characterized by better glucose control (e.g., lower hyperglycemia or HbA1c), lower insulin resistance, more sulphonylureas use, and features of less inflammation (e.g., lower obesity, neutrophils, platelets, anti-inflammatory use). We posit that differences underlying glucose dysregulation and inflammation in elderly vs. younger adults with T2D, contribute to their differential association with TB. Studies in the elderly provide valuable insights into TB-T2D pathogenesis, e.g., here we identified insulin resistance as a novel candidate mechanism by which T2D may increase active TB risk.
Collapse
Affiliation(s)
- Blanca I. Restrepo
- School of Public Health and UTHealth Consortium on Aging, University of Texas Health Science Center at Houston, Brownsville Campus, Brownsville, TX 78520, USA
- School of Medicine, South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA
- Host Pathogen Interactions and Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Julia M. Scordo
- Host Pathogen Interactions and Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Barshop Institute, The University of Texas Health Science Center of San Antonio, San Antonio, TX 78229, USA
| | | | - Doris Ayala
- School of Public Health and UTHealth Consortium on Aging, University of Texas Health Science Center at Houston, Brownsville Campus, Brownsville, TX 78520, USA
| | - Ana Paulina Quirino-Cerrillo
- School of Public Health and UTHealth Consortium on Aging, University of Texas Health Science Center at Houston, Brownsville Campus, Brownsville, TX 78520, USA
| | - Raúl Loera-Salazar
- Secretaría de Salud de Tamaulipas, Reynosa 88630, Matamoros 87370 and Ciudad Victoria 87000, Mexico
| | - America Cruz-González
- Secretaría de Salud de Tamaulipas, Reynosa 88630, Matamoros 87370 and Ciudad Victoria 87000, Mexico
| | - Jose A. Caso
- Biology Department, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA
| | - Mateo Joya-Ayala
- School of Public Health and UTHealth Consortium on Aging, University of Texas Health Science Center at Houston, Brownsville Campus, Brownsville, TX 78520, USA
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA
| | - Esperanza M. García-Oropesa
- Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas Reynosa-Aztlán, Reynosa 88779, Mexico
| | - Alejandra B. Salinas
- School of Public Health and UTHealth Consortium on Aging, University of Texas Health Science Center at Houston, Brownsville Campus, Brownsville, TX 78520, USA
| | - Leonardo Martinez
- Department of Epidemiology, School of Public Health, Boston University, Boston, MA 02118, USA
| | - Larry S. Schlesinger
- Host Pathogen Interactions and Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Jordi B. Torrelles
- Host Pathogen Interactions and Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Joanne Turner
- Host Pathogen Interactions and Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| |
Collapse
|
13
|
Park HE, Lee W, Choi S, Jung M, Shin MK, Shin SJ. Modulating macrophage function to reinforce host innate resistance against Mycobacterium avium complex infection. Front Immunol 2022; 13:931876. [PMID: 36505429 PMCID: PMC9730288 DOI: 10.3389/fimmu.2022.931876] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 10/21/2022] [Indexed: 11/25/2022] Open
Abstract
Mycobacterium avium complex (MAC) is the main causative agent of infectious diseases in humans among nontuberculous mycobacteria (NTM) that are ubiquitous organisms found in environmental media such as soil as well as in domestic and natural waters. MAC is a primary causative agent of NTM-lung disease that threaten immunocompromised or structural lung disease patients. The incidence and the prevalence of M. tuberculosis infection have been reduced, while MAC infections and mortality rates have increased, making it a cause of global health concern. The emergence of drug resistance and the side effects of long-term drug use have led to a poor outcome of treatment regimens against MAC infections. Therefore, the development of host-directed therapy (HDT) has recently gained interest, aiming to accelerate mycobacterial clearance and reversing lung damage by employing the immune system using a novel adjuvant strategy to improve the clinical outcome of MAC infection. Therefore, in this review, we discuss the innate immune responses that contribute to MAC infection focusing on macrophages, chief innate immune cells, and host susceptibility factors in patients. We also discuss potential HDTs that can act on the signaling pathway of macrophages, thereby contributing to antimycobacterial activity as a part of the innate immune response during MAC infection. Furthermore, this review provides new insights into MAC infection control that modulates and enhances macrophage function, promoting host antimicrobial activity in response to potential HDTs and thus presenting a deeper understanding of the interactions between macrophages and MACs during infection.
Collapse
Affiliation(s)
- Hyun-Eui Park
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Sangwon Choi
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Myunghwan Jung
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea,*Correspondence: Min-Kyoung Shin, ; Sung Jae Shin,
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea,*Correspondence: Min-Kyoung Shin, ; Sung Jae Shin,
| |
Collapse
|
14
|
Korotetskaya MV, Rubakova EI. Metabolic biological markers for diagnosing and monitoring the course of tuberculosis. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2022. [DOI: 10.15789/2220-7619-mbm-1947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The international biomedical community has been currently facing a need to find a simple and most accessible type of analysis that helps to diagnose tuberculosis (TB) with the maximum reliability even before the onset of clinical manifestations. Tuberculosis results in more deaths than any other pathogen, second only to pneumonia caused by the SARS-CoV-2 virus, but the majority of infected people remain asymptomatic. In addition, it is important to develop methods to distinguish various forms of tuberculosis infection course at early stages and to reliably stratify patients into appropriate groups (persons with a rapidly progressing infection, chronic course, latent infection carriers). Immunometabolism investigates a relationship between bioenergetic pathways and specific functions of immune cells that has recently become increasingly important in scientific research. The host anti-mycobacteria immune response in tuberculosis is regulated by a number of metabolic networks that can interact both cooperatively and antagonistically, influencing an outcome of the disease. The balance between inflammatory and immune reactions limits the spread of mycobacteria in vivo and protects from developing tuberculosis. Cytokines are essential for host defense, but if uncontrolled, some mediators may contribute to developing disease and pathology. Differences in plasma levels of metabolites between individuals with advanced infection, LTBI and healthy individuals can be detected long before the onset of the major related clinical signs. Changes in amino acid and cortisol level may be detected as early as 12 months before the onset of the disease and become more prominent at verifying clinical diagnosis. Assessing serum level of certain amino acids and their ratios may be used as additional diagnostic markers of active pulmonary TB. Metabolites, including serum fatty acids, amino acids and lipids may contribute to detecting active TB. Metabolic profiles indicate about increased indolamine 2.3-dioxygenase 1 (IDO1) activity, decreased phospholipase activity, increased adenosine metabolite level, and fibrous lesions in active vs. latent infection. TB treatment can be adjusted based on individual patient metabolism and biomarker profiles. Thus, exploring immunometabolism in tuberculosis is necessary for development of new therapeutic strategies.
Collapse
|
15
|
Reddy DS, Sinha A, Kumar A, Saini VK. Drug re-engineering and repurposing: A significant and rapid approach to tuberculosis drug discovery. Arch Pharm (Weinheim) 2022; 355:e2200214. [PMID: 35841594 DOI: 10.1002/ardp.202200214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 01/11/2023]
Abstract
The prevalence of tuberculosis (TB) remains the leading cause of death from a single infectious agent, ranking it above all other contagious diseases. The problem to tackle this disease seems to become even worse due to the outbreak of SARS-CoV-2. Further, the complications related to drug-resistant TB, prolonged treatment regimens, and synergy between TB and HIV are significant drawbacks. There are several drugs to treat TB, but there is still no rapid and accurate treatment available. Intensive research is, therefore, necessary to discover newer molecular analogs that can probably eliminate this disease within a short span. An increase in efficacy can be achieved through re-engineering old TB-drug families and repurposing known drugs. These two approaches have led to the production of newer classes of compounds with novel mechanisms to treat multidrug-resistant strains. With respect to this context, we discuss structural aspects of developing new anti-TB drugs as well as examine advances in TB drug discovery. It was found that the fluoroquinolone, oxazolidinone, and nitroimidazole classes of compounds have greater potential to be further explored for TB drug development. Most of the TB drug candidates in the clinical phase are modified versions of these classes of compounds. Therefore, here we anticipate that modification or repurposing of these classes of compounds has a higher probability to reach the clinical phase of drug development. The information provided will pave the way for researchers to design and identify newer molecular analogs for TB drug development and also broaden the scope of exploring future-generation potent, yet safer anti-TB drugs.
Collapse
Affiliation(s)
- Dinesh S Reddy
- Centre for Nano and Material Sciences, Jain University, Bangalore, India
| | - Anamika Sinha
- Centre for Nano and Material Sciences, Jain University, Bangalore, India
| | - Amit Kumar
- Centre for Nano and Material Sciences, Jain University, Bangalore, India
| | - Vipin K Saini
- Materials and Environmental Chemistry Research Laboratory, School of Environment & Natural Resources, Doon University, Dehradun, India
| |
Collapse
|
16
|
Clinical Manifestations and Outcomes of Tubercular Uveitis in Taiwan—A Ten-Year Multicenter Retrospective Study. Medicina (B Aires) 2022; 58:medicina58030376. [PMID: 35334552 PMCID: PMC8950060 DOI: 10.3390/medicina58030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background andObjectives: This 10-year multicenter retrospective study reviewed the clinical manifestations, diagnostic tests, and treatment modalities of tubercular uveitis (TBU), including direct infection and indirect immune-mediated hypersensitivity to mycobacterial antigens in Taiwan. Materials and Methods: This retrospective chart review of patients with TBU was conducted at 11 centers from 1 January 2008 to 31 December 2017. We used a multiple regression model to analyze which factors influenced best-corrected visual acuity (BCVA) improvement. Results: A total of 79 eyes from 51 patients were included in the study. The mean age was 48.9 ± 16.4 years. The mean change of LogMAR BCVA at last visit was −0.21 ± 0.45. Diagnostic tools used include chest X-ray, chest computed tomography, Mantoux test, interferon gamma release test (QuantiFERON-TB Gold test), intraocular fluid tuberculosis polymerase chain reaction, and bronchial alveolar lavage. The clinical manifestations included 48% posterior uveitis and 37% panuveitis. In the sample, 55% of the cases were bilateral and 45% unilateral. There was 60.76% retinal vasculitis, 35.44% choroiditis, 21.52% serpiginous-like choroiditis, 17.72% vitreous hemorrhage, 12.66% posterior synechiae, 6.33% retinal detachment, and 3.80% choroidal granuloma. Treatment modalities included rifampicin, isoniazid, pyrazinamide, ethambutol, oral steroid, posterior triamcinolone, non-steroidal anti-inflammatory drugs, vitrectomy, and immunosuppressants. BCVA improved in 53.2% of eyes and remained stable in 32.9% of eyes. In the final model of multiple regression, worse initial BCVA, pyrazinamide, and receiving vitrectomy predicted better BCVA improvement. Ethambutol was associated with worse visual outcomes. Seven eyes experienced recurrence. Conclusions: This is the largest 10-year multicenter retrospective study of TBU in Taiwan to date, demonstrating the distribution of clinical manifestations and clinical associations with better treatment outcomes. The study provides a comprehensive description of TBU phenotypes in Taiwan and highlights considerations for the design of further prospective studies to reliably assess the role of ATT and vitrectomy in patients with TBU.
