1
|
Singh M, Sarhan MO, Damiba NNL, Singh AK, Villabona-Rueda A, Nino-Meza OJ, Chen X, Masias-Leon Y, Ruiz-Gonzalez CE, Ordonez AA, D'Alessio FR, Aboagye EO, Carroll LS, Jain SK. Proapoptotic Bcl-2 inhibitor as potential host directed therapy for pulmonary tuberculosis. Nat Commun 2025; 16:3003. [PMID: 40148277 PMCID: PMC11950383 DOI: 10.1038/s41467-025-58190-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Mycobacterium tuberculosis establishes within host cells by inducing anti-apoptotic Bcl-2 family proteins, triggering necrosis, inflammation, and fibrosis. Here, we demonstrate that navitoclax, an orally bioavailable, small-molecule Bcl-2 inhibitor, significantly improves pulmonary tuberculosis (TB) treatments as a host-directed therapy. Addition of navitoclax to standard TB treatments at human equipotent dosing in mouse models of TB, inhibits Bcl-2 expression, leading to improved bacterial clearance, reduced tissue necrosis, fibrosis and decreased extrapulmonary bacterial dissemination. Using immunohistochemistry and flow cytometry, we show that navitoclax induces apoptosis in several immune cells, including CD68+ and CD11b+ cells. Finally, positron emission tomography (PET) in live animals using clinically translatable biomarkers for apoptosis (18F-ICMT-11) and fibrosis (18F-FAPI-74), demonstrates that navitoclax significantly increases apoptosis and reduces fibrosis in pulmonary tissues, which are confirmed in postmortem analysis. Our studies suggest that proapoptotic drugs such as navitoclax can potentially improve pulmonary TB treatments, reduce lung damage / fibrosis and may be protective against post-TB lung disease.
Collapse
Affiliation(s)
- Medha Singh
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mona O Sarhan
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nerketa N L Damiba
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alok K Singh
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | | | - Oscar J Nino-Meza
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xueyi Chen
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuderleys Masias-Leon
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carlos E Ruiz-Gonzalez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Franco R D'Alessio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Department of Surgery & Cancer, Hammersmith Campus, Imperial College, London, UK
| | - Laurence S Carroll
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Yun JS, Shin E, Lee YR, Lee JA, Lee H, Kim JS, Shin SJ, Ha SJ, Lee SW, Kim D, Yoo JS, Jeong HS. Immunogenicity and protective efficacy of a multi-antigenic adenovirus-based vaccine candidate against Mycobacterium tuberculosis. Front Microbiol 2025; 16:1492268. [PMID: 39927262 PMCID: PMC11802578 DOI: 10.3389/fmicb.2025.1492268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Introduction The inadequate efficacy of the Bacillus Calmette-Guérin (BCG) vaccine against adult pulmonary tuberculosis (TB) necessitates the development of new and effective vaccines. Human adenovirus serotype 5 (Ad5), which induces T-cell response, is a widely used viral vector. In this study, we aimed to evaluate the efficacy of a multi-antigenic recombinant Ad5 vectored vaccine and determine the optimal immunization route for enhanced immune response against Mycobacterium tuberculosis. Methods We constructed a multi-antigenic recombinant Ad5 vectored vaccine expressing four antigens (Ag85B-ESAT6-MPT64-Rv2660c) of M. tuberculosis (rAd-TB4), immunized with rAd-TB4 (5 × 107 infectious virus units/mouse) twice at an interval of 4 weeks starting at 10 weeks after BCG priming, and evaluated its boosting efficacy in a BCG-primed mouse model, and determined the optimal immunization route. Results Compared with the BCG-only (2 × 105 colony forming units/mouse), subcutaneous injection of rAd-TB4 (1 × 107 infectious virus units/mL; two doses) elicited a T-cell response and cytokine production in lung lymphocytes and splenocytes. rAd-TB4 immunization significantly reduced bacterial loads and inflamed lung areas compared to BCG immunization (p < 0.01) and protected against the H37Rv challenge performed at 17 weeks of BCG priming. RNA sequencing of the whole blood of rAd-TB4-vaccinated mice collected pre- and, 1 and 4 weeks post-infection, identified differentially expressed genes associated with immune and inflammatory responses, especially those in the Wnt signaling pathway. Conclusion Our results indicate that rAd-TB4 immunization enhances the immune response to the vaccine boosting antigens in BCG-primed mice, making it a potential adult pulmonary TB vaccine candidate.
Collapse
Affiliation(s)
- Jin-Seung Yun
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Eunkyung Shin
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Young-Ran Lee
- Bio-Convergence R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, Chungbuk, Republic of Korea
| | - Jung-Ah Lee
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Hyeokjin Lee
- Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Jong-Seok Kim
- Department of Cell Biology, College of Medicine, Myunggok Medical Research Institute, Konyang University, Daejeon, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sang-Won Lee
- Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Dokeun Kim
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Jung-Sik Yoo
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Hye-Sook Jeong
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| |
Collapse
|
3
|
Doghish AS, Abulsoud AI, Nassar YA, Nasr SM, Mohammed OA, Abdel-Reheim MA, Rizk NI, Lutfy RH, Abdel Mageed SS, Ismail MA, Abd-Elhalim HM, Awad FA, Fayez SZ, Elimam H, Mansour RM. Harnessing miRNAs: A Novel Approach to Diagnosis and Treatment of Tuberculosis. J Biochem Mol Toxicol 2025; 39:e70119. [PMID: 39799557 DOI: 10.1002/jbt.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/09/2024] [Accepted: 12/21/2024] [Indexed: 01/15/2025]
Abstract
Mycobacterium tuberculosis (Mtb) complex, responsible for tuberculosis (TB) infection, continues to be a predominant global cause of mortality due to intricate host-pathogen interactions that affect disease progression. MicroRNAs (miRNAs), essential posttranscriptional regulators, have become pivotal modulators of these relationships. Recent findings indicate that miRNAs actively regulate immunological responses to Mtb complex by modulating autophagy, apoptosis, and immune cell activities. This has resulted in increased interest in miRNAs as prospective diagnostic indicators for TB, especially in differentiating active infection from latent or inactive stages. Variations in miRNA expression during Mtb infection indicate disease progression and offer insights into the immune response. Furthermore, miRNAs present potential as therapeutic targets in host-directed therapy (HDT) techniques for TB infection. This work examines the function of miRNAs in TB pathogenesis, with the objective of identifying particular miRNAs that regulate the immune response to the Mtb complex, evaluating their diagnostic value and exploring their therapeutic implications in host-directed therapy for TB infection. The objective is to enhance comprehension of how miRNAs can facilitate improved diagnosis and treatment of TB.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Yara A Nassar
- Department of Botany, Biotechnology and Its Application Program, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Sami Mohamed Nasr
- Biochemistry and Molecular Biology, Theodor Bilharz Research Institute, Giza, Egypt
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | | | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Radwa H Lutfy
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
| | - Menattallah A Ismail
- Applied Biotechnology Program, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Haytham M Abd-Elhalim
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
- Agricultural Research Center, Agricultural Genetic Engineering Research Institute, Giza, Egypt
| | - Farah A Awad
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Salma Zaki Fayez
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Reda M Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan, Egypt
- Molecular Biology and Biotechnology Department, School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo, Egypt
| |
Collapse
|
4
|
Chen S, Lei Z, Sun T. The critical role of miRNA in bacterial zoonosis. Int Immunopharmacol 2024; 143:113267. [PMID: 39374566 DOI: 10.1016/j.intimp.2024.113267] [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: 05/06/2024] [Revised: 08/21/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024]
Abstract
The public's health and the financial sustainability of international societies remain threatened by bacterial zoonoses, with limited reliable diagnostic and therapeutic options available for bacterial diseases. Bacterial infections influence mammalian miRNA expression in host-pathogen interactions. In order to counteract bacterial infections, miRNAs participate in gene-specific expression and play important regulatory roles that rely on translational inhibition and target gene degradation by binding to the 3' non-coding region of target genes. Intriguingly, according to current studies, that exogenous miRNAs derived from plants could potentially serve as effective medicinal components sourced from traditional Chinese medicine plants. These exogenous miRNAs exhibit stable functionality in mammals and mimic the regulatory roles of endogenous miRNAs, illuminating the molecular processes behind the therapeutic effects of plants. This review details the immune defense mechanisms of inflammation, apoptosis, autophagy and cell cycle disturbance caused by some typical bacterial infections, summarizes the role of some mammalian miRNAs in regulating these mechanisms, and introduces the cGAS-STING signaling pathway in detail. Evidence suggests that this newly discovered immune defense mechanism in mammalian cells can also be affected by miRNAs. Meanwhile, some examples of transboundary regulation of mammalian mRNA and even bacterial diseases by exogenous miRNAs from plants are also summarized. This viewpoint provides fresh understanding of microbial tactics and host mechanisms in the management of bacterial illnesses.