Collapse
|
17
|
Abdel Shaheed C, Beardsley J, Day RO, McLachlan AJ. Immunomodulatory effects of pharmaceutical opioids and antipyretic analgesics: Mechanisms and relevance to infection. Br J Clin Pharmacol 2022; 88:3114-3131. [PMID: 35229890 DOI: 10.1111/bcp.15281] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/12/2022] Open
Abstract
Understanding how pharmaceutical opioids and antipyretic analgesics interact with the immune system potentially has major clinical implications for management of patients with infectious diseases and surgical and critical care patients. An electronic search was carried out on MEDLINE, EMBASE, PsycINFO, CENTRAL and the Cochrane library to identify reports describing the immunomodulatory effects of opioid analgesics and antipyretic analgesics, and their effects in infectious diseases. In adaptive immunity, nonsteroidal anti-inflammatory drugs have divergent effects: augmenting cell-mediated immunity but inhibiting humoral immunity. Nonsteroidal anti-inflammatory drugs have demonstrated a beneficial role in Mycobacterium tuberculosis infection and histoplasmosis in animals, and may be plausible adjuvants to antimicrobial agents in these diseases. There is a need to evaluate these findings rigorously in human clinical trials. There is preliminary evidence demonstrating antiviral effects of indomethacin in SARS CoV-2 in vitro; however, uncertainty regarding its clinical benefit in humans needs to be resolved in large clinical trials. Certain opioid analgesics are associated with immunosuppressive effects, with a developing understanding that fentanyl, morphine, methadone and buprenorphine suppress innate immunity, whilst having diverse effects on adaptive immunity. Morphine suppresses key cells of the innate immunity and is associated with greater risk of infection in the postsurgical setting. Efforts are needed to achieve adequate analgesia whilst avoiding suppression of the innate immunity in the immediate postoperative period caused by certain opioids, particularly in cancer surgery.
Collapse
Affiliation(s)
- Christina Abdel Shaheed
- Faculty of Medicine and Health, Sydney School of Public Health, University of Sydney, Sydney, Australia.,Institute for Musculoskeletal Health, University of Sydney and Sydney Local Health District, Sydney, Australia
| | - Justin Beardsley
- Westmead Institute for Medical Research, Sydney, Australia.,Sydney Institute for Infectious Diseases, University of Sydney, Australia
| | - Richard O Day
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Andrew J McLachlan
- Faculty of Medicine and Health, Sydney Pharmacy School, University of Sydney, Sydney, Australia
| |
Collapse
|
18
|
Mirocki A, Sikorski A. Structural Characterization of Multicomponent Crystals Formed from Diclofenac and Acridines. MATERIALS 2022; 15:ma15041518. [PMID: 35208056 PMCID: PMC8876612 DOI: 10.3390/ma15041518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 12/28/2022]
Abstract
Multicomponent crystals containing diclofenac and acridine (1) and diclofenac and 6,9-diamino-2-ethoxyacridine (2) were synthesized and structurally characterized. The single-crystal XRD measurements showed that compound 1 crystallizes in the triclinic P-1 space group as a salt cocrystal with one acridinium cation, one diclofenac anion, and one diclofenac molecule in the asymmetric unit, whereas compound 2 crystallizes in the triclinic P-1 space group as an ethanol solvate monohydrate salt with one 6,9-diamino-2-ethoxyacridinium cation, one diclofenac anion, one ethanol molecule, and one water molecule in the asymmetric unit. In the crystals of the title compounds, diclofenac and acridines ions and solvent molecules interact via N–H⋯O, O–H⋯O, and C–H⋯O hydrogen bonds, as well as C–H⋯π and π–π interactions, and form heterotetramer bis[⋯cation⋯anion⋯] (1) or heterohexamer bis[⋯cation⋯ethanol⋯anion⋯] (2). Moreover, in the crystal of compound 1, acridine cations and diclofenac anions interact via N–H⋯O hydrogen bond, C–H⋯π and π–π interactions to produce blocks, while diclofenac molecules interact via C–Cl⋯π interactions to form columns. In the crystal of compound 2, the ethacridine cations interact via C–H⋯π and π–π interactions building blocks, while diclofenac anions interact via π–π interactions to form columns.
Collapse
Affiliation(s)
- Artur Mirocki
- Correspondence: (A.M.); (A.S.); Tel.: +48-58-523-5112 (A.M. & A.S.)
| | - Artur Sikorski
- Correspondence: (A.M.); (A.S.); Tel.: +48-58-523-5112 (A.M. & A.S.)
| |
Collapse
|
19
|
Nienaber A, Ozturk M, Dolman R, Blaauw R, Zandberg LL, van Rensburg S, Britz M, Hayford FEA, Brombacher F, Loots DT, Smuts CM, Parihar SP, Malan L. n-3 long-chain PUFA promote antibacterial and inflammation-resolving effects in Mycobacterium tuberculosis-infected C3HeB/FeJ mice, dependent on fatty acid status. Br J Nutr 2022; 127:384-397. [PMID: 33814018 DOI: 10.1017/s0007114521001124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Non-resolving inflammation is characteristic of tuberculosis (TB). Given their inflammation-resolving properties, n-3 long-chain PUFA (n-3 LCPUFA) may support TB treatment. This research aimed to investigate the effects of n-3 LCPUFA on clinical and inflammatory outcomes of Mycobacterium tuberculosis-infected C3HeB/FeJ mice with either normal or low n-3 PUFA status before infection. Using a two-by-two design, uninfected mice were conditioned on either an n-3 PUFA-sufficient (n-3FAS) or -deficient (n-3FAD) diet for 6 weeks. One week post-infection, mice were randomised to either n-3 LCPUFA supplemented (n-3FAS/n-3+ and n-3FAD/n-3+) or continued on n-3FAS or n-3FAD diets for 3 weeks. Mice were euthanised and fatty acid status, lung bacterial load and pathology, cytokine, lipid mediator and immune cell phenotype analysed. n-3 LCPUFA supplementation in n-3FAS mice lowered lung bacterial loads (P = 0·003), T cells (P = 0·019), CD4+ T cells (P = 0·014) and interferon (IFN)-γ (P < 0·001) and promoted a pro-resolving lung lipid mediator profile. Compared with n-3FAS mice, the n-3FAD group had lower bacterial loads (P = 0·037), significantly higher immune cell recruitment and a more pro-inflammatory lipid mediator profile, however, significantly lower lung IFN-γ, IL-1α, IL-1β and IL-17, and supplementation in the n-3FAD group provided no beneficial effect on lung bacterial load or inflammation. Our study provides the first evidence that n-3 LCPUFA supplementation has antibacterial and inflammation-resolving benefits in TB when provided 1 week after infection in the context of a sufficient n-3 PUFA status, whilst a low n-3 PUFA status may promote better bacterial control and lower lung inflammation not benefiting from n-3 LCPUFA supplementation.
Collapse
Affiliation(s)
- Arista Nienaber
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Mumin Ozturk
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town, Western Cape, South Africa
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town, Western Cape, South Africa
| | - Robin Dolman
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Renee Blaauw
- Division of Human Nutrition, Stellenbosch University, Tygerberg, Cape Town, Western Cape, South Africa
| | - Lizelle L Zandberg
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Simone van Rensburg
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Melinda Britz
- 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, University of Ghana, Accra, Ghana
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town, Western Cape, South Africa
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town, Western Cape, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, Western Cape, South Africa
| | - Du Toit Loots
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Cornelius M Smuts
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Suraj P Parihar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town, Western Cape, South Africa
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town, Western Cape, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, Western Cape, South Africa
- Division of Medical Microbiology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, Western Cape, South Africa
| | - Linda Malan
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| |
Collapse
|
20
|
Perveen S, Kumari D, Singh K, Sharma R. Tuberculosis drug discovery: Progression and future interventions in the wake of emerging resistance. Eur J Med Chem 2022; 229:114066. [PMID: 34973508 DOI: 10.1016/j.ejmech.2021.114066] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/13/2021] [Accepted: 12/19/2021] [Indexed: 02/06/2023]
Abstract
The emergence of drug resistance continues to afflict TB control where drug resistant strains have become a global health concern. Contrary to drug-sensitive TB, the treatment of MDR/XDR-TB is more complicated requiring the administration of second-line drugs that are inefficient than the first line drugs and are associated with greater side effects. The emergence of drug resistant Mtb strains had coincided with an innovation void in the field of drug discovery of anti-mycobacterials. However, the approval of bedaquiline and delamanid recently for use in MDR/XDR-TB has given an impetus to the TB drug discovery. The review discusses the drug discovery efforts in the field of tuberculosis with a focus on the strategies adopted and challenges confronted by TB research community. Here, we discuss the diverse clinical candidates in the current TB drug discovery pipeline. There is an urgent need to combat the current TB menace through multidisciplinary approaches and strategies making use of the recent advances in understanding the molecular biology and pathogenesis of Mtb. The review highlights the recent advances in drug discovery, with the host directed therapeutics and nanoparticles-drug delivery coming up as important tools to fight tuberculosis in the future.
Collapse
Affiliation(s)
- Summaya Perveen
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Diksha Kumari
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rashmi Sharma
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
21
|
Wu X, Jin S, Ding C, Wang Y, He D, Liu Y. Mesenchymal Stem Cell-Derived Exosome Therapy of Microbial Diseases: From Bench to Bed. Front Microbiol 2022; 12:804813. [PMID: 35046923 PMCID: PMC8761948 DOI: 10.3389/fmicb.2021.804813] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Microbial diseases are a global health threat, leading to tremendous casualties and economic losses. The strategy to treat microbial diseases falls into two broad categories: pathogen-directed therapy (PDT) and host-directed therapy (HDT). As the typical PDT, antibiotics or antiviral drugs directly attack bacteria or viruses through discerning specific molecules. However, drug abuse could result in antimicrobial resistance and increase infectious disease morbidity. Recently, the exosome therapy, as a HDT, has attracted extensive attentions for its potential in limiting infectious complications and targeted drug delivery. Mesenchymal stem cell-derived exosomes (MSC-Exos) are the most broadly investigated. In this review, we mainly focus on the development and recent advances of the application of MSC-Exos on microbial diseases. The review starts with the difficulties and current strategies in antimicrobial treatments, followed by a comprehensive overview of exosomes in aspect of isolation, identification, contents, and applications. Then, the underlying mechanisms of the MSC-Exo therapy in microbial diseases are discussed in depth, mainly including immunomodulation, repression of excessive inflammation, and promotion of tissue regeneration. In addition, we highlight the latest progress in the clinical translation of the MSC-Exo therapy, by summarizing related clinical trials, routes of administration, and exosome modifications. This review will provide fundamental insights and future perspectives on MSC-Exo therapy in microbial diseases from bench to bedside.
Collapse
Affiliation(s)
| | | | | | | | | | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology and National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Digital and Material Technology of Stomatology and Beijing Key Laboratory of Digital Stomatology and Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health and NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|
22
|
Biswas P, Datta HK, Dastidar P. Designing Coordination Polymers as Multi-drug-self-delivery System for Tuberculosis and Cancer Therapy: in vitro Viability and in vivo Toxicity Assessment. Biomater Sci 2022; 10:6201-6216. [PMID: 36097681 DOI: 10.1039/d2bm00752e] [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: 11/21/2022]
Abstract
A proof of the concept for designing multi-drug-delivery system suitable for self-drug-delivery is disclosed. Simple coordination chemistry was employed to anchor two kinds of drugs namely isoniazid (IZ – anti-tuberculosis),...
Collapse
Affiliation(s)
- Protap Biswas
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A and 2B Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India.
| | - Hemanta Kumar Datta
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A and 2B Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India.
| | - Parthasarathi Dastidar
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A and 2B Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India.
| |
Collapse
|
23
|
Paradoxical manifestations during tuberculous meningitis treatment among HIV-negative patients: a retrospective descriptive study and literature review. Neurol Sci 2021; 43:2699-2708. [PMID: 34705128 DOI: 10.1007/s10072-021-05693-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Tuberculous meningitis (TBM) is the most frequent, severe, and disabling form of central nervous system (CNS) tuberculosis (TB). TBM paradoxical manifestations are characterized by clinical or paraclinical worsening after 1 month of effective anti-TB treatment in patients who initially responded to treatment despite the use of adjunctive corticosteroids. METHODS Retrospective descriptive study of consecutive HIV-negative adult patients (≥ 18 years) with definitive TBM who developed a paradoxical manifestation following anti-TB in a tertiary-care hospital in Mexico from 2009 to 2019; we also conducted a literature review of published cases/series of paradoxical manifestations in HIV-negative patients from 1980 to 2020. RESULTS We detected 84 cases of definitive TBM; 55 (68.7%) HIV-negative patients and 29 (36.3%) HIV-infected patients. Among HIV-negative patients, four (7.3%), three female and one male (19-49 years old), developed a paradoxical manifestation within 4-14 weeks following treatment initiation despite receiving adequate corticosteroid doses; Mycobacterium bovis was isolated from the cerebrospinal fluid of three cases and Mycobacterium tuberculosis in one more. Two patients developed vasculopathy-related cerebral infarctions, one severe basilar meningitis, and hydrocephalus, one more a tuberculoma. Two were treated with intravenous cyclophosphamide, and two with steroids. One of the patients treated with steroids died; patients who received cyclophosphamide had a good clinical response. CONCLUSIONS This case series illustrates the diverse clinical/radiologic paradoxical manifestations of TBM in HIV-negative patients. Cyclophosphamide may be safe and effective in treating TBM-associated paradoxical manifestations. Specific diagnostic and care protocols for these patients are needed.
Collapse
|
24
|
Bhatt K, Bhagavathula M, Verma S, Timmins GS, Deretic VP, Ellner JJ, Salgame P. Rapamycin modulates pulmonary pathology in a murine model of Mycobacterium tuberculosis infection. Dis Model Mech 2021; 14:272048. [PMID: 34486033 PMCID: PMC8560501 DOI: 10.1242/dmm.049018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB) treatment regimens are lengthy, causing non-adherence to treatment. Inadequate treatment can lead to relapse and the development of drug resistance TB. Furthermore, patients often exhibit residual lung damage even after cure, increasing the risk for relapse and development of other chronic respiratory illnesses. Host-directed therapeutics are emerging as an attractive means to augment the success of TB treatment. In this study, we used C3HeB/FeJ mice as an experimental model to investigate the potential role of rapamycin, a mammalian target of rapamycin inhibitor, as an adjunctive therapy candidate during the treatment of Mycobacterium tuberculosis infection with moxifloxacin. We report that administration of rapamycin with or without moxifloxacin reduced infection-induced lung inflammation, and the number and size of caseating necrotic granulomas. Results from this study strengthen the potential use of rapamycin and its analogs as adjunct TB therapy, and importantly underscore the utility of the C3HeB/FeJ mouse model as a preclinical tool for evaluating host-directed therapy candidates for the treatment of TB. Summary: Rapamycin, an mTOR inhibitor, with or without moxifloxacin, reduces lung inflammation and the number and size of caseating necrotic granulomas in Mycobacterium tuberculosis-infected C3HeB/FeJ mice.
Collapse
Affiliation(s)
- Kamlesh Bhatt
- Center for Emerging Pathogens, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Madhuri Bhagavathula
- Center for Emerging Pathogens, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Sheetal Verma
- Center for Emerging Pathogens, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Graham S Timmins
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Vojo P Deretic
- Autophagy Inflammation and Metabolism (AIM) Center of Biomedical Research Excellence University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.,Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Jerrold J Ellner
- Center for Emerging Pathogens, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Padmini Salgame
- Center for Emerging Pathogens, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
25
|
Review of the Effectiveness of Various Adjuvant Therapies in Treating Mycobacterium tuberculosis. Infect Dis Rep 2021; 13:821-834. [PMID: 34562999 PMCID: PMC8482146 DOI: 10.3390/idr13030074] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 12/18/2022] Open
Abstract
Tuberculosis disease is caused by the bacterium Mycobacterium tuberculosis. It is estimated that 10 million people have developed tuberculosis disease globally, leading to 1.4 million deaths in 2019. Treatment of tuberculosis has been especially challenging due to the rise of multidrug-resistant (MDR-TB) and extensive drug-resistant (XDR-TB) tuberculosis. In addition to drug-resistant genotypes, the standard treatment of tuberculosis by first-line agents is also challenging due to toxicity and costs. In the last four decades, there have only been two new anti-tuberculosis agents—bedaquiline and delamanid. Therefore, shorter, safer, and more cost-effective therapies are needed to adequately treat tuberculosis. In this review, we explore various adjuvants such as glutathione, everolimus, vitamin D, steroid, aspirin, statin, and metformin and their usefulness in reducing the burden of tuberculosis. Glutathione, everolimus, aspirin, and metformin showed the most promise in alleviating the burden of tuberculosis. Despite their potential, more clinical trials are needed to unequivocally establish the effectiveness of these adjuvants as future clinical therapies. Methods: The journals for this review were selected by conducting a search via PubMed, Google Scholar, and The Lancet. Our first search included keywords such as “tuberculosis” and “adjuvant therapy.” From the search, we made a list of adjuvants associated with tuberculosis, and this helped guide us with our second online database search. Using the same three online databases, we searched “tuberculosis” and “respective therapy.” The adjuvants included in the paper were selected based on the availability of sufficient research and support between the therapy and tuberculosis. Adjuvants with minimal research support were excluded. There were no specific search criteria regarding the timing of publication, with our citations ranging between 1979 to 2021.