Collapse
Affiliation(s)
- Si Chen
- School of Chemistry, Chemical Engineering and Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Disease, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Disease, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Disease, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| |
Collapse
|
5
|
Jain S, Singh M, Sarhan M, Damiba N, Singh A, Villabona-Rueda A, Meza ON, Chen X, Ordonez A, D'Alessio F, Aboagye E, Carroll L. Proapoptotic Bcl-2 inhibitor as host directed therapy for pulmonary tuberculosis. RESEARCH SQUARE 2024:rs.3.rs-4926508. [PMID: 39281866 PMCID: PMC11398574 DOI: 10.21203/rs.3.rs-4926508/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Mycobacterium tuberculosis establishes within host cells by inducing anti-apoptotic Bcl-2 family proteins, triggering necrosis, inflammation, and fibrosis. Here, we demonstrate that navitoclax, an orally bioavailable, small-molecule Bcl-2 inhibitor, significantly improves pulmonary tuberculosis (TB) treatments as a host-directed therapy. Addition of navitoclax to standard TB treatments at human equipotent dosing in mouse models of TB, inhibits Bcl-2 expression, leading to improved bacterial clearance, reduced tissue damage / fibrosis and decreased extrapulmonary bacterial dissemination. Using immunohistochemistry and flow cytometry, we show that navitoclax induces apoptosis in several immune cells, including CD68 + and CD11b + cells. Finally, positron emission tomography (PET) in live animals using novel, clinically translatable biomarkers for apoptosis (18F-ICMT-11) and fibrosis (18F-FAPI-74) demonstrates that navitoclax significantly increases apoptosis and reduces fibrosis in pulmonary tissues, which are confirmed using post-mortem studies. Our studies suggest that proapoptotic drugs such as navitoclax can improve pulmonary TB treatments, and should be evaluated in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Alok Singh
- Johns Hopkins University School of Medicine
| | | | | | - Xueyi Chen
- Johns Hopkins University School of Medicine
| | | | | | | | | |
Collapse
|
6
|
Rodríguez-González J, Gutiérrez-Kobeh L. Apoptosis and its pathways as targets for intracellular pathogens to persist in cells. Parasitol Res 2023; 123:60. [PMID: 38112844 PMCID: PMC10730641 DOI: 10.1007/s00436-023-08031-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
Apoptosis is a finely programmed process of cell death in which cells silently dismantle and actively participate in several operations such as immune response, differentiation, and cell growth. It can be initiated by three main pathways: the extrinsic, the perforin granzyme, and the intrinsic that culminate in the activation of several proteins in charge of tearing down the cell. On the other hand, apoptosis represents an ordeal for pathogens that live inside cells and maintain a strong dependency with them; thus, they have evolved multiple strategies to manipulate host cell apoptosis on their behalf. It has been widely documented that diverse intracellular bacteria, fungi, and parasites can interfere with most steps of the host cell apoptotic machinery to inhibit or induce apoptosis. Indeed, the inhibition of apoptosis is considered a virulence property shared by many intracellular pathogens to ensure productive replication. Some pathogens intervene at an early stage by interfering with the sensing of extracellular signals or transduction pathways. Others sense cellular stress or target the apoptosis regulator proteins of the Bcl-2 family or caspases. In many cases, the exact molecular mechanisms leading to the interference with the host cell apoptotic cascade are still unknown. However, intense research has been conducted to elucidate the strategies employed by intracellular pathogens to modulate host cell death. In this review, we summarize the main routes of activation of apoptosis and present several processes used by different bacteria, fungi, and parasites to modulate the apoptosis of their host cells.
Collapse
Affiliation(s)
- Jorge Rodríguez-González
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología "Ignacio Chávez,", Juan Badiano No. 1, Col. Belisario Domínguez, Sección XVI, Delegación Tlalpan, C.P. 14080, Ciudad de México, México
- Laboratorio de Estudios Epidemiológicos, Clínicos, Diseños Experimentales e Investigación, Facultad de Ciencias Químicas, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, Mexico
| | - Laila Gutiérrez-Kobeh
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología "Ignacio Chávez,", Juan Badiano No. 1, Col. Belisario Domínguez, Sección XVI, Delegación Tlalpan, C.P. 14080, Ciudad de México, México.
| |
Collapse
|
7
|
Rahlwes KC, Dias BR, Campos PC, Alvarez-Arguedas S, Shiloh MU. Pathogenicity and virulence of Mycobacterium tuberculosis. Virulence 2023; 14:2150449. [PMID: 36419223 PMCID: PMC9817126 DOI: 10.1080/21505594.2022.2150449] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 11/17/2022] [Indexed: 11/27/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis, an infectious disease with one of the highest morbidity and mortality rates worldwide. Leveraging its highly evolved repertoire of non-protein and protein virulence factors, Mtb invades through the airway, subverts host immunity, establishes its survival niche, and ultimately escapes in the setting of active disease to initiate another round of infection in a naive host. In this review, we will provide a concise synopsis of the infectious life cycle of Mtb and its clinical and epidemiologic significance. We will also take stock of its virulence factors and pathogenic mechanisms that modulate host immunity and facilitate its spread. Developing a greater understanding of the interface between Mtb virulence factors and host defences will enable progress toward improved vaccines and therapeutics to prevent and treat tuberculosis.
Collapse
Affiliation(s)
- Kathryn C. Rahlwes
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Beatriz R.S. Dias
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Priscila C. Campos
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Samuel Alvarez-Arguedas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael U. Shiloh
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
8
|
Li LS, Yang L, Zhuang L, Ye ZY, Zhao WG, Gong WP. From immunology to artificial intelligence: revolutionizing latent tuberculosis infection diagnosis with machine learning. Mil Med Res 2023; 10:58. [PMID: 38017571 PMCID: PMC10685516 DOI: 10.1186/s40779-023-00490-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023] Open
Abstract
Latent tuberculosis infection (LTBI) has become a major source of active tuberculosis (ATB). Although the tuberculin skin test and interferon-gamma release assay can be used to diagnose LTBI, these methods can only differentiate infected individuals from healthy ones but cannot discriminate between LTBI and ATB. Thus, the diagnosis of LTBI faces many challenges, such as the lack of effective biomarkers from Mycobacterium tuberculosis (MTB) for distinguishing LTBI, the low diagnostic efficacy of biomarkers derived from the human host, and the absence of a gold standard to differentiate between LTBI and ATB. Sputum culture, as the gold standard for diagnosing tuberculosis, is time-consuming and cannot distinguish between ATB and LTBI. In this article, we review the pathogenesis of MTB and the immune mechanisms of the host in LTBI, including the innate and adaptive immune responses, multiple immune evasion mechanisms of MTB, and epigenetic regulation. Based on this knowledge, we summarize the current status and challenges in diagnosing LTBI and present the application of machine learning (ML) in LTBI diagnosis, as well as the advantages and limitations of ML in this context. Finally, we discuss the future development directions of ML applied to LTBI diagnosis.
Collapse
Affiliation(s)
- Lin-Sheng Li
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, 100091, China
- Hebei North University, Zhangjiakou, 075000, Hebei, China
- Senior Department of Respiratory and Critical Care Medicine, the Eighth Medical Center of PLA General Hospital, Beijing, 100091, China
| | - Ling Yang
- Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Li Zhuang
- Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Zhao-Yang Ye
- Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Wei-Guo Zhao
- Senior Department of Respiratory and Critical Care Medicine, the Eighth Medical Center of PLA General Hospital, Beijing, 100091, China.
| | - Wen-Ping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, 100091, China.
| |
Collapse
|
9
|
Muhammad N, Khan MT, Ali S, Khan TA, Khan AS, Ullah N, Higazi H, Ali S, Mohamed S, Qasim M. Novel Mutations in MPT64 Secretory Protein of Mycobacterium tuberculosis Complex. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2530. [PMID: 36767896 PMCID: PMC9915896 DOI: 10.3390/ijerph20032530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Tuberculosis (TB) is a global health problem caused by the Mycobacterium tuberculosis complex (MTBC). These bacteria secrete various proteins involved in the pathogenesis and persistence of MTBC. Among the secretory proteins, MPT64 (Rv1980C) is highly conserved and is also known as a major culture filtrate that is used in rapid diagnosis of MTBC. In the current study, we aimed to find the mutation in this highly conserved protein in isolates from the Pashtun-dominant province of Pakistan. We analyzed 470 M. tuberculosis whole-genome sequences of Khyber Pakhtunkhwa Province. Mutations in the MPT64 gene were screened through TB-Profiler and BioEdit software tools. The DynaMut web server was used to analyze the impact of the mutation on protein dynamics and stability. Among 470 MTB genomes, three non-synonymous mutations were detected in nine isolates, and one synonymous mutation (G208A) was found in four isolates. Mutation G211T (F159L), which was detected at the C-terminal domain of the protein in six isolates, was the most prominent. The second novel mutation, T480C (I70V), was detected in two isolates at the C-terminal side of the protein structure. The third novel mutation, A491C (L66R), was detected in a single isolate at the N-terminal side of the MPT64 protein. The effect of these three mutations was destabilizing on the protein structure. The molecular flexibility of the first two mutations increased, and the last one decreased. MPT64 is a highly conserved secretory protein, harboring only a few mutations. This study provides useful information for better managing the diagnosis of MTB isolates in high TB-burden countries.
Collapse
Affiliation(s)
- Noor Muhammad
- Department of Microbiology, Kohat University of Science and Technology, Kohat 26000, Pakistan
| | - Muhammad Tahir Khan
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang 473006, China
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, KM Defense Road, Lahore 54770, Pakistan
| | - Sajid Ali
- Department of Microbiology & Biotechnology, Bacha Khan University, Charsadda 24550, Pakistan
| | - Taj Ali Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25100, Pakistan
| | - Anwar Sheed Khan
- Department of Microbiology, Kohat University of Science and Technology, Kohat 26000, Pakistan
| | - Nadeem Ullah
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden
| | - Hassan Higazi
- Department of Medical Laboratory Sciences, College of Health Sciences, Gulf Medical University, Ajman P.O. Box 4184, United Arab Emirates
| | - Sara Ali
- Department of Medical Laboratory Sciences, College of Health Sciences, Gulf Medical University, Ajman P.O. Box 4184, United Arab Emirates
| | - Salma Mohamed
- Department of Medical Laboratory Sciences, College of Health Sciences, Gulf Medical University, Ajman P.O. Box 4184, United Arab Emirates
| | - Muhammad Qasim
- Department of Microbiology, Kohat University of Science and Technology, Kohat 26000, Pakistan
| |
Collapse
|
10
|
Nisa A, Kipper FC, Panigrahy D, Tiwari S, Kupz A, Subbian S. Different modalities of host cell death and their impact on Mycobacterium tuberculosis infection. Am J Physiol Cell Physiol 2022; 323:C1444-C1474. [PMID: 36189975 PMCID: PMC9662802 DOI: 10.1152/ajpcell.00246.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 11/22/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is the pathogen that causes tuberculosis (TB), a leading infectious disease of humans worldwide. One of the main histopathological hallmarks of TB is the formation of granulomas comprised of elaborately organized aggregates of immune cells containing the pathogen. Dissemination of Mtb from infected cells in the granulomas due to host and mycobacterial factors induces multiple cell death modalities in infected cells. Based on molecular mechanism, morphological characteristics, and signal dependency, there are two main categories of cell death: programmed and nonprogrammed. Programmed cell death (PCD), such as apoptosis and autophagy, is associated with a protective response to Mtb by keeping the bacteria encased within dead macrophages that can be readily phagocytosed by arriving in uninfected or neighboring cells. In contrast, non-PCD necrotic cell death favors the pathogen, resulting in bacterial release into the extracellular environment. Multiple types of cell death in the PCD category, including pyroptosis, necroptosis, ferroptosis, ETosis, parthanatos, and PANoptosis, may be involved in Mtb infection. Since PCD pathways are essential for host immunity to Mtb, therapeutic compounds targeting cell death signaling pathways have been experimentally tested for TB treatment. This review summarizes different modalities of Mtb-mediated host cell deaths, the molecular mechanisms underpinning host cell death during Mtb infection, and its potential implications for host immunity. In addition, targeting host cell death pathways as potential therapeutic and preventive approaches against Mtb infection is also discussed.
Collapse
Affiliation(s)
- Annuurun Nisa
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Franciele C Kipper
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Sangeeta Tiwari
- Department of Biological Sciences, Border Biomedical Research Center (BBRC), University of Texas, El Paso, Texas
| | - Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Townsville, Queensland, Australia
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
11
|
Singh S, Maurya SK, Aqdas M, Bashir H, Arora A, Bhalla V, Agrewala JN. Mycobacterium tuberculosis exploits MPT64 to generate myeloid-derived suppressor cells to evade the immune system. Cell Mol Life Sci 2022; 79:567. [PMID: 36283989 PMCID: PMC11803053 DOI: 10.1007/s00018-022-04596-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/19/2022] [Accepted: 10/09/2022] [Indexed: 11/24/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is a smart and successful pathogen since it can persist in the intimidating environment of the host by taming and tuning the immune system. Mtb releases MPT64 (Rv1980c) protein in high amounts in patients with active tuberculosis (TB). Consequently, we were curious to decipher the role of MPT64 on the differentiating dendritic cells (DCs) and its relation to evading the immune system. We observed that pre-exposure of differentiating DCs to MPT64 (DCMPT64) transformed them into a phenotype of myeloid-derived suppressor cells (MDSCs). DCMPT64 expressed a high level of immunosuppressive molecules PD-L1, TIM-3, nitric oxide (NO), arginase 1, IDO-1, IL-10 and TGF-β, but inhibited the production of pro-inflammatory cytokines TNF-α, IL-6 and IL-12. DCMPT64 chemotaxis function was diminished due to the reduced expression of CCR7. DCMPT64 promoted the generation of regulatory T cells (Tregs) but inhibited the differentiation of Th1 cells and Th17 cells. Further, high lipid and methylglyoxal content, and reduced glucose consumption by DCMPT64, rendered them metabolically quiescent and consequently, reduced DCMPT64 ability to phagocytose Mtb and provided a safer shelter for the intracellular survival of the mycobacterium. The mechanism identified in impairing the function of DCMPT64 was through the increased production and accumulation of methylglyoxal. Hence, for the first time, we demonstrate the novel role of MPT64 in promoting the generation of MDSCs to favor Mtb survival and escape its destruction by the immune system.
Collapse
Affiliation(s)
- Sanpreet Singh
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Sudeep K Maurya
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Mohammad Aqdas
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Hilal Bashir
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Ashish Arora
- Department of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Vijayender Bhalla
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
- Biosensor Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, 160036, India.
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, India.
| |
Collapse
|
12
|
Li W, Deng W, Zhang N, Peng H, Xu Y. Mycobacterium tuberculosis Rv2387 Facilitates Mycobacterial Survival by Silencing TLR2/p38/JNK Signaling. Pathogens 2022; 11:pathogens11090981. [PMID: 36145413 PMCID: PMC9504853 DOI: 10.3390/pathogens11090981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/15/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) can evade antimicrobial immunity and persist within macrophages by interfering with multiple host cellular functions through its virulence factors, causing latent tuberculosis. The Rv2387 protein has been identified as a putative effector that potentially participates in Mtb pathogenicity. To explore the role of the Rv2387 protein in host–mycobacteria interactions, we established recombinant M. smegmatis strains and RAW264.7 cell lines that stably express the Rv2387 protein. We found that this protein suppresses mycobacteria infection-induced macrophage apoptosis by inactivating caspase-3/-8, thus facilitating the intracellular survival of mycobacteria. In addition, Rv2387 inhibits the production of inflammatory cytokines in macrophages by specifically suppressing TLR2-dependent stimulation of p38 and JNK MAPK pathways. Moreover, we further determined that the Rv2387 protein conferred a growth advantage over recombinant M. smegmatis and suppressed the inflammatory response in a mouse infection model. Overall, these data suggested that Rv2387 facilitates mycobacteria to escape host immunity and might be an essential virulence factor in Mtb.
Collapse
Affiliation(s)
- Wu Li
- The Joint Center for Infection and Immunity, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Regional Characteristic Agricultural Resources, College of Life Sciences, Neijiang Normal University, Neijiang 641100, China
- Correspondence: (W.L.); (Y.X.)
| | - Wanyan Deng
- The Joint Center for Infection and Immunity, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nan Zhang
- Key Laboratory of Regional Characteristic Agricultural Resources, College of Life Sciences, Neijiang Normal University, Neijiang 641100, China
| | - Huijuan Peng
- Key Laboratory of Regional Characteristic Agricultural Resources, College of Life Sciences, Neijiang Normal University, Neijiang 641100, China
| | - Yi Xu
- The Joint Center for Infection and Immunity, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- Correspondence: (W.L.); (Y.X.)
| |
Collapse
|
13
|
Qiu X, Tang H, Dong J, Wang C, Li Y. Stochastic Collision Electrochemistry from Single Pt Nanoparticles: Electrocatalytic Amplification and MicroRNA Sensing. Anal Chem 2022; 94:8202-8208. [PMID: 35642339 DOI: 10.1021/acs.analchem.2c00116] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Single-particle collisions have made many achievements in basic research, but challenges still exist due to their low collision frequency and selectivity in complex samples. In this work, we developed an "on-off-on" strategy based on Pt nanoparticles (PtNPs) that catalyze N2H4 collision signals on the surface of carbon ultramicroelectrodes and established a new method for the detection of miRNA21 with high selectivity and sensitivity. PtNPs catalyze the reduction of N2H4 on the surface of carbon ultramicroelectrodes to generate a stepped collision signal, which is in the "on" state. The single-stranded DNA paired with miRNA21 is coupled with PtNPs to form the complex DNA/PtNPs. Because PtNPs are covered by DNA, the electrocatalytic collision of N2H4 oxidation is inhibited. At this time, the signal is in the "off" state. When miRNA21 is added, the strong complementary pairing between miRNA21 and DNA destroys the electrostatic adsorption of DNA/PtNP conjugates and restores the electrocatalytic performance of PtNPs, and the signal is in the "on" state again. Based on this, a new method for detecting miRNA21 was established. It provides a new way for small-molecule sensing and has a wide range of applications in electroanalysis, electrocatalysis, and biosensing.
Collapse
Affiliation(s)
- Xia Qiu
- Anhui Key Laboratory of Chemo/Biosensing, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241000, P.R. China
| | - Haoran Tang
- Anhui Key Laboratory of Chemo/Biosensing, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241000, P.R. China
| | - Jingyi Dong
- Anhui Key Laboratory of Chemo/Biosensing, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241000, P.R. China
| | - Chaohui Wang
- Anhui Key Laboratory of Chemo/Biosensing, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241000, P.R. China
| | - Yongxin Li
- Anhui Key Laboratory of Chemo/Biosensing, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241000, P.R. China
| |
Collapse
|
14
|
Banerjee N, Wang H, Wang G, Boor PJ, Khan MF. Differential Expression of miRNAs in Trichloroethene-Mediated Inflammatory/Autoimmune Response and Its Modulation by Sulforaphane: Delineating the Role of miRNA-21 and miRNA-690. Front Immunol 2022; 13:868539. [PMID: 35422807 PMCID: PMC9001960 DOI: 10.3389/fimmu.2022.868539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Trichloroethene (TCE), an occupational and ubiquitous environmental contaminant, is associated with the induction of autoimmune diseases (ADs). Although oxidative stress plays a major role in TCE-mediated autoimmunity, the underlying molecular mechanisms still need to be delineated. Altered non-coding RNAs, including the expression of microRNAs (miRNAs), can influence target genes, especially related to apoptosis and inflammation, and contribute to ADs. Therefore, the objective of this study was to delineate the contribution of miRNAs in TCE-mediated inflammatory and autoimmune response. To achieve this, we treated female MRL+/+ mice with TCE (10 mmol/kg in corn oil, i.p., every fourth day) with/without antioxidant sulforaphane (SFN; 8 mg/kg in corn oil, i.p., every other day) for 6 weeks. With the use of miRNA microarray, 293 miRNAs were analyzed, which included 35 miRNAs that were relevant to inflammation and ADs. Among those 35 miRNAs, 8 were modulated by TCE and/or TCE+SFN exposure. TCE treatment led to increased expression of 3 miRNAs and also decreased expression of 3 miRNAs. Interestingly, among the 35 differentially expressed miRNAs, antioxidant SFN modulated the expression of 6 miRNAs. Based on the microarray findings, we subsequently focused on two miRNAs (miRNA-21 and miRNA-690), which are known to be involved in inflammation and autoimmune response. The increases in miRNA-21 and miR-690 (observed using miRNA microarray) were further validated by RT-PCR, and the TCE-mediated increases in miR-21 and miR-690 were ameliorated by SFN treatment. Modulating miR-21 and miR-690 by respective inhibitors or mimics suppressed the expression of NF-κB (p65) and IL-12 in RAW 264.7 cells. Our findings suggest a contributory role of miR-21 and miR-690 in TCE-mediated and its metabolite dichloroacetyl chloride (DCAC)-mediated inflammation and autoimmune response and support that antioxidant SFN could be a potential therapeutic candidate for inflammatory responses and ADs.