Collapse
|
26
|
Jøntvedt Jørgensen M, Nore KG, Aass HCD, Layre E, Nigou J, Mortensen R, Tasken K, Kvale D, Jenum S, Tonby K, Dyrhol-Riise AM. Plasma LOX-Products and Monocyte Signaling Is Reduced by Adjunctive Cyclooxygenase-2 Inhibitor in a Phase I Clinical Trial of Tuberculosis Patients. Front Cell Infect Microbiol 2021; 11:669623. [PMID: 34307194 PMCID: PMC8299478 DOI: 10.3389/fcimb.2021.669623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/21/2021] [Indexed: 11/22/2022] Open
Abstract
Introduction Eicosanoids and intracellular signaling pathways are potential targets for host-directed therapy (HDT) in tuberculosis (TB). We have explored the effect of cyclooxygenase 2 inhibitor (COX-2i) treatment on eicosanoid levels and signaling pathways in monocytes. Methods Peripheral blood mononuclear cells isolated from TB patients included in a randomized phase I clinical trial of standard TB treatment with (n=21) or without (n=18) adjunctive COX-2i (etoricoxib) were analyzed at baseline, day 14 and day 56. Plasma eicosanoids were analyzed by ELISA and liquid chromatography-mass spectrometry (LC-MS), plasma cytokines by multiplex, and monocyte signaling by phospho-flow with a defined set of phospho-specific antibodies. Results Lipoxygenase (LOX)-derived products (LXA4 and 12-HETE) and pro-inflammatory cytokines were associated with TB disease severity and were reduced during TB therapy, possibly accelerated by adjunctive COX-2i. Phosphorylation of p38 MAPK, NFkB, Erk1/2, and Akt in monocytes as well as plasma levels of MIG/CXCL9 and procalcitonin were reduced in the COX-2i group compared to controls. Conclusion COX-2i may reduce excess inflammation in TB via the LOX-pathway in addition to modulation of phosphorylation patterns in monocytes. Immunomodulatory effects of adjunctive COX-2i in TB should be further investigated before recommended for use as a HDT strategy.
Collapse
Affiliation(s)
- Marthe Jøntvedt Jørgensen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Kristin G Nore
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Emilie Layre
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Kjetil Tasken
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Deparment of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Dag Kvale
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
27
|
Kirwan DE, Chong DLW, Friedland JS. Platelet Activation and the Immune Response to Tuberculosis. Front Immunol 2021; 12:631696. [PMID: 34093524 PMCID: PMC8170316 DOI: 10.3389/fimmu.2021.631696] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/04/2021] [Indexed: 12/24/2022] Open
Abstract
In 2019 10 million people developed symptomatic tuberculosis (TB) disease and 1.2 million died. In active TB the inflammatory response causes tissue destruction, which leads to both acute morbidity and mortality. Tissue destruction in TB is driven by host innate immunity and mediated via enzymes, chiefly matrix metalloproteinases (MMPs) which are secreted by leukocytes and stromal cells and degrade the extracellular matrix. Here we review the growing evidence implicating platelets in TB immunopathology. TB patients typically have high platelet counts, which correlate with disease severity, and a hypercoagulable profile. Platelets are present in human TB granulomas and platelet-associated gene transcripts are increased in TB patients versus healthy controls. Platelets most likely drive TB immunopathology through their effect on other immune cells, particularly monocytes, to lead to upregulation of activation markers, increased MMP secretion, and enhanced phagocytosis. Finally, we consider current evidence supporting use of targeted anti-platelet agents in the treatment of TB due to growing interest in developing host-directed therapies to limit tissue damage and improve treatment outcomes. In summary, platelets are implicated in TB disease and contribute to MMP-mediated tissue damage via their cellular interactions with other leukocytes, and are potential targets for novel host-directed therapies.
Collapse
Affiliation(s)
- Daniela E Kirwan
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| | - Deborah L W Chong
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| | - Jon S Friedland
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| |
Collapse
|
28
|
Hayford FEA, Ozturk M, Dolman RC, Blaauw R, Nienaber A, Loots DT, Brombacher F, Smuts CM, Parihar SP, Malan L. Longer-Term Omega-3 LCPUFA More Effective Adjunct Therapy for Tuberculosis Than Ibuprofen in a C3HeB/FeJ Tuberculosis Mouse Model. Front Immunol 2021; 12:659943. [PMID: 33995381 PMCID: PMC8113969 DOI: 10.3389/fimmu.2021.659943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022] Open
Abstract
Advancement in the understanding of inflammation regulation during tuberculosis (TB) treatment has led to novel therapeutic approaches being proposed. The use of immune mediators like anti-inflammatory and pro-resolving molecules for such, merits attention. Drug repurposing is a widely used strategy that seeks to identify new targets to treat or manage diseases. The widely explored nonsteroidal anti-inflammatory drug (NSAID) ibuprofen and a more recently explored pharmaconutrition therapy using omega-3 long-chain polyunsaturated fatty acids (n-3 LCPUFAs), have the potential to modulate the immune system and are thus considered potential repurposed drugs in this context. These approaches may be beneficial as supportive therapy to the already existing treatment regimen to improve clinical outcomes. Here, we applied adjunct ibuprofen and n-3 LCPUFA therapy, respectively, with standard anti-TB treatment, in a C3HeB/FeJ murine model of TB. Bacterial loads, lung pathology, lung cytokines/chemokines and lung lipid mediators were measured as outcomes. Lung bacterial load on day 14 post-treatment (PT) was lower in the n-3 LCPUFA, compared to the ibuprofen group (p = 0.039), but was higher in the ibuprofen group than the treated control group (p = 0.0315). Treated control and ibuprofen groups had more free alveolar space initially as compared to the n-3 LCPUFA group (4 days PT, p= 0.0114 and p= 0.002, respectively); however, significantly more alveolar space was present in the n-3 LCPUFA group as compared to the ibuprofen group by end of treatment (14 days PT, p = 0.035). Interleukin 6 (IL-6) was lower in the ibuprofen group as compared to the treated control, EPA/DHA and untreated control groups at 4 days PT (p = 0.019, p = 0.019 and p = 0.002, respectively). Importantly, pro-resolving EPA derived 9-HEPE, 11-HEPE, 12-HEPE and 18-HEPE lipid mediators (LMs) were significantly higher in the EPA/DHA group as compared to the ibuprofen and treated control groups. This suggests that n-3 LCPUFAs do improve pro-resolving and anti-inflammatory properties in TB, and it may be safe and effective to co-administer as adjunct therapy with standard TB treatment, particularly longer-term. Also, our results show host benefits upon short-term co-administration of ibuprofen, but not throughout the entire TB treatment course.
Collapse
Affiliation(s)
- Frank E. A. Hayford
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
- Department of Dietetics, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Mumin Ozturk
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town, South Africa
| | - Robin C. Dolman
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Renee Blaauw
- Division of Human Nutrition, Stellenbosch University, Tygerberg, Cape Town, South Africa
| | - Arista Nienaber
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Laboratory of Infectious Disease Metabolomics, Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Cornelius M. Smuts
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Suraj P. Parihar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Division of Medical Microbiology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Linda Malan
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| |
Collapse
|
29
|
Rankine-Wilson LI, Shapira T, Sao Emani C, Av-Gay Y. From infection niche to therapeutic target: the intracellular lifestyle of Mycobacterium tuberculosis. MICROBIOLOGY (READING, ENGLAND) 2021; 167:001041. [PMID: 33826491 PMCID: PMC8289223 DOI: 10.1099/mic.0.001041] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/15/2021] [Indexed: 12/16/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is an obligate human pathogen killing millions of people annually. Treatment for tuberculosis is lengthy and complicated, involving multiple drugs and often resulting in serious side effects and non-compliance. Mtb has developed numerous complex mechanisms enabling it to not only survive but replicate inside professional phagocytes. These mechanisms include, among others, overcoming the phagosome maturation process, inhibiting the acidification of the phagosome and inhibiting apoptosis. Within the past decade, technologies have been developed that enable a more accurate understanding of Mtb physiology within its intracellular niche, paving the way for more clinically relevant drug-development programmes. Here we review the molecular biology of Mtb pathogenesis offering a unique perspective on the use and development of therapies that target Mtb during its intracellular life stage.
Collapse
Affiliation(s)
| | - Tirosh Shapira
- Division of Infectious Disease, Department of Medicine, The University of British Columbia, Vancouver, Canada
| | - Carine Sao Emani
- Division of Infectious Disease, Department of Medicine, The University of British Columbia, Vancouver, Canada
| | - Yossef Av-Gay
- Department of Microbiology & Immunology, The University of British Columbia, Vancouver, Canada
- Division of Infectious Disease, Department of Medicine, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
30
|
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
|
31
|
Crilly NP, Ayeh SK, Karakousis PC. The New Frontier of Host-Directed Therapies for Mycobacterium avium Complex. Front Immunol 2021; 11:623119. [PMID: 33552087 PMCID: PMC7862709 DOI: 10.3389/fimmu.2020.623119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/14/2020] [Indexed: 01/03/2023] Open
Abstract
Mycobacterium avium complex (MAC) is an increasingly important cause of morbidity and mortality, and is responsible for pulmonary infection in patients with underlying lung disease and disseminated disease in patients with AIDS. MAC has evolved various virulence strategies to subvert immune responses and persist in the infected host. Current treatment for MAC is challenging, requiring a combination of multiple antibiotics given over a long time period (for at least 12 months after negative sputum culture conversion). Moreover, even after eradication of infection, many patients are left with residual lung dysfunction. In order to address similar challenges facing the management of patients with tuberculosis, recent attention has focused on the development of novel adjunctive, host-directed therapies (HDTs), with the goal of accelerating the clearance of mycobacteria by immune defenses and reducing or reversing mycobacterial-induced lung damage. In this review, we will summarize the evidence supporting specific adjunctive, HDTs for MAC, with a focus on the repurposing of existing immune-modulatory agents targeting a variety of different cellular pathways. We also highlight areas meriting further investigation.