Collapse
Affiliation(s)
- Nivedita Banerjee
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Hui Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Gangduo Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Paul J Boor
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - M Firoze Khan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
15
|
Magdalena D, Magdalena G. Biological functions and diagnostic implications of microRNAs in Mycobacterium tuberculosis infection. Asian Pac J Trop Biomed 2022. [DOI: 10.4103/2221-1691.333208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
16
|
Kusuma SAF, Subroto T, Parwati I, Rukayadi Y, Fadhlillah M, Pardede RM, Berlian AV, Sabila G. Improving of pelB-Secreted MPT64 protein released by Escherichia coli BL21 (DE3) using Triton X-100 and Tween-80. J Adv Pharm Technol Res 2022; 13:171-176. [PMID: 35935695 PMCID: PMC9355052 DOI: 10.4103/japtr.japtr_25_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
pelB has been known as a successful signal peptide to translocate the protein target extracellularly in the Escherichia coli system. However, in our previous study, the yield of MPT64 protein extracellular recovery was still low and plenty of this protein was remain trapped in cytoplasm and periplasm. Recently, nonionic surfactants were efficiently reported to secrete recombinant protein extracellularly. Nonetheless, it must be clarified whether the surfactant supplementation can improve the yield of MPT64 extracellular protein significantly without giving impact on the structure of isolated MPT64 protein and can minimized the cell lysis effect. MPT64 protein secretion was carried out by comparing the effects of surfactants Tween 80 and Triton × 100 at various concentrations. Triton × 100 was able to increase the extracellular MPT64 protein gain up to 3 times higher than Tween 80 and it was in line with the greater level ratio of cell leakage of Triton × 100 compared to that of Tween 80. Similarly, the viable cell of the cultures decreased dramatically. However, both surfactants did not interfere the structure of MPT64 protein. In conclusion, Triton × 100 can be chosen as the supporting surfactant to assist the act of peptide signal in improving the resulting of MPT64 extracellular protein.
Collapse
Affiliation(s)
- Sri Agung Fitri Kusuma
- Department of Biology Pharmacy, Faculty of Pharmacy, Padjadjaran University, Bandung, West Java, Indonesia,Address for correspondence: Dr. Sri Agung Fitri Kusuma, Department of Biology Pharmacy, Faculty of Pharmacy, Padjadjaran University, Indonesia. E-mail:
| | - Toto Subroto
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Padjadjaran University,Research Center of Molecular Biotechnology and Bioinformatics, Padjadjaran University, Bandung, West Java, Indonesia
| | - Ida Parwati
- Department of Clinical Pathology, Faculty of Medical, Padjadjaran University, Bandung, West Java,Department of Clinical Pathology, Dr. Hasan Sadikin General Hospital, Bandung, Indonesia
| | - Yaya Rukayadi
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Sedang, Malaysia
| | - Muhammad Fadhlillah
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Padjadjaran University,Research Center of Molecular Biotechnology and Bioinformatics, Padjadjaran University, Bandung, West Java, Indonesia
| | - Ruth Michellee Pardede
- Department of Biology Pharmacy, Faculty of Pharmacy, Padjadjaran University, Bandung, West Java, Indonesia
| | - Alif Virisy Berlian
- Department of Biology Pharmacy, Faculty of Pharmacy, Padjadjaran University, Bandung, West Java, Indonesia
| | - Gina Sabila
- Department of Biology Pharmacy, Faculty of Pharmacy, Padjadjaran University, Bandung, West Java, Indonesia
| |
Collapse
|
17
|
Hassan HM, Mahran YF, Ghanim AMH. Ganoderma lucidum ameliorates the diabetic nephropathy via down-regulatory effect on TGFβ-1 and TLR-4/NFκB signalling pathways. J Pharm Pharmacol 2021; 73:1250-1261. [PMID: 33847358 DOI: 10.1093/jpp/rgab058] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 03/18/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Diabetic nephropathy (DN) is one of the most important complications of diabetes mellitus and it is considered as a principal cause for end-stage renal failure. Ganoderma lucidum (GL) has been studied for its reno-protective effect against different kidney injury models. The aim of our study is to investigate the mechanisms by which GL can improve kidney injury and consequent renal inflammation and fibrosis. METHODS GL either in a low dose (250 mg/kg, i.p.) or high dose (500 mg/kg, i.p.) was administered to DN rat model, and nephropathy indices were investigated. KEY FINDINGS GL treatment significantly down-regulated kidney injury molecule-1 (KIM-1) gene expression and inhibited TLR-4 (Toll-like receptor-4)/NFκB (nuclear factor kappa B) signalling pathway. As well, GL treatment significantly decreased the pro-inflammatory mediator; IL-1β (interleukin-1 beta) level and fibrosis-associated growth factors; FGF-23 (fibroblast growth factor-23) and TGFβ-1 (transforming growth factor beta-1) levels. In addition, GL remarkably inhibited (Bax) the pro-apoptotic protein and induced (Bcl-2) the anti-apoptotic protein expression in kidneys. Moreover, GL treatment significantly alleviates kidney injury indicated by correcting the deteriorated kidney function and improving oxidative stress status in DN rats. CONCLUSIONS GL significantly improved renal function indices through dose-dependent kidney function restoration, oxidative stress reduction, down-regulation of gene expression of KIM-1 and TLR4/NFκB signalling pathway blockage with subsequent alleviation of renal inflammation and fibrosis.
Collapse
Affiliation(s)
- Hanan M Hassan
- Department of pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Mansoura, Egypt
| | - Yasmen F Mahran
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amal M H Ghanim
- Department of Biochemistry, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| |
Collapse
|
18
|
Ordonez AA, Abhishek S, Singh AK, Klunk MH, Azad BB, Aboagye EO, Carroll L, Jain SK. Caspase-Based PET for Evaluating Pro-Apoptotic Treatments in a Tuberculosis Mouse Model. Mol Imaging Biol 2021; 22:1489-1494. [PMID: 32232626 DOI: 10.1007/s11307-020-01494-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE Despite recent advances in antimicrobial treatments, tuberculosis (TB) remains a major global health threat. Mycobacterium tuberculosis proliferates in macrophages, preventing apoptosis by inducing anti-apoptotic proteins leading to necrosis of the infected cells. Necrosis then leads to increased tissue destruction, reducing the penetration of antimicrobials and immune cells to the areas where they are needed most. Pro-apoptotic drugs could be used as host-directed therapies in TB to improve antimicrobial treatments and patient outcomes. PROCEDURE We evaluated [18F]-ICMT-11, a caspase-3/7-specific positron emission tomography (PET) radiotracer, in macrophage cell cultures and in an animal model of pulmonary TB that closely resembles human disease. RESULTS Cells infected with M. tuberculosis and treated with cisplatin accumulated [18F]-ICMT-11 at significantly higher levels compared with that of controls, which correlated with levels of caspase-3/7 activity. Infected mice treated with cisplatin with increased caspase-3/7 activity also had a higher [18F]-ICMT-11 PET signal compared with that of untreated infected animals. CONCLUSIONS [18F]-ICMT-11 PET could be used as a noninvasive approach to measure intralesional pro-apoptotic responses in situ in pulmonary TB models and support the development of pro-apoptotic host-directed therapies for TB.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sudhanshu Abhishek
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alok K Singh
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Mariah H Klunk
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Babak Benham Azad
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Department of Surgery & Cancer Hammersmith Campus, Imperial College, London, UK
| | - Laurence Carroll
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Sampath P, Periyasamy KM, Ranganathan UD, Bethunaickan R. Monocyte and Macrophage miRNA: Potent Biomarker and Target for Host-Directed Therapy for Tuberculosis. Front Immunol 2021; 12:667206. [PMID: 34248945 PMCID: PMC8267585 DOI: 10.3389/fimmu.2021.667206] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
The end TB strategy reinforces the essentiality of readily accessible biomarkers for early tuberculosis diagnosis. Exploration of microRNA (miRNA) and pathway analysis opens an avenue for the discovery of possible therapeutic targets. miRNA is a small, non-coding oligonucleotide characterized by the mechanism of gene regulation, transcription, and immunomodulation. Studies on miRNA define their importance as an immune marker for active disease progression and as an immunomodulator for innate mechanisms, such as apoptosis and autophagy. Monocyte research is highly advancing toward TB pathogenesis and biomarker efficiency because of its innate and adaptive response connectivity. The combination of monocytes/macrophages and their relative miRNA expression furnish newer insight on the unresolved mechanism for Mycobacterium survival, exploitation of host defense, latent infection, and disease resistance. This review deals with miRNA from monocytes, their relative expression in different disease stages of TB, multiple gene regulating mechanisms in shaping immunity against tuberculosis, and their functionality as biomarker and host-mediated therapeutics. Future collaborative efforts involving multidisciplinary approach in various ethnic population with multiple factors (age, gender, mycobacterial strain, disease stage, other chronic lung infections, and inflammatory disease criteria) on these short miRNAs from body fluids and cells could predict the valuable miRNA biosignature network as a potent tool for biomarkers and host-directed therapy.
Collapse
Affiliation(s)
- Pavithra Sampath
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | | | - Uma Devi Ranganathan
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai, India
| | | |
Collapse
|
20
|
Curcumin suppresses oxidative stress via regulation of ROS/NF-κB signaling pathway to protect retinal vascular endothelial cell in diabetic retinopathy. Mol Cell Toxicol 2021. [DOI: 10.1007/s13273-021-00144-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
21
|
Kim H, Lee K, Kim JM, Kim MY, Kim JR, Lee HW, Chung YW, Shin HI, Kim T, Park ES, Rho J, Lee SH, Kim N, Lee SY, Choi Y, Jeong D. Selenoprotein W ensures physiological bone remodeling by preventing hyperactivity of osteoclasts. Nat Commun 2021; 12:2258. [PMID: 33859201 PMCID: PMC8050258 DOI: 10.1038/s41467-021-22565-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/18/2021] [Indexed: 11/09/2022] Open
Abstract
Selenoproteins containing selenium in the form of selenocysteine are critical for bone remodeling. However, their underlying mechanism of action is not fully understood. Herein, we report the identification of selenoprotein W (SELENOW) through large-scale mRNA profiling of receptor activator of nuclear factor (NF)-κΒ ligand (RANKL)-induced osteoclast differentiation, as a protein that is downregulated via RANKL/RANK/tumour necrosis factor receptor-associated factor 6/p38 signaling. RNA-sequencing analysis revealed that SELENOW regulates osteoclastogenic genes. SELENOW overexpression enhances osteoclastogenesis in vitro via nuclear translocation of NF-κB and nuclear factor of activated T-cells cytoplasmic 1 mediated by 14-3-3γ, whereas its deficiency suppresses osteoclast formation. SELENOW-deficient and SELENOW-overexpressing mice exhibit high bone mass phenotype and osteoporosis, respectively. Ectopic SELENOW expression stimulates cell-cell fusion critical for osteoclast maturation as well as bone resorption. Thus, RANKL-dependent repression of SELENOW regulates osteoclast differentiation and blocks osteoporosis caused by overactive osteoclasts. These findings demonstrate a biological link between selenium and bone metabolism. Selenoproteins containing selenium have a variety of physiological functions including redox homeostasis and thyroid hormone metabolism. Here, the authors show that RANKL-dependent repression of selenoprotein W regulates cell fusion during osteoclast differentiation and bone remodelling in mice.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, Korea.,Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Kyunghee Lee
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, Korea
| | - Jin Man Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, Korea
| | - Mi Yeong Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, Korea
| | - Jae-Ryong Kim
- Department of Biochemistry and Molecular Biology, Smart-aging Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Youn Wook Chung
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hong-In Shin
- IHBR, Department of Oral Pathology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Taesoo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Eui-Soon Park
- Department of Microbiology and BK21 Bio Brain Center, Chungnam National University, Daejeon, Korea
| | - Jaerang Rho
- Department of Microbiology and BK21 Bio Brain Center, Chungnam National University, Daejeon, Korea
| | - Seoung Hoon Lee
- Department of Oral Microbiology and Immunology, Wonkwang University School of Dentistry, Iksan, Korea
| | - Nacksung Kim
- National Research Laboratory for Regulation of Bone Metabolism and Disease, Chonnam National University Medical School, Gwangju, Korea
| | - Soo Young Lee
- Division of Life and Pharmaceutical Sciences, Department of Life Science, Center for Cell Signaling & Drug Discovery Research, College of Natural Sciences, Ewha Womans University, Seoul, Korea
| | - Yongwon Choi
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Daewon Jeong
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu, Korea.
| |
Collapse
|
22
|
Riahi Rad Z, Riahi Rad Z, Goudarzi H, Goudarzi M, Mahmoudi M, Yasbolaghi Sharahi J, Hashemi A. MicroRNAs in the interaction between host-bacterial pathogens: A new perspective. J Cell Physiol 2021; 236:6249-6270. [PMID: 33599300 DOI: 10.1002/jcp.30333] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022]
Abstract
Gene expression regulation plays a critical role in host-pathogen interactions, and RNAs function is essential in this process. miRNAs are small noncoding, endogenous RNA fragments that affect stability and/or translation of mRNAs, act as major posttranscriptional regulators of gene expression. miRNA is involved in regulating many biological or pathological processes through targeting specific mRNAs, including development, differentiation, apoptosis, cell cycle, cytoskeleton organization, and autophagy. Deregulated microRNA expression is associated with many types of diseases, including cancers, immune disturbances, and infection. miRNAs are a vital section of the host immune response to bacterial-made infection. Bacterial pathogens suppress host miRNA expression for their benefit, promoting survival, replication, and persistence. The role played through miRNAs in interaction with host-bacterial pathogen has been extensively studied in the past 10 years, and knowledge about these staggering molecules' function can clarify the complicated and ambiguous interactions of the host-bacterial pathogen. Here, we review how pathogens prevent the host miRNA expression. We briefly discuss emerging themes in this field, including their role as biomarkers in identifying bacterial infections, as part of the gut microbiota, on host miRNA expression.
Collapse
Affiliation(s)
- Zohreh Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Yasbolaghi Sharahi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Quarleri J, Cevallos C, Delpino MV. Apoptosis in infectious diseases as a mechanism of immune evasion and survival. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 125:1-24. [PMID: 33931136 DOI: 10.1016/bs.apcsb.2021.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In pluricellular organisms, apoptosis is indispensable for the development and homeostasis. During infection, apoptosis plays the main role in the elimination of infected cells. Infectious diseases control apoptosis, and this contributes to disease pathogenesis. Increased apoptosis may participate in two different ways. It can assist the dissemination of intracellular pathogens or induce immunosuppression to favor pathogen dissemination. In other conditions, apoptosis can benefit eradicate infectious agents from the host. Accordingly, bacteria, viruses, fungi, and parasites have developed strategies to inhibit host cell death by apoptosis to allow intracellular survival and persistence of the pathogen. The clarification of the intracellular signaling pathways, the receptors involved and the pathogen factors that interfere with apoptosis could disclose new therapeutic targets for blocking microbial actions on apoptotic pathways. In this review, we summarize the current knowledge on pathogen anti-apoptotic and apoptotic approaches and the mechanisms involving in disease.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Cintia Cevallos
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - María Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
24
|
Huang YL, Zhang FL, Tang XL, Yang XJ. Telocytes Enhances M1 Differentiation and Phagocytosis While Inhibits Mitochondria-Mediated Apoptosis Via Activation of NF-κB in Macrophages. Cell Transplant 2021; 30:9636897211002762. [PMID: 33787355 PMCID: PMC8020100 DOI: 10.1177/09636897211002762] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/17/2022] Open
Abstract
Telocytes (TCs), which are a recently discovered interstitial cell type present in various organs and tissues, perform multiple biological functions and participate in extensive crosstalk with neighboring cells. Endometriosis (EMs) is a gynecological disease characterized by the presence of viable endometrial debris and impaired macrophage phagocytosis in the peritoneal environment. Here, CD34/vimentin-positive TCs were co-cultured with RAW264.7 cells in vitro. M1/M2 differentiation-related markers were detected; phagocytosis, energy metabolism, proliferation, apoptosis, and pathway mechanisms were studied; and the mitochondrial membrane potential (ΔΨm) was measured. Furthermore, in an EMs mouse model, the differentiation of macrophages in response to treatment with TC-conditioned medium (TCM) in vivo was studied. The results showed that upon in vitro co-culture with TCM, RAW264.7 cells differentiated more toward the M1 phenotype with enhancement of phagocytosis, increase in energy metabolism and proliferation owing to reduced the loss of ΔΨm, and suppression of dexamethasone-induced apoptosis. Further, along with the activation of NF-κB, Bcl-2 and Bcl-xl, the expression of Bax, cleaved-caspase9, and cleaved-caspase3 reduced in RAW264.7 cells. In addition, the M1 subtype was found to be the dominant phenotype among tissue and peritoneal macrophages in the EMs model subjected to in vivo TCM treatment. In conclusion, TCs enhanced M1 differentiation and phagocytosis while inhibiting apoptosis via the activation of NF-κB in macrophages, which potentially inhibited the onset of EMs. Our findings provide a potential research target and the scope for developing a promising therapeutic strategy for EMs.
Collapse
Affiliation(s)
- Yue-Lin Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou city, Jiangsu Province, PR China
| | - Fei-Lei Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou city, Jiangsu Province, PR China
| | - Xue-Ling Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou city, Jiangsu Province, PR China
| | - Xiao-Jun Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou city, Jiangsu Province, PR China
| |
Collapse
|
25
|
Dahiya B, Prasad T, Singh V, Khan A, Kamra E, Mor P, Yadav A, Gupta KB, Mehta PK. Diagnosis of tuberculosis by nanoparticle-based immuno-PCR assay based on mycobacterial MPT64 and CFP-10 detection. Nanomedicine (Lond) 2020; 15:2609-2624. [PMID: 33090059 DOI: 10.2217/nnm-2020-0258] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To improve the diagnostic accuracy of immuno-PCR (I-PCR) in tuberculosis (TB) patients by using functionalized gold nanoparticles (AuNPs) coupled with detection antibodies and oligonucleotides, and magnetic beads (MBs) conjugated with capture antibodies in the liquid phase. Materials & methods: MB-coupled AuNP-based I-PCR (MB-AuNP-I-PCR) assay was designed to detect a cocktail of Mycobacterium tuberculosis MPT64 and CFP-10 proteins in bodily fluids of TB patients. Results: The sensitivities of 89.3 (n = 94) and 78.1% (n = 73) were observed in pulmonary TB and extrapulmonary TB patients, respectively, with specificities of 97.9-98.3%. Notably, the sensitivities attained by MB-AuNP-I-PCR in smear-negative pulmonary TB and extrapulmonary TB patients were significantly higher (p < 0.05-0.001) than Magneto-ELISA and GeneXpert assay. Conclusion: The improved technology, as well as enhanced diagnostic accuracy of MB-AuNP-I-PCR, may lead to development of an attractive diagnostic kit.
Collapse
Affiliation(s)
- Bhawna Dahiya
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak 124001, Haryana, India
| | - Tulika Prasad
- Special Centre for Nano Science & Advanced Instrumentation Research & Facility (AIRF), Jawaharlal Nehru University (JNU), New Delhi 110067, India
| | - Vishwajeet Singh
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Anish Khan
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak 124001, Haryana, India
| | - Ekta Kamra
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak 124001, Haryana, India
| | - Preeti Mor
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak 124001, Haryana, India
| | - Aparna Yadav
- Department of Microbiology, University of Health Sciences (UHS), Rohtak 124001, Haryana, India
| | - Krishna B Gupta
- Department of TB & Respiratory Medicine, UHS, Rohtak 124001, Haryana, India
| | - Promod K Mehta
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak 124001, Haryana, India
| |
Collapse
|
26
|
Wang WH, Takeuchi R, Jain SH, Jiang YH, Watanuki S, Ohtaki Y, Nakaishi K, Watabe S, Lu PL, Ito E. A novel, rapid (within hours) culture-free diagnostic method for detecting live Mycobacterium tuberculosis with high sensitivity. EBioMedicine 2020; 60:103007. [PMID: 32949995 PMCID: PMC7501073 DOI: 10.1016/j.ebiom.2020.103007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Nucleic acid amplification tests (NAATs) are widely used to diagnose tuberculosis (TB), but cannot discriminate live bacilli from dead bacilli. Live bacilli can be isolated by culture methods, but this is time-consuming. We developed a de novo TB diagnostic method that detects only live bacilli with high sensitivity within hours. METHODS A prospective study was performed in Taiwan from 2017 to 2018. Sputum was collected consecutively from 1102 patients with suspected TB infection. The sputum was pretreated and heated at 46°C for 1 h to induce the secretion of MPT64 protein from live Mycobacterium tuberculosis. MPT64 was detected with our ultrasensitive enzyme-linked immunosorbent assay (ELISA) coupled with thionicotinamide-adenine dinucleotide (thio-NAD) cycling. We compared our data with those obtained using a culture test (MGIT), a smear test (Kinyoun staining), and a NAAT (Xpert). FINDINGS The limit of detection for MPT64 in our culture-free ultrasensitive ELISA was 2.0 × 10-19 moles/assay. When the criterion for a positive response was set as an absorbance value ≥17 mAbs, this value corresponded to ca. 330 CFU/mL in the culture method - almost the same high-detection sensitivity as the culture method. To confirm that MPT64 is secreted from only live bacilli, M. bovis BCG was killed using 8 μg/mL rifampicin and then heated. Following this procedure, our method detected no MPT64. Our rapid ultra-sensitive ELISA-based method required only 5 h to complete. Comparing the results of our method with those of culture tests for 944 specimens revealed a sensitivity of 86.9% (93/107, 95% CI: 79.0-92.7%) and a specificity of 92.0% (770/837, 95% CI: 89.9-93.7%). The performance data were not significantly different (McNemar's test, P = 0.887) from those of the Xpert tests. In addition, at a ≥1+ titer in the smear test, the positive predictive value of our culture-free ultrasensitive ELISA tests was in a good agreement with that of the culture tests. Furthermore, our culture-free ultrasensitive ELISA test had better validity for drug effectiveness examination than Xpert tests because our test detected only live bacilli. INTERPRETATION Our culture-free ultrasensitive ELISA method detects only live TB bacilli with high sensitivity within hours, allowing for rapid diagnosis of TB and monitoring drug efficacy. FUNDING Matching Planner Program from JST (VP29117939087), the A-STEP Program from JST (AS3015096U), Waseda University grants for Specific Research Projects (2017A-015 and 2019C-123), the Precise Measurement Technology Promotion Foundation to E.I.