Collapse
Affiliation(s)
- Nathan P Crilly
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Samuel K Ayeh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Petros C Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
32
|
Jøntvedt Jørgensen M, Jenum S, Tonby K, Mortensen R, Walzl G, Du Plessis N, Dyrhol-Riise AM. Monocytic myeloid-derived suppressor cells reflect tuberculosis severity and are influenced by cyclooxygenase-2 inhibitors. J Leukoc Biol 2020; 110:177-186. [PMID: 33155730 PMCID: PMC8359170 DOI: 10.1002/jlb.4a0720-409rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 10/02/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
Myeloid‐derived suppressor cells (MDSCs) increase in tuberculosis (TB) and may be targets for host‐directed therapy (HDT). In this study, we use flow cytometry to analyze the effects of cyclooxygenase‐2 inhibitors (COX‐2i) on monocytic (M)‐MDSCs in blood from TB patients attending a clinical trial of COX‐2i. The effects of COX‐2i on M‐MDSCs and mycobacterial uptake were also studied by an in vitro mycobacterial infection model. We found that M‐MDSC frequencies correlated with TB disease severity. Reduced M‐MDSC (P = 0.05) and IDO (P = 0.03) expression was observed in the COX‐2i group. We show that peripheral blood‐derived M‐MDSCs successfully internalized Mycobacterium bovis and that in vitro mycobacterial infection increased COX‐2 (P = 0.002), PD‐L1 (P = 0.01), and Arginase‐1 (P = 0.002) expression in M‐MDSCs. Soluble IL‐1β, IL‐10, and S100A9 were reduced in COX‐2i‐treated M‐MDSCs cultures (P < 0.05). We show novel data that COX‐2i had limited effect in vivo but reduced M‐MDSC cytokine production in vitro. The relevance of COX‐2i in a HDT strategy needs to be further explored.
Collapse
Affiliation(s)
- Marthe Jøntvedt Jørgensen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Gerhard Walzl
- 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, South Africa
| | - Nelita Du Plessis
- 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, South Africa
| | - Anne Ma Dyrhol-Riise
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
33
|
Negatu DA, Gengenbacher M, Dartois V, Dick T. Indole Propionic Acid, an Unusual Antibiotic Produced by the Gut Microbiota, With Anti-inflammatory and Antioxidant Properties. Front Microbiol 2020; 11:575586. [PMID: 33193190 PMCID: PMC7652848 DOI: 10.3389/fmicb.2020.575586] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
Most antibiotics are produced by soil microbes and typically interfere with macromolecular synthesis processes as their antibacterial mechanism of action. These natural products are often large and suffer from poor chemical tractability. Here, we discuss discovery, mechanism of action, and the therapeutic potentials of an unusual antibiotic, indole propionic acid (IPA). IPA is produced by the human gut microbiota. The molecule is small, chemically tractable, and targets amino acid biosynthesis. IPA is active against a broad spectrum of mycobacteria, including drug resistant Mycobacterium tuberculosis and non-tuberculous mycobacteria (NTM). Interestingly, the microbiota-produced metabolite is detectable in the serum of healthy individuals, tuberculosis (TB) patients, and several animal models. Thus, the microbiota in our gut may influence susceptibility to mycobacterial diseases. If a gut-lung microbiome axis can be demonstrated, IPA may have potential as a biomarker of disease progression, and development of microbiota-based therapies could be explored. In addition to its antimycobacterial activity, the molecule displays anti-inflammatory and antioxidant properties. This raises the possibility that IPA has therapeutic potential as both antibiotic and add-on host-directed drug for the treatment of TB in patient populations where disease morbidity and mortality is driven by excessive inflammation and tissue damage, such as TB-associated immune reconstitution inflammatory syndrome, TB-meningitis, and TB-diabetes.
Collapse
Affiliation(s)
- Dereje Abate Negatu
- Center for Innovative Drug Development and Therapeutic Trials for Africa, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia.,Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Martin Gengenbacher
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States.,Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States.,Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States.,Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, United States.,Department of Microbiology and Immunology, Georgetown University, Washington, DC, United States
| |
Collapse
|
34
|
Seddon JA, Johnson S, Palmer M, van der Zalm MM, Lopez-Varela E, Hughes J, Schaaf HS. Multidrug-resistant tuberculosis in children and adolescents: current strategies for prevention and treatment. Expert Rev Respir Med 2020; 15:221-237. [PMID: 32965141 DOI: 10.1080/17476348.2021.1828069] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION An estimated 30,000 children develop multidrug-resistant (MDR) tuberculosis (TB) each year, with only a small proportion diagnosed and treated. This field has historically been neglected due to the perception that children with MDR-TB are challenging to diagnose and treat. Diagnostic and therapeutic developments in adults have improved pediatric management, yet further pediatric-specific research and wider implementation of evidence-based practices are required. AREAS COVERED This review combines the most recent data with expert opinion to highlight best practice in the evaluation, diagnosis, treatment, and support of children and adolescents with MDR-TB disease. A literature search of PubMed was carried out on topics related to MDR-TB in children. This review provides practical advice on MDR-TB prevention and gives updates on new regimens and novel treatments. The review also addresses host-directed therapy, comorbid conditions, special populations, psychosocial support, and post-TB morbidity, as well as identifying outstanding research questions. EXPERT OPINION Increased availability of molecular diagnostics has the potential to aid with the diagnosis of MDR-TB in children. Shorter MDR-TB disease treatment regimens have made therapy safer and shorter and further developments with novel agents and repurposed drugs should lead to additional improvements. The evidence base for MDR-TB preventive therapy is increasing.
Collapse
Affiliation(s)
- James A Seddon
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University , Stellenbosch, South Africa.,Section of Paediatric Infectious Diseases, Department of Infectious Diseases, Imperial College London , London, UK
| | - Sarah Johnson
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University , Stellenbosch, South Africa.,Section of Paediatric Infectious Diseases, Department of Infectious Diseases, Imperial College London , London, UK
| | - Megan Palmer
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University , Stellenbosch, South Africa
| | - Marieke M van der Zalm
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University , Stellenbosch, South Africa
| | - Elisa Lopez-Varela
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University , Stellenbosch, South Africa.,ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic - Universitat De Barcelona , Barcelona, Spain
| | - Jennifer Hughes
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University , Stellenbosch, South Africa
| | - H Simon Schaaf
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University , Stellenbosch, South Africa
| |
Collapse
|
35
|
Shapira T, Rankine-Wilson L, Chao JD, Pichler V, Rens C, Pfeifer T, Av-Gay Y. High-Content Screening of Eukaryotic Kinase Inhibitors Identify CHK2 Inhibitor Activity Against Mycobacterium tuberculosis. Front Microbiol 2020; 11:553962. [PMID: 33042061 PMCID: PMC7530171 DOI: 10.3389/fmicb.2020.553962] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022] Open
Abstract
A screen of a eukaryotic kinase inhibitor library in an established intracellular infection model identified a set of drug candidates enabling intracellular killing of Mycobacterium tuberculosis (M.tb). Screen validity was confirmed internally by a Z′ = 0.5 and externally by detecting previously reported host-targeting anti-M.tb compounds. Inhibitors of the CHK kinase family, specifically checkpoint kinase 2 (CHK2), showed the highest inhibition and lowest toxicity of all kinase families. The screen identified and validated DDUG, a CHK2 inhibitor, as a novel bactericidal anti-M.tb compound. CHK2 inhibition by RNAi phenocopied the intracellular inhibitory effect of DDUG. DDUG was active intracellularly against M.tb, but not other mycobacteria. DDUG also had extracellular activity against 4 of 12 bacteria tested, including M.tb. Combined, these observations suggest DDUG acts in tandem against both host and pathogen. Importantly, DDUG’s validation highlights the screening and analysis methodology developed for this screen, which identified novel host-directed anti-M.tb compounds.
Collapse
Affiliation(s)
- Tirosh Shapira
- Division of Infectious Diseases, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Leah Rankine-Wilson
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Joseph D Chao
- Division of Infectious Diseases, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Virginia Pichler
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Celine Rens
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Tom Pfeifer
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Yossef Av-Gay
- Division of Infectious Diseases, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
36
|
Hayford FEA, Dolman RC, Blaauw R, Nienaber A, Smuts CM, Malan L, Ricci C. The effects of anti-inflammatory agents as host-directed adjunct treatment of tuberculosis in humans: a systematic review and meta-analysis. Respir Res 2020; 21:223. [PMID: 32847532 PMCID: PMC7448999 DOI: 10.1186/s12931-020-01488-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The potential role of adjunctive anti-inflammatory therapy to enhance tuberculosis (TB) treatment has recently received increasing interest. There is, therefore, a need to broadly examine current host-directed therapies (HDTs) that could accelerate treatment response and improve TB outcomes. METHODS This systematic review and meta-analysis included randomised controlled trials of vitamin D and other HDT agents in patients receiving antibiotic treatment for pulmonary TB. Sputum smear conversion rate at 4-8 weeks was the primary outcome. Secondary outcomes included blood indices associated with infectivity and inflammation, chest radiology and incidence of adverse events. RESULTS Fifty-five studies were screened for eligibility after the initial search, which yielded more than 1000 records. Of the 2540 participants in the 15 trials included in the meta-analysis, 1898 (74.7%) were male, and the age at entry ranged from 18 to 70 years. There was a 38% significantly (RR 1.38, 95% CI = 1.03-1.84) increased sputum smear negativity in patients administered with vitamin D in addition to standard TB treatment than those receiving only the TB treatment. Patients treated with other HDT anti-inflammatory agents in addition to TB treatment also had a 29% significantly increased sputum smear conversion rate (RR 1.29, 95% CI = 1.09-1.563). Lymphocyte to monocyte ratio was significantly higher in the vitamin D treatment groups compared to the controls (3.52 vs 2.70, 95% CI for difference 0.16-1.11, p = 0.009) and (adjusted mean difference 0.4, 95% CI 0.2 -- 0.6; p = 0.001); whilst tumour necrosis factor-alpha (TNF-α) showed a trend towards a reduction in prednisolone (p < 0.001) and pentoxifylline (p = 0.27) treatment groups. Vitamin D and N-acetylcysteine also accelerated radiographic resolution in treatment compared to placebo at 8 weeks. No differences were observed in the occurrence of adverse events among all HDT treatments. CONCLUSIONS Vitamin D and other anti-inflammatory HDT medications used as adjunct TB treatment may be well tolerated and effective. They significantly improved sputum smear conversion rate and chest radiological appearance, and also exhibited an inflammation resolution effect.
Collapse
Affiliation(s)
- Frank Ekow Atta Hayford
- Centre of Excellence for Nutrition, Faculty of Health Sciences, Building G16, North- West University, Potchefstroom Campus, Potchefstroom, South Africa. .,Department of Nutrition and Dietetics, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana.
| | - Robin Claire Dolman
- Centre of Excellence for Nutrition, Faculty of Health Sciences, Building G16, North- West University, Potchefstroom Campus, Potchefstroom, South Africa
| | - Renee Blaauw
- Division of Human Nutrition, Stellenbosch University, Cape Town, South Africa
| | - Arista Nienaber
- Centre of Excellence for Nutrition, Faculty of Health Sciences, Building G16, North- West University, Potchefstroom Campus, Potchefstroom, South Africa
| | - Cornelius Mattheus Smuts
- Centre of Excellence for Nutrition, Faculty of Health Sciences, Building G16, North- West University, Potchefstroom Campus, Potchefstroom, South Africa
| | - Linda Malan
- Centre of Excellence for Nutrition, Faculty of Health Sciences, Building G16, North- West University, Potchefstroom Campus, Potchefstroom, South Africa
| | - Cristian Ricci
- Centre of Excellence for Nutrition, Faculty of Health Sciences, Building G16, North- West University, Potchefstroom Campus, Potchefstroom, South Africa.,Department of Pediatric Epidemiology, Department of Pediatrics, Medical Faculty , University/Institution: Leipzig University, Leipzig, Germany
| |
Collapse
|
37
|
Wang W, Du Z, Ni M, Wang Z, Liang M, Sheng H, Zhang A, Yang J. Aspirin enhances the clinical efficacy of anti-tuberculosis therapy in pulmonary tuberculosis in patients with type 2 diabetes mellitus. Infect Dis (Lond) 2020; 52:721-729. [PMID: 32552387 DOI: 10.1080/23744235.2020.1778177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: Tuberculosis in patients with diabetes mellitus is characterised by rapid disease progression, poor treatment efficacy, poor prognosis and poses a new challenge in tuberculosis treatment and control.Methods: Patients with pulmonary TB and type 2 DM were recruited at Yijishan Hospital of Wannan Medical College. A total of 348 patients were randomly assigned to two groups. The aspirin group (aspirin + TB/DM) included 174 patients who received anti-TB therapy and enteric-coated aspirin tablets (100 mg/tablet). The control group (placebo + TB/DM) included 174 patients who received anti-TB therapy and enteric-coated placebo tablets (an identical tablet containing no drug). Eighty-two patients in the aspirin group and 86 in the control group completed the trial and were included in the analysis. Clinical characteristics, laboratory test results, imaging data and side effects of aspirin were monitored.Results: Aspirin treatment affect certain signs and symptoms. The erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) levels were lower in the aspirin group than in the control group after treatment (Both p = .000). The sputum-negative conversion rate was 86.7% in the aspirin group, significantly higher than in the control group (53.8%) (p = .031). After two months of treatment, the differences in the number of cases with cavities, the number of cavities, and maximum diameter of cavities in the aspirin group were statistically significant (p = .003, p = .023 and p = .015 respectively).Conclusion: Our findings suggest that aspirin may improve treatment in patients with pulmonary TB and type 2 DM.
Collapse
Affiliation(s)
- Wenjie Wang
- Department of Infectious Diseases, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| | - Zhixiang Du
- Department of Infectious Diseases, The People's Hospital of Taizhou, Taizhou, P. R. China
| | - Mingyue Ni
- Department of Infectious Diseases, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| | - Zijian Wang
- Department of Infectious Diseases, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| | - Manman Liang
- Department of Infectious Diseases, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| | - Haoyu Sheng
- Department of Infectious Diseases, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| | - Aiping Zhang
- Department of Infectious Diseases, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| | - Jianghua Yang
- Department of Infectious Diseases, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| |
Collapse
|
38
|
Ahmad S, Bhattacharya D, Gupta N, Rawat V, Tousif S, Van Kaer L, Das G. Clofazimine enhances the efficacy of BCG revaccination via stem cell-like memory T cells. PLoS Pathog 2020; 16:e1008356. [PMID: 32437421 PMCID: PMC7269335 DOI: 10.1371/journal.ppat.1008356] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/03/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022] Open
Abstract
Tuberculosis (TB) is one of the deadliest diseases, claiming ~2 million deaths annually worldwide. The majority of people in TB endemic regions are vaccinated with Bacillus Calmette Guerin (BCG), which is the only usable vaccine available. BCG is efficacious against meningeal and disseminated TB in children, but protective responses are relatively short-lived and fail to protect against adult pulmonary TB. The longevity of vaccine efficacy critically depends on the magnitude of long-lasting central memory T (TCM) cells, a major source of which is stem cell-like memory T (TSM) cells. These TSM cells exhibit enhanced self-renewal capacity as well as to rapidly respond to antigen and generate protective poly-functional T cells producing IFN-γ, TNF-α, IL-2 and IL-17. It is now evident that T helper Th 1 and Th17 cells are essential for host protection against TB. Recent reports have indicated that Th17 cells preserve the molecular signature for TSM cells, which eventually differentiate into IFN-γ-producing effector cells. BCG is ineffective in inducing Th17 cell responses, which might explain its inadequate vaccine efficacy. Here, we show that revaccination with BCG along with clofazimine treatment promotes TSM differentiation, which continuously restores TCM and T effector memory (TEM) cells and drastically increases vaccine efficacy in BCG-primed animals. Analyses of these TSM cells revealed that they are predominantly precursors to host protective Th1 and Th17 cells. Taken together, these findings revealed that clofazimine treatment at the time of BCG revaccination provides superior host protection against TB by increasing long-lasting TSM cells. Tuberculosis (TB) is one of the deadliest diseases, claiming ~2 million deaths annually worldwide. Bacillus Calmette Guerin (BCG) is the only usable vaccine available and exhibits efficacy against meningeal and disseminated TB in children. Consequently, the vast majority of people in TB endemic regions are vaccinated with BCG. However, host protective immune responses diminish over time due to gradual depletion of T central memory (TCM) cells, which are responsible for long-term host protection. Here, we provide evidence that revaccination with BCG along with the clofazimine, an approved drug for treatment of leprosy and drug-resistant TB, induces stem cell-like memory T (TSM) cells. TSM cells are precursors to TCM cells, and provide long-term host protection to TB by continuous supply of TCM cells. Interestingly, these TSM cells were generated from IL-17-producing T helper (Th)17 cells. These TSM cells differentiated into TCM and T effector memory (TEM) cells and maintained a stable pool of critically important Th1 and Th17 cells to provide optimal host protection against TB.
Collapse
Affiliation(s)
- Shaheer Ahmad
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | | | - Neeta Gupta
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Varsha Rawat
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Sultan Tousif
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Gobardhan Das
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
- * E-mail:
| |
Collapse
|
39
|
Kewcharoenwong C, Saenwongsa W, Willcocks SJ, Bancroft GJ, Fletcher HA, Lertmemongkolchai G. Glibenclamide alters interleukin-8 and interleukin-1β of primary human monocytes from diabetes patients against Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 2020; 123:101939. [PMID: 32452426 DOI: 10.1016/j.tube.2020.101939] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/25/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is an important risk factor for development of tuberculosis (TB). Our previous study showed glibenclamide, an anti-diabetic drug used to control blood glucose concentration, reduced interleukin (IL)-8 secretion from primary human monocytes challenged with M. tuberculosis (Mtb). In mice infected with Mtb, IL-1β is essential for host resistance through the enhancement of cyclooxygenase that limits excessive Type I interferon (IFN) production and fosters Mtb containment. We hypothesize that glibenclamide may also interfere with monocyte mediated immune responses against Mtb and alter the balance between IL-1β and IFNα-mediated immunity. Purified monocytes from non-diabetic and diabetic individuals were infected with Mtb or M. bovis BCG. We demonstrate that monocytes from diabetes patients who were being treated with glibenclamide showed reduced IL-1β and IL-8 secretion when exposed to Mtb. Additionally, these responses also occurred when monocytes from non-diabetic individuals were pre-treated with glibenclamide in vitro. Moreover, this pre-treatment enhanced IFNa1 expression but was not involved with prostaglandin E2 (PGE2) expression in response to Mtb infection. Taken together, our data show that glibenclamide might exacerbate susceptibility of diabetes patients to Mtb infection by reducing IL-1β and IL-8 production by monocytes.
Collapse
Affiliation(s)
- Chidchamai Kewcharoenwong
- The Centre for Research & Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Thailand
| | - Wipawee Saenwongsa
- The Centre for Research & Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Thailand; Disease Prevention and Control Region 10th, Ubonratchathani, Ministry of Public Healthy, Thailand
| | - Samuel J Willcocks
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, UK
| | - Gregory J Bancroft
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, UK; Tuberculosis Centre, London School of Hygiene and Tropical Medicine, UK
| | - Helen A Fletcher
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, UK; Tuberculosis Centre, London School of Hygiene and Tropical Medicine, UK
| | - Ganjana Lertmemongkolchai
- The Centre for Research & Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Thailand.
| |
Collapse
|
40
|
Potential anti-TB investigational compounds and drugs with repurposing potential in TB therapy: a conspectus. Appl Microbiol Biotechnol 2020; 104:5633-5662. [PMID: 32372202 DOI: 10.1007/s00253-020-10606-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/27/2020] [Accepted: 04/05/2020] [Indexed: 02/07/2023]
Abstract
The latest WHO report estimates about 1.6 million global deaths annually from TB, which is further exacerbated by drug-resistant (DR) TB and comorbidities with diabetes and HIV. Exiguous dosing, incomplete treatment course, and the ability of the tuberculosis bacilli to tolerate and survive current first-line and second-line anti-TB drugs, in either their latent state or active state, has resulted in an increased prevalence of multidrug-resistant (MDR), extensively drug-resistant (XDR), and totally drug-resistant TB (TDR-TB). Although a better understanding of the TB microanatomy, genome, transcriptome, proteome, and metabolome, has resulted in the discovery of a few novel promising anti-TB drug targets and diagnostic biomarkers of late, no new anti-TB drug candidates have been approved for routine therapy in over 50 years, with only bedaquiline, delamanid, and pretomanid recently receiving tentative regulatory approval. Considering this, alternative approaches for identifying possible new anti-TB drug candidates, for effectively eradicating both replicating and non-replicating Mycobacterium tuberculosis, are still urgently required. Subsequently, several antibiotic and non-antibiotic drugs with known treatment indications (TB targeted and non-TB targeted) are now being repurposed and/or derivatized as novel antibiotics for possible use in TB therapy. Insights gathered here reveal that more studies focused on drug-drug interactions between licensed and potential lead anti-TB drug candidates need to be prioritized. This write-up encapsulates the most recent findings regarding investigational compounds with promising anti-TB potential and drugs with repurposing potential in TB therapy.