Collapse
Affiliation(s)
- Wen-Hung Wang
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, 100 TzYou 1st Rd., Kaohsiung 80756, Taiwan
| | - Rikiya Takeuchi
- R&D Department, TAUNS Laboratories, Inc., 761-1 Kamishima, Izunokuni, Shizuoka 410-2325, Japan
| | - Shu-Huei Jain
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, 100 TzYou 1st Rd., Kaohsiung 80756, Taiwan
| | - Yong-Huang Jiang
- R&D Department, TAUNS Laboratories, Inc., 761-1 Kamishima, Izunokuni, Shizuoka 410-2325, Japan
| | - Sonoko Watanuki
- R&D Department, TAUNS Laboratories, Inc., 761-1 Kamishima, Izunokuni, Shizuoka 410-2325, Japan
| | - Yoshiharu Ohtaki
- R&D Department, TAUNS Laboratories, Inc., 761-1 Kamishima, Izunokuni, Shizuoka 410-2325, Japan
| | - Kazunari Nakaishi
- R&D Headquarters, TAUNS Laboratories, Inc., 761-1 Kamishima, Izunokuni, Shizuoka 410-2325, Japan; Waseda Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan
| | - Satoshi Watabe
- R&D Headquarters, TAUNS Laboratories, Inc., 761-1 Kamishima, Izunokuni, Shizuoka 410-2325, Japan; Waseda Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan
| | - Po-Liang Lu
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, 100 TzYou 1st Rd., Kaohsiung 80756, Taiwan; College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., Kaohsiung 80756, Taiwan.
| | - Etsuro Ito
- Waseda Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan; Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., Kaohsiung 80756, Taiwan; Department of Biology, Waseda University, 2-2 Wakamatsucho, Shinjuku, Tokyo 162-8480, Japan.
| |
Collapse
|
27
|
Yimer SA, Kalayou S, Homberset H, Birhanu AG, Riaz T, Zegeye ED, Lutter T, Abebe M, Holm-Hansen C, Aseffa A, Tønjum T. Lineage-Specific Proteomic Signatures in the Mycobacterium tuberculosis Complex Reveal Differential Abundance of Proteins Involved in Virulence, DNA Repair, CRISPR-Cas, Bioenergetics and Lipid Metabolism. Front Microbiol 2020; 11:550760. [PMID: 33072011 PMCID: PMC7536270 DOI: 10.3389/fmicb.2020.550760] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/17/2020] [Indexed: 01/17/2023] Open
Abstract
Despite the discovery of the tubercle bacillus more than 130 years ago, its physiology and the mechanisms of virulence are still not fully understood. A comprehensive analysis of the proteomes of members of the human-adapted Mycobacterium tuberculosis complex (MTBC) lineages 3, 4, 5, and 7 was conducted to better understand the evolution of virulence and other physiological characteristics. Unique and shared proteomic signatures in these modern, pre-modern and ancient MTBC lineages, as deduced from quantitative bioinformatics analyses of high-resolution mass spectrometry data, were delineated. The main proteomic findings were verified by using immunoblotting. In addition, analysis of multiple genome alignment of members of the same lineages was performed. Label-free peptide quantification of whole cells from MTBC lineages 3, 4, 5, and 7 yielded a total of 38,346 unique peptides derived from 3092 proteins, representing 77% coverage of the predicted proteome. MTBC lineage-specific differential expression was observed for 539 proteins. Lineage 7 exhibited a markedly reduced abundance of proteins involved in DNA repair, type VII ESX-3 and ESX-1 secretion systems, lipid metabolism and inorganic phosphate uptake, and an increased abundance of proteins involved in alternative pathways of the TCA cycle and the CRISPR-Cas system as compared to the other lineages. Lineages 3 and 4 exhibited a higher abundance of proteins involved in virulence, DNA repair, drug resistance and other metabolic pathways. The high throughput analysis of the MTBC proteome by super-resolution mass spectrometry provided an insight into the differential expression of proteins between MTBC lineages 3, 4, 5, and 7 that may explain the slow growth and reduced virulence, metabolic flexibility, and the ability to survive under adverse growth conditions of lineage 7.
Collapse
Affiliation(s)
- Solomon Abebe Yimer
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo, Oslo, Norway.,Coalition for Epidemic Preparedness Innovations, Oslo, Norway
| | - Shewit Kalayou
- Division of Laboratory Medicine, Department of Microbiology, Oslo University Hospital, Oslo, Norway.,International Centre of Insect Physiology and Ecology, Nairobi, Kenya
| | - Håvard Homberset
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo, Oslo, Norway
| | - Alemayehu Godana Birhanu
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo, Oslo, Norway.,Division of Laboratory Medicine, Department of Microbiology, Oslo University Hospital, Oslo, Norway.,Institute of Biotechnology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tahira Riaz
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo, Oslo, Norway
| | - Ephrem Debebe Zegeye
- NORCE Norwegian Research Centre AS, Centre for Applied Biotechnology, Bergen, Norway
| | - Timo Lutter
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo, Oslo, Norway
| | - Markos Abebe
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Carol Holm-Hansen
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Abraham Aseffa
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Tone Tønjum
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo, Oslo, Norway.,Division of Laboratory Medicine, Department of Microbiology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
28
|
Alipoor SD, Adcock IM, Tabarsi P, Folkerts G, Mortaz E. MiRNAs in tuberculosis: Their decisive role in the fate of TB. Eur J Pharmacol 2020; 886:173529. [PMID: 32919937 DOI: 10.1016/j.ejphar.2020.173529] [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] [Received: 06/04/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 12/20/2022]
Abstract
Tuberculosis (TB) is one of the most lethal global infectious diseases. Despite the availability of much higher levels of technology in health and medicine, tuberculosis still remains a serious global health problem. Mycobacterium tuberculosis has the capacity for prolonged survival inside macrophages by exploiting host metabolic and energy pathways and perturbing autophagy and apoptosis of infected cells. The mechanism(s) underlying this process are not completely understood but evidence suggests that mycobacteria subvert the host miRNA network to enable mycobacterial survival. We present here a comprehensive review on the role of miRNAs in TB immune escape mechanisms and the potential for miRNA-based TB therapeutics. Further validation studies are required to (i) elucidate the precise effect of TB on host miRNAs, (ii) determine the inhibition of mycobacterial burden using miRNA-based therapies and (iii) identify novel miRNA biomarkers that may prove useful in TB diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Shamila D Alipoor
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Payam Tabarsi
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gert Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Kavunja HW, Biegas KJ, Banahene N, Stewart JA, Piligian BF, Groenevelt JM, Sein CE, Morita YS, Niederweis M, Siegrist MS, Swarts BM. Photoactivatable Glycolipid Probes for Identifying Mycolate-Protein Interactions in Live Mycobacteria. J Am Chem Soc 2020; 142:7725-7731. [PMID: 32293873 PMCID: PMC7949286 DOI: 10.1021/jacs.0c01065] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mycobacteria have a distinctive glycolipid-rich outer membrane, the mycomembrane, which is a critical target for tuberculosis drug development. However, proteins that associate with the mycomembrane, or that are involved in its metabolism and host interactions, are not well-characterized. To facilitate the study of mycomembrane-related proteins, we developed photoactivatable trehalose monomycolate analogues that metabolically incorporate into the mycomembrane in live mycobacteria, enabling in vivo photo-cross-linking and click-chemistry-mediated analysis of mycolate-interacting proteins. When deployed in Mycobacterium smegmatis with quantitative proteomics, this strategy enriched over 100 proteins, including the mycomembrane porin (MspA), several proteins with known mycomembrane synthesis or remodeling functions (CmrA, MmpL3, Ag85, Tdmh), and numerous candidate mycolate-interacting proteins. Our approach is highly versatile, as it (i) enlists click chemistry for flexible protein functionalization; (ii) in principle can be applied to any mycobacterial species to identify endogenous bacterial proteins or host proteins that interact with mycolates; and (iii) can potentially be expanded to investigate protein interactions with other mycobacterial lipids. This tool is expected to help elucidate fundamental physiological and pathological processes related to the mycomembrane and may reveal novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Herbert W Kavunja
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan 48859, United States
| | - Kyle J Biegas
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan 48859, United States
| | - Nicholas Banahene
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan 48859, United States
| | - Jessica A Stewart
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan 48859, United States
| | - Brent F Piligian
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan 48859, United States
| | - Jessica M Groenevelt
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan 48859, United States
| | - Caralyn E Sein
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Yasu S Morita
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Michael Niederweis
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - M Sloan Siegrist
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Benjamin M Swarts
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan 48859, United States
| |
Collapse
|
30
|
Intelligent Mechanisms of Macrophage Apoptosis Subversion by Mycobacterium. Pathogens 2020; 9:pathogens9030218. [PMID: 32188164 PMCID: PMC7157668 DOI: 10.3390/pathogens9030218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/12/2020] [Accepted: 03/15/2020] [Indexed: 02/07/2023] Open
Abstract
Macrophages are one of the first innate defense barriers and play an indispensable role in communication between innate and adaptive immune responses, leading to restricted Mycobacterium tuberculosis (Mtb) infection. The macrophages can undergo programmed cell death (apoptosis), which is a crucial step to limit the intracellular growth of bacilli by liberating them into extracellular milieu in the form of apoptotic bodies. These bodies can be taken up by the macrophages for the further degradation of bacilli or by the dendritic cells, thereby leading to the activation of T lymphocytes. However, Mtb has the ability to interplay with complex signaling networks to subvert macrophage apoptosis. Here, we describe the intelligent strategies of Mtb inhibition of macrophages apoptosis. This review provides a platform for the future study of unrevealed Mtb anti-apoptotic mechanisms and the design of therapeutic interventions.