Collapse
|
41
|
Host-Directed Therapies and Anti-Virulence Compounds to Address Anti-Microbial Resistant Tuberculosis Infection. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10082688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite global efforts to contain tuberculosis (TB), the disease remains a leading cause of morbidity and mortality worldwide, further exacerbated by the increased resistance to antibiotics displayed by the tubercle bacillus Mycobacterium tuberculosis. In order to treat drug-resistant TB, alternative or complementary approaches to standard anti-TB regimens are being explored. An area of active research is represented by host-directed therapies which aim to modulate the host immune response by mitigating inflammation and by promoting the antimicrobial activity of immune cells. Additionally, compounds that reduce the virulence of M. tuberculosis, for instance by targeting the major virulence factor ESX-1, are being given increased attention by the TB research community. This review article summarizes the current state of the art in the development of these emerging therapies against TB.
Collapse
|
42
|
Rizvi I, Garg RK, Malhotra HS, Kumar N, Uniyal R. Role of Aspirin in Tuberculous Meningitis: A Systematic Review and Meta-analysis. Neurol India 2020; 67:993-1002. [PMID: 31512620 DOI: 10.4103/0028-3886.266232] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Objectives Aspirin is a drug that has been found to be useful in reducing the incidence of infarctions. This systemic is aimed at review evaluating the benefits of aspirin in the management of tuberculous meningitis. Methods A systematic literature search was performed using PubMed, Cochrane Central Register of Controlled Trials, and SCOPUS for articles published on or before September 22, 2018. The meta-register of controlled trials and bibliography was also searched. Randomized controlled trials wherein aspirin was used were included in the systematic review. The data was extracted using a predetermined format. The risk ratio (RR) for dichotomous data was calculated and a random-effects model was used to combine the data. Death and occurrence of new infarctions were considered as primary outcomes. The quality of evidence was assessed using the GRADE approach. Results Four trials including 546 patients were found eligible. The addition of aspirin to anti-tuberculosis drug regimens did not significantly reduce mortality [RR = 0.66 (0.42-1.02); low-quality evidence] but significantly reduced the risk of new infarctions [RR = 0.52 (0.29-0.92); moderate-quality evidence]. Aspirin did not differ from the placebo with regard to the adverse event outcome. Conclusion Aspirin reduces the risk of new infarctions in patients with tuberculous meningitis but does not affect mortality (moderate-to-low level of evidence).
Collapse
Affiliation(s)
- Imran Rizvi
- Department of Neurology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Ravindra K Garg
- Department of Neurology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Hardeep S Malhotra
- Department of Neurology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Neeraj Kumar
- Department of Neurology, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Ravi Uniyal
- Department of Neurology, King George Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
43
|
Hydrogen sulfide dysregulates the immune response by suppressing central carbon metabolism to promote tuberculosis. Proc Natl Acad Sci U S A 2020; 117:6663-6674. [PMID: 32139610 PMCID: PMC7104411 DOI: 10.1073/pnas.1919211117] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ubiquitous gasotransmitter hydrogen sulfide (H2S) has been recognized to play a crucial role in human health. Using cystathionine γ-lyase (CSE)-deficient mice, we demonstrate an unexpected role of H2S in Mycobacterium tuberculosis (Mtb) pathogenesis. We showed that Mtb-infected CSE-/- mice survive longer than WT mice, and support reduced pathology and lower bacterial burdens in the lung, spleen, and liver. Similarly, in vitro Mtb infection of macrophages resulted in reduced colony forming units in CSE-/- cells. Chemical complementation of infected WT and CSE-/- macrophages using the slow H2S releaser GYY3147 and the CSE inhibitor DL-propargylglycine demonstrated that H2S is the effector molecule regulating Mtb survival in macrophages. Furthermore, we demonstrate that CSE promotes an excessive innate immune response, suppresses the adaptive immune response, and reduces circulating IL-1β, IL-6, TNF-α, and IFN-γ levels in response to Mtb infection. Notably, Mtb infected CSE-/- macrophages show increased flux through glycolysis and the pentose phosphate pathway, thereby establishing a critical link between H2S and central metabolism. Our data suggest that excessive H2S produced by the infected WT mice reduce HIF-1α levels, thereby suppressing glycolysis and production of IL-1β, IL-6, and IL-12, and increasing bacterial burden. Clinical relevance was demonstrated by the spatial distribution of H2S-producing enzymes in human necrotic, nonnecrotic, and cavitary pulmonary tuberculosis (TB) lesions. In summary, CSE exacerbates TB pathogenesis by altering immunometabolism in mice and inhibiting CSE or modulating glycolysis are potential targets for host-directed TB control.
Collapse
|
44
|
Young C, Walzl G, Du Plessis N. Therapeutic host-directed strategies to improve outcome in tuberculosis. Mucosal Immunol 2020; 13:190-204. [PMID: 31772320 PMCID: PMC7039813 DOI: 10.1038/s41385-019-0226-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/15/2019] [Accepted: 10/23/2019] [Indexed: 02/04/2023]
Abstract
Bacille Calmette-Guérin (BCG) is the only licenced tuberculosis (TB) vaccine, but has limited efficacy against pulmonary TB disease development and modest protection against extrapulmonary TB. Preventative antibiotic treatment for Mycobacterium tuberculosis (Mtb) infections in high-prevalence settings is unfeasible due to unclear treatment durability, drug toxicity, logistical constraints related to directly observed treatment strategy (DOTS) and the lengthy treatment protocols. Together, these factors promote non-adherence, contributing to relapse and establishment of drug-resistant Mtb strains. Although antibiotic treatment of drug-susceptible Mtb is generally effective, drug-resistant TB has a treatment efficacy below 50% and can, in a proportion, develop into progressive, untreatable disease. Other immune compromising co-infections and/or co-morbidities require more complex prevention/treatment approaches, posing huge financial burdens to national health services. Novel TB treatment strategies, such as host-directed therapeutics, are required to complement pathogen-targeted approaches. Pre-clinical studies have highlighted promising candidates that enhance endogenous pathways and/or limit destructive host responses. This review discusses promising pre-clinical candidates and forerunning compounds at advanced stages of clinical investigation in TB host-directed therapeutic (HDT) efficacy trials. Such approaches are rationalized to improve outcome in TB and shorten treatment strategies.
Collapse
Affiliation(s)
- C Young
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology/DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - G Walzl
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology/DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - N Du Plessis
- South African Medical Research Council, Centre for Tuberculosis Research, Department of Science and Technology/DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
45
|
Rohwer N, Kühl AA, Ostermann AI, Hartung NM, Schebb NH, Zopf D, McDonald FM, Weylandt KH. Effects of chronic low-dose aspirin treatment on tumor prevention in three mouse models of intestinal tumorigenesis. Cancer Med 2020; 9:2535-2550. [PMID: 31994315 PMCID: PMC7131863 DOI: 10.1002/cam4.2881] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 12/29/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Although early detection and treatment of colorectal cancer (CRC) have improved, it remains a significant health-care problem with high morbidity and mortality. Data indicate that long-term intake of low-dose aspirin reduces the risk of CRC; however, the mechanisms underlying this chemopreventive effect are still unclear. Different mouse models for inflammation-associated, sporadic, and hereditary CRC were applied to assess the efficacy and mechanism of low-dose aspirin on tumor prevention. An initial dosing study performed in healthy mice indicates that aspirin at a dose of 25 mg/kg/d has a similar pharmacodynamic effect as low-dose aspirin treatment in human subjects (100 mg/d). Chronic low-dose aspirin treatment suppresses colitis-associated and to a lesser extent spontaneous tumorigenesis in mice. Aspirin's antitumor effect is most pronounced in a preventive approach when aspirin administration starts before the tumor-initiating genotoxic event and continues for the duration of the experiment. These effects are not associated with alterations in cell proliferation, apoptosis, or activation of signaling pathways involved in CRC. Aspirin-induced reduction in tumor burden is accompanied by inhibition of thromboxane B2 formation, indicating reduced platelet activation. Aspirin treatment also results in decreased colonic prostaglandin E2 formation and tumor angiogenesis. With respect to colitis-triggered tumorigenesis, aspirin administration is associated with a reduction in inflammatory activity in the colon, as indicated by decreased levels of pro-inflammatory mediators, and tumor-associated iNOS-positive macrophages. Our results suggest that low-dose aspirin represents an effective antitumor agent in the context of colon tumorigenesis primarily due to its well-established cyclooxygenase inhibition effects.
Collapse
Affiliation(s)
- Nadine Rohwer
- Medical Department, Division of Hepatology and Gastroenterology, Charite-Universitätsmedizin Berlin, Berlin, Germany.,Medical Department B, Divisions of Hepatology, Gastroenterology, Oncology, Hematology, Rheumatology, Endocrinology and Diabetes, Brandenburg Medical School, Ruppin General Hospital, Neuruppin, Germany.,Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Anja A Kühl
- iPATH.Berlin-Immunopathology for Experimental Models, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Annika I Ostermann
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nicole Marie Hartung
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Dieter Zopf
- Research and Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Fiona M McDonald
- Research and Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Karsten-H Weylandt
- Medical Department, Division of Hepatology and Gastroenterology, Charite-Universitätsmedizin Berlin, Berlin, Germany.,Medical Department B, Divisions of Hepatology, Gastroenterology, Oncology, Hematology, Rheumatology, Endocrinology and Diabetes, Brandenburg Medical School, Ruppin General Hospital, Neuruppin, Germany
| |
Collapse
|
46
|
Pierson E, Haufroid M, Gosain TP, Chopra P, Singh R, Wouters J. Identification and Repurposing of Trisubstituted Harmine Derivatives as Novel Inhibitors of Mycobacterium tuberculosis Phosphoserine Phosphatase. Molecules 2020; 25:E415. [PMID: 31963843 PMCID: PMC7024313 DOI: 10.3390/molecules25020415] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
Mycobacterium tuberculosis is still the deadliest bacterial pathogen worldwide and the increasing number of multidrug-resistant tuberculosis cases further complicates this global health issue. M. tuberculosis phosphoserine phosphatase SerB2 is a promising target for drug design. Besides being a key essential metabolic enzyme of the pathogen's serine pathway, it appears to be involved in immune evasion mechanisms. In this work, a malachite green-based phosphatase assay has been used to screen 122 compounds from an internal chemolibrary. Trisubstituted harmine derivatives were found among the best hits that inhibited SerB2 activity. Synthesis of an original compound helped to discuss a brief structure activity relationship evaluation. Kinetics experiments showed that the most potent derivatives inhibit the phosphatase in a parabolic competitive fashion with apparent inhibition constants ( K i ) values in the micromolar range. Their interaction modes with the enzyme were investigated through induced fit docking experiments, leading to results consistent with the experimental data. Cellular assays showed that the selected compounds also inhibited M. tuberculosis growth in vitro. Those promising results may provide a basis for the development of new antimycobacterial agents targeting SerB2.
Collapse
Affiliation(s)
- Elise Pierson
- Laboratoire de Chimie Biologique Structurale (CBS), Namur Medicine and Drug Innovation Center (NAMEDIC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), B-5000 Namur, Belgium
| | - Marie Haufroid
- Laboratoire de Chimie Biologique Structurale (CBS), Namur Medicine and Drug Innovation Center (NAMEDIC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), B-5000 Namur, Belgium
| | - Tannu Priya Gosain
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India
| | - Pankaj Chopra
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India
| | - Ramandeep Singh
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, Faridabad 121001, Haryana, India
| | - Johan Wouters
- Laboratoire de Chimie Biologique Structurale (CBS), Namur Medicine and Drug Innovation Center (NAMEDIC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), B-5000 Namur, Belgium
| |
Collapse
|
47
|
Phase III, placebo-controlled, randomized, double-blind trial of tableted, therapeutic TB vaccine (V7) containing heat-killed M. vaccae administered daily for one month. J Clin Tuberc Other Mycobact Dis 2019; 18:100141. [PMID: 31890902 PMCID: PMC6933248 DOI: 10.1016/j.jctube.2019.100141] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective Immunotherapy of tuberculosis (TB) to shorten treatment duration represents an unmet medical need. Orally delivered, tableted TB vaccine (V7) containing heat-killed Mycobacterium vaccae (NCTC 11659) has been demonstrated in prior clinical studies to be safe and fast-acting immune adjunct. Methods The outcome of Phase III trial of V7 containing 10 µg of hydrolyzed M. vaccae was evaluated in 152 patients randomized at 2:1 ratio: V7 (N = 100), placebo (N = 52). Both arms received conventional 1st or 2nd line TB drugs co-administered with daily pill of V7 or placebo. Results After one month mycobacterial clearance was observed in 68% (P < 0.0001) and 23.1% (P = 0.04) of patients on V7 and placebo. Stratified conversion rates in V7 recipients with drug-sensitive and multidrug-resistant TB were 86.7% and 55.6% vs 27.2% and 15% in placebo. Patients on V7 gained on average 2.4 kg (P < 0.0001) vs 0.3 kg (P = 0.18) in placebo. Improvements in hemoglobin levels, erythrocyte sedimentation rate and leukocyte counts were significantly better than in controls. Liver function tests revealed that V7 can prevent chemotherapy-induced hepatic damage. Conclusion Oral M. vaccae is safe, can overcome TB-associated weight loss and inflammation, reduce hepatotoxicity of TB drugs, improve sputum conversion three-fold OR 3.15; 95%CI (2.3,4.6), and cut treatment length by at least six-fold. Longer follow-up studies might be needed to further substantiate our findings (Clinicaltrials.gov: NCT01977768).
Collapse
|
48
|
Mortensen R, Clemmensen HS, Woodworth JS, Therkelsen ML, Mustafa T, Tonby K, Jenum S, Agger EM, Dyrhol-Riise AM, Andersen P. Cyclooxygenase inhibitors impair CD4 T cell immunity and exacerbate Mycobacterium tuberculosis infection in aerosol-challenged mice. Commun Biol 2019; 2:288. [PMID: 31396568 PMCID: PMC6683187 DOI: 10.1038/s42003-019-0530-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/02/2019] [Indexed: 01/06/2023] Open
Abstract
Tuberculosis, caused by infection with Mycobacterium tuberculosis (Mtb), kills over 1.6 million people each year despite availability of antibiotics. The increase in drug resistant Mtb strains is a major public health emergency and host-directed therapy as adjunct to antibiotic treatment has gained increased interest. Cyclooxygenase inhibitors (COXi) are frequently used drugs to alleviate tuberculosis related symptoms. Mouse studies of acute intravenous Mtb infection have suggested a potential benefit of COXi for host-directed therapy. Here we show that COXi treatment (ibuprofen and celecoxib) is detrimental to Mtb control in different mouse models of respiratory infection. This effect links to impairments of the Type-1 helper (Th1) T-cell response as CD4 T-cells in COXi-treated animals have significantly decreased Th1 differentiation, reduced IFNγ expression and decreased protective capacity upon adoptive transfer. If confirmed in clinical trials, these findings could have major impact on global health and question the use of COXi for host-directed therapy.
Collapse
Affiliation(s)
- Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | | | - Joshua S. Woodworth
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | - Marie Louise Therkelsen
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | - Tehmina Mustafa
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen & Department of Thoracic Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | - Anne Ma Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
49
|
Rao M, Dodoo E, Zumla A, Maeurer M. Immunometabolism and Pulmonary Infections: Implications for Protective Immune Responses and Host-Directed Therapies. Front Microbiol 2019; 10:962. [PMID: 31134013 PMCID: PMC6514247 DOI: 10.3389/fmicb.2019.00962] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022] Open
Abstract
The biology and clinical efficacy of immune cells from patients with infectious diseases or cancer are associated with metabolic programming. Host immune- and stromal-cell genetic and epigenetic signatures in response to the invading pathogen shape disease pathophysiology and disease outcomes. Directly linked to the immunometabolic axis is the role of the host microbiome, which is also discussed here in the context of productive immune responses to lung infections. We also present host-directed therapies (HDT) as a clinically viable strategy to refocus dysregulated immunometabolism in patients with infectious diseases, which requires validation in early phase clinical trials as adjuncts to conventional antimicrobial therapy. These efforts are expected to be continuously supported by newly generated basic and translational research data to gain a better understanding of disease pathology while devising new molecularly defined platforms and therapeutic options to improve the treatment of patients with pulmonary infections, particularly in relation to multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Ernest Dodoo
- Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London, NIHR Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| |
Collapse
|
50
|
Rao M, Ippolito G, Mfinanga S, Ntoumi F, Yeboah-Manu D, Vilaplana C, Zumla A, Maeurer M. Improving treatment outcomes for MDR-TB - Novel host-directed therapies and personalised medicine of the future. Int J Infect Dis 2019; 80S:S62-S67. [PMID: 30685590 DOI: 10.1016/j.ijid.2019.01.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/17/2019] [Accepted: 01/19/2019] [Indexed: 12/12/2022] Open
Abstract
Multidrug-resistant TB (MDR-TB) is a major threat to global health security. In 2017, only 50% of patients with MDR-TB who received WHO-recommended treatment were cured. Most MDR-TB patients who recover continue to suffer from functional disability due to long-term lung damage. Whilst new MDR-TB treatment regimens are becoming available, conventional drug therapies need to be complemented with host-directed therapies (HDTs) to reduce tissue damage and improve functional treatment outcomes. This viewpoint highlights recent data on biomarkers, immune cells, circulating effector molecules and genetics which could be utilised for developing personalised HDTs. Novel technologies currently used for cancer therapy which could facilitate in-depth understanding of host genetics and the microbiome in patients with MDR-TB are discussed. Against this background, personalised cell-based HDTs for adjunct MDR-TB treatment to improve clinical outcomes are proposed as a possibility for complementing standard therapy and other HDT agents. Insights into the molecular biology of the mechanisms of action of cellular HDTs may also aid to devise non-cell-based therapies targeting defined inflammatory pathway(s) in Mtb-driven immunopathology.
Collapse
Affiliation(s)
- Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| | - Giuseppe Ippolito
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy.
| | - Sayoki Mfinanga
- National Institute of Medical Research Muhimbili, Dar es Salaam, Tanzania.
| | - Francine Ntoumi
- University Marien NGouabi and Fondation Congolaise pour la Recherche Médicale (FCRM), Brazzaville, Congo; Institute for Tropical Medicine, University of Tübingen, Germany.
| | - Dorothy Yeboah-Manu
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, Accra, Ghana.
| | - Cris Vilaplana
- Experimental Tuberculosis Unit (UTE), Fundació Institut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona (UAB), Badalona, Catalonia, Spain.
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK.
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal; Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt am Main, Germany.
| |
Collapse
|