Collapse
|
31
|
Ruiz-Tagle C, Naves R, Balcells ME. Unraveling the Role of MicroRNAs in Mycobacterium tuberculosis Infection and Disease: Advances and Pitfalls. Infect Immun 2020; 88:e00649-19. [PMID: 31871103 PMCID: PMC7035921 DOI: 10.1128/iai.00649-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) is an infectious disease of extremely high epidemiological burden worldwide that is easily acquired through the inhalation of infected respiratory droplets. The complex pathogenesis of this infection spans from subjects never developing this disease despite intense exposure, to others in which immune containment fails catastrophically and severe or disseminated forms of disease ensue. In recent decades, microRNAs (miRNAs) have gained increasing attention due to their role as gene silencers and because of their altered expression in diverse human diseases, including some infections. Recent research regarding miRNAs and TB has revealed that the expression profile for particular miRNAs clearly changes upon Mycobacterium tuberculosis infection and also varies in the different stages of this disease. However, despite the growing number of studies-some of which have even proposed some miRNAs as potential biomarkers-methodological variations and key differences in relevant factors, such as sex and age, cell type analyzed, M. tuberculosis strain, and antimicrobial therapy status, strongly hinder the comparison of data. In this review, we summarize and discuss the literature and highlight the role of selected miRNAs that have specifically and more consistently been associated with M. tuberculosis infection, together with a discussion of the possible gene and immune regulation pathways involved.
Collapse
Affiliation(s)
- Cinthya Ruiz-Tagle
- Departamento de Enfermedades Infecciosas del Adulto, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Naves
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - María Elvira Balcells
- Departamento de Enfermedades Infecciosas del Adulto, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
32
|
Ali MK, Zhen G, Nzungize L, Stojkoska A, Duan X, Li C, Duan W, Xu J, Xie J. Mycobacterium tuberculosis PE31 ( Rv3477) Attenuates Host Cell Apoptosis and Promotes Recombinant M. smegmatis Intracellular Survival via Up-regulating GTPase Guanylate Binding Protein-1. Front Cell Infect Microbiol 2020; 10:40. [PMID: 32117813 PMCID: PMC7020884 DOI: 10.3389/fcimb.2020.00040] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
The Mycobacterium (M.) tuberculosis comprising proline–glutamic acid (PE) subfamily proteins associate with virulence, pathogenesis, and host-immune modulations. While the functions of most of this family members are not yet explored. Here, we explore the functions of “PE only” subfamily member PE31 (Rv3477) in virulence and host-pathogen interactions. We have expressed the M. tuberculosis PE31 in non-pathogenic Mycobacterium smegmatis strain (Ms_PE31) and demonstrated that PE31 significantly altered the cell facet features including colony morphology and biofilm formation. PE31 expressing M. smegmatis showed more resistant to the low pH, diamide, H2O2 and surface stress. Moreover, Ms_PE31 showed higher intracellular survival in macrophage THP-1 cells. Ms_PE31 significantly down-regulated the production of IL-12p40 and IL-6, while up-regulates the production of IL-10 in macrophages. Ms_PE31 also induced the expression of guanylate-binding protein-1 (GBP-1) in macrophages. Further analysis demonstrates that Ms_PE31 inhibits the caspase-3 activation and reduces the macrophages apoptosis. Besides, the NF-κB signaling pathway involves the interplay between Ms_PE31 and macrophages. Collectively, our finding identified that PE31 act as a functionally relevant virulence factor of M. tuberculosis.
Collapse
Affiliation(s)
- Md Kaisar Ali
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Gong Zhen
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Lambert Nzungize
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Andrea Stojkoska
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Xiangke Duan
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Chunyan Li
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Wei Duan
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Junqi Xu
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Jianping Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| |
Collapse
|
33
|
Luo L, Li X, Hu X, Hu C, Tang W, Deng S, Feng J. Anaphylatoxins Enhance Recruitment of Nonclassical Monocytes via Chemokines Produced by Pleural Mesothelial Cells in Tuberculous Pleural Effusion. Am J Respir Cell Mol Biol 2019; 60:454-464. [PMID: 30422670 DOI: 10.1165/rcmb.2018-0075oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
In the present study, we sought to elucidate the mechanisms by which monocytes migrate into the pleural space in the presence of anaphylatoxins in tuberculous pleural effusion (TPE). Monocytes in both pleural effusion and blood were counted, and their phenotypic characteristics were analyzed. Activation of the complement system was detected in TPE. The effects of Mpt64 and anaphylatoxins on the production of chemokines in pleural mesothelial cells (PMCs) were measured. The chemoattractant activity of chemokines produced by PMCs for monocytes was observed. Levels of CD14+CD16+ monocytes were significantly higher in TPE than in blood. Three pathways of the complement system were activated in TPE. C3a-C3aR1, C5a-C5aR1, CCL2-CCR2, CCL7-CCR2, and CX3CL1-CX3CR1 were coexpressed in PMCs and monocytes isolated from TPE. Moreover, we initially found that Mpt64 stimulated the expression of C3a and C5a in PMCs. C3a and C5a not only induced CCL2, CCL7, and CX3CL1 expression in PMCs but also stimulated production of IL-1β, IL-17, and IL-27 in monocytes. C3a and C5a stimulated PMCs to secrete CCL2, CCL7, and CX3CL1, which recruited CD14+CD16+ monocytes to the pleural cavity. As a result, the infiltration of CD14+CD16+ monocytes engaged in the pathogenesis of TPE by excessive production of inflammatory cytokines.
Collapse
Affiliation(s)
- Lisha Luo
- 1 Department of Respiratory and Critical Care Medicine, Key Site of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; and
| | - Xiaozhao Li
- 2 Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinyue Hu
- 1 Department of Respiratory and Critical Care Medicine, Key Site of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; and
| | - Chengping Hu
- 1 Department of Respiratory and Critical Care Medicine, Key Site of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; and
| | - Wei Tang
- 1 Department of Respiratory and Critical Care Medicine, Key Site of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; and
| | - Shuanglinzi Deng
- 1 Department of Respiratory and Critical Care Medicine, Key Site of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; and
| | - Juntao Feng
- 1 Department of Respiratory and Critical Care Medicine, Key Site of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; and
| |
Collapse
|
34
|
Ji Y, Shen J, Li M, Zhu X, Wang Y, Ding J, Jiang S, Chen L, Wei W. RMP/URI inhibits both intrinsic and extrinsic apoptosis through different signaling pathways. Int J Biol Sci 2019; 15:2692-2706. [PMID: 31754340 PMCID: PMC6854365 DOI: 10.7150/ijbs.36829] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/24/2019] [Indexed: 02/06/2023] Open
Abstract
The evading apoptosis of tumor cells may result in chemotherapy resistance. Therefore, investigating what molecular events contribute to drug-induced apoptosis, and how tumors evade apoptotic death, provides a paradigm to explain the relationship between cancer genetics and treatment sensitivity. In this study, we focused on the role of RMP/URI both in cisplatin-induced endogenous apoptosis and in TRAIL-induced exogenous apoptosis in HCC cells. Although flow cytometric analysis indicated that RMP overexpression reduced the apoptosis rate of HCC cells treated with both cisplatin and TRAIL, there was a difference in mechanism between the two treatments. Western blot showed that in intrinsic apoptosis induced by cisplatin, the overexpression of RMP promoted the Bcl-xl expression both in vitro and in vivo. Besides, RMP activated NF-κB/p65(rel) through the phosphorylation of ATM. However, in TRAIL-induced extrinsic apoptosis, RMP significantly suppressed the transcription and expression of P53. Moreover, the forced expression of P53 could offset this inhibitory effect. In conclusion, we presumed that RMP inhibited both intrinsic and extrinsic apoptosis through different signaling pathways. NF-κB was distinctively involved in the RMP circumvention of intrinsic apoptosis, but not in the extrinsic apoptosis of HCC cells. RMP might play an important role in defects of apoptosis, hence the chemotherapeutic resistance in hepatocellular carcinoma. These studies are promising to shed light on a more rational approach to clinical anticancer drug design and therapy.
Collapse
Affiliation(s)
- Yuan Ji
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| | - Jian Shen
- Department of Interventional Radiology, First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Min Li
- Department of Tumor, People Hospital of Maanshan, Maanshan, 243000, China
| | - Xiaoxiao Zhu
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| | - Yanyan Wang
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| | - Jiazheng Ding
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| | - Shunyao Jiang
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| | - Linqi Chen
- Department of Endocrinology, Children's Hospital affiliated to Soochow University, Suzhou, 215000, China
| | - Wenxiang Wei
- Department of Cell Biology, Institute of Bioengineering, School of Medicine, Soochow University, Suzhou 215123, China
| |
Collapse
|
35
|
Agarwal RG, Sharma P, Nyati KK. microRNAs in Mycobacterial Infection: Modulation of Host Immune Response and Apoptotic Pathways. Immune Netw 2019; 19:e30. [PMID: 31720041 PMCID: PMC6829074 DOI: 10.4110/in.2019.19.e30] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/19/2019] [Accepted: 08/29/2019] [Indexed: 01/02/2023] Open
Abstract
Our current knowledge of mycobacterial infections in humans has progressively increased over the past few decades. The infection of Mycobacterium tuberculosis causes tuberculosis (TB) disease, which has reasoned for excessive morbidity and mortality worldwide, and has become a foremost issue of health problem globally. Mycobacterium leprae, another member of the family Mycobacteriaceae, is responsible for causing a chronic disease known as leprosy that mainly affects mucosa of the upper respiratory tract, skin, peripheral nerves, and eyes. Ample amount of existing data suggests that pathogenic mycobacteria have skilled in utilizing different mechanisms to escape or offset the host immune responses. They hijack the machinery of immune cells through the modulation of microRNAs (miRs), which regulate gene expression and immune responses of the host. Evidence shows that miRs have now gained considerable attention in the research, owing to their involvement in a broad range of inflammatory processes that are further implicated in the pathogenesis of several diseases. However, the knowledge of functions of miRs during mycobacterial infections remains limited. This review summarises recent findings of differential expression of miRs, which are used to good advantage by mycobacteria in offsetting host immune responses generated against them.
Collapse
Affiliation(s)
- Riddhi Girdhar Agarwal
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur 342005, India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur 342005, India
| | - Kishan Kumar Nyati
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur 342005, India
| |
Collapse
|
36
|
Aguilar C, Mano M, Eulalio A. Multifaceted Roles of microRNAs in Host-Bacterial Pathogen Interaction. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0002-2019. [PMID: 31152522 PMCID: PMC11026079 DOI: 10.1128/microbiolspec.bai-0002-2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a well-characterized class of small noncoding RNAs that act as major posttranscriptional regulators of gene expression. Accordingly, miRNAs have been associated with a wide range of fundamental biological processes and implicated in human diseases. During the past decade, miRNAs have also been recognized for their role in the complex interplay between the host and bacterial pathogens, either as part of the host response to counteract infection or as a molecular strategy employed by bacteria to subvert host pathways for their own benefit. Importantly, the characterization of downstream miRNA targets and their underlying mechanisms of action has uncovered novel molecular factors and pathways relevant to infection. In this article, we review the current knowledge of the miRNA response to bacterial infection, focusing on different bacterial pathogens, including Salmonella enterica, Listeria monocytogenes, Mycobacterium spp., and Helicobacter pylori, among others.
Collapse
Affiliation(s)
- Carmen Aguilar
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- Functional Genomics and RNA-Based Therapeutics Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
- RNA & Infection Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
37
|
Naderer T, Fulcher MC. Targeting apoptosis pathways in infections. J Leukoc Biol 2019; 103:275-285. [PMID: 29372933 DOI: 10.1189/jlb.4mr0717-286r] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/29/2017] [Accepted: 09/13/2017] [Indexed: 11/24/2022] Open
Abstract
The programmed cell death pathway of apoptosis is essential for mammalian development and immunity as it eliminates unwanted and dangerous cells. As part of the cellular immune response, apoptosis removes the replicative niche of intracellular pathogens and enables the resolution of infections. To subvert apoptosis, pathogens have evolved a diverse range of mechanisms. In some circumstances, however, pathogens express effector molecules that induce apoptotic cell death. In this review, we focus on selected host-pathogen interactions that affect apoptotic pathways. We discuss how pathogens control the fate of host cells and how this determines the outcome of infections. Finally, small molecule inhibitors that activate apoptosis in cancer cells can also induce apoptotic cell death of infected cells. This suggests that targeting host death factors to kill infected cells is a potential therapeutic option to treat infectious diseases.
Collapse
Affiliation(s)
- Thomas Naderer
- Biomedicine Discovery Institute and Department of Biochemistry & Molecular Biology, Monash University, Clayton, Australia
| | - Maria Cecilia Fulcher
- Biomedicine Discovery Institute and Department of Biochemistry & Molecular Biology, Monash University, Clayton, Australia
| |
Collapse
|
38
|
Curto P, Riley SP, Simões I, Martinez JJ. Macrophages Infected by a Pathogen and a Non-pathogen Spotted Fever Group Rickettsia Reveal Differential Reprogramming Signatures Early in Infection. Front Cell Infect Microbiol 2019; 9:97. [PMID: 31024862 PMCID: PMC6467950 DOI: 10.3389/fcimb.2019.00097] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/22/2019] [Indexed: 12/22/2022] Open
Abstract
Despite their high degree of genomic similarity, different spotted fever group (SFG) Rickettsia are often associated with very different clinical presentations. For example, Rickettsia conorii causes Mediterranean spotted fever, a life-threatening disease for humans, whereas Rickettsia montanensis is associated with limited or no pathogenicity to humans. However, the molecular basis responsible for the different pathogenicity attributes are still not understood. Although killing microbes is a critical function of macrophages, the ability to survive and/or proliferate within phagocytic cells seems to be a phenotypic feature of several intracellular pathogens. We have previously shown that R. conorii and R. montanensis exhibit different intracellular fates within macrophage-like cells. By evaluating early macrophage responses upon insult with each of these rickettsial species, herein we demonstrate that infection with R. conorii results in a profound reprogramming of host gene expression profiles. Transcriptional programs generated upon infection with this pathogenic bacteria point toward a sophisticated ability to evade innate immune signals, by modulating the expression of several anti-inflammatory molecules. Moreover, R. conorii induce the expression of several pro-survival genes, which may result in the ability to prolong host cell survival, thus protecting its replicative niche. Remarkably, R. conorii-infection promoted a robust modulation of different transcription factors, suggesting that an early manipulation of the host gene expression machinery may be key to R. conorii proliferation in THP-1 macrophages. This work provides new insights into the early molecular processes hijacked by a pathogenic SFG Rickettsia to establish a replicative niche in macrophages, opening several avenues of research in host-rickettsiae interactions.
Collapse
Affiliation(s)
- Pedro Curto
- Ph.D. Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Sean P. Riley
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Isaura Simões
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Juan J. Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
39
|
Park HS, Back YW, Shin KW, Bae HS, Lee KI, Choi HG, Choi S, Lee HH, Choi CH, Park JK, Kim HJ. Mycobacterium tuberculosis Rv3463 induces mycobactericidal activity in macrophages by enhancing phagolysosomal fusion and exhibits therapeutic potential. Sci Rep 2019; 9:4246. [PMID: 30862819 PMCID: PMC6414722 DOI: 10.1038/s41598-019-38982-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/04/2019] [Indexed: 12/21/2022] Open
Abstract
Macrophages are responsible for innate and adaptive immune response activation necessary for eliminating infections. Optimal activation of macrophages to phagocytize Mycobacterium tuberculosis is critical in anti-mycobacterial defense. Here, we identified a novel Rv3463 hypothetical protein that induces macrophage activation in Mtb culture filtrate. Recombinant Rv3463 activated mouse bone marrow-derived macrophages to induce the expression of surface molecules and secrete pro-inflammatory cytokines via the TLR2 and TLR4 pathways. Mitogen activated protein kinase, phospatidylinositol-4,5-bisphosphate 3-kinases, and the NF-κB signaling pathways are involved in Rv3463-mediated macrophage activation. Furthermore, Rv3463 induced bactericidal effects in Mtb-infected macrophages through phagosome maturation and phagolysosomal fusion enhanced by phospatidylinositol-4,5-bisphosphate 3-kinases and Ca2+ signaling pathways and exhibited therapeutic effects in a short-term Mtb-infection mouse model. Overexpression of Rv3463 in M. smegmatis caused rapid clearance of bacteria in macrophages and mice. Our study suggests that Rv3463 is a promising target for the development of post-exposure tuberculosis vaccines or adjunct immune-therapy.
Collapse
Affiliation(s)
- Hye-Soo Park
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yong Woo Back
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ki-Won Shin
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun Shik Bae
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kang-In Lee
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Han-Gyu Choi
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seunga Choi
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hwang-Ho Lee
- Department of Microbiology and Immunology, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Chul Hee Choi
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong-Kyu Park
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hwa-Jung Kim
- Department of Microbiology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
40
|
Total Flavonoids from Carya cathayensis Sarg. Leaves Alleviate H9c2 Cells Hypoxia/Reoxygenation Injury via Effects on miR-21 Expression, PTEN/Akt, and the Bcl-2/Bax Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:8617314. [PMID: 30622615 PMCID: PMC6304542 DOI: 10.1155/2018/8617314] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/29/2018] [Accepted: 11/21/2018] [Indexed: 11/21/2022]
Abstract
This study aimed to investigate whether the total flavonoids (TFs) from Carya cathayensis Sarg. leaves alleviate hypoxia/reoxygenation (H/R) injury in H9c2 cardiomyocytes and to explore potential mechanisms. H9c2 cells pretreated with TFs for 24h were exposed to H/R treatment. The results indicated that TFs significantly alleviate H/R injury, which include inhibiting apoptosis and enhancing antioxidant capacity. The protective effects of TFs resulted in higher expression of miR-21 in H/R-induced H9c2 cells than that of controls, which in turn upregulated Akt signaling activity via suppressing the expression of PTEN together with decreasing the ratio of Bax/Bcl-2, caspase3, and cleaved-caspase3 expression in H/R-induced H9c2 cells. Conversely, blocking miR-21 expression with miR-21 inhibitor effectively suppressed the protective effects of TFs against H/R-induced injury. Our study suggests that TFs can decrease cell apoptosis, which may be mediated by altering the expression of miR-21, PTEN/Akt, and Bcl/Bax.
Collapse
|
41
|
Bragato JP, Melo LM, Venturin GL, Rebech GT, Garcia LE, Lopes FL, de Lima VMF. Relationship of peripheral blood mononuclear cells miRNA expression and parasitic load in canine visceral leishmaniasis. PLoS One 2018; 13:e0206876. [PMID: 30517108 PMCID: PMC6281177 DOI: 10.1371/journal.pone.0206876] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023] Open
Abstract
Visceral leishmaniasis (VL) in humans is a chronic and often fatal disease if left untreated. Dogs appear to be the main reservoir host for L. infantum infection, however, in many regions other canids such as jackals, foxes, wolves and other mammals, such as hares or black rats, have been implicated as wild reservoirs. Most dogs cannot form an effective immune response against this infection, and this could be modulated by small non-coding RNAs, called microRNAs, responsible for post-transcriptional control of gene expression. Here, we evaluated the expression of miRNAs in peripheral blood mononuclear cells (PBMC) of symptomatic dogs naturally infected with Leishmania (L.) infantum (n = 10) and compared to those of healthy dogs (n = 5). Microarray analysis revealed that miR-21, miR-424, miR-194 and miR-451 had a 3-fold increase in expression, miR-192, miR-503, and miR-371 had a 2-fold increase in expression, whereas a 2-fold reduction in expression was observed for miR-150 and miR-574. Real-time PCR validated the differential expression of miR-21, miR-150, miR-451, miR-192, miR-194, and miR-371. Parasite load of PBMC was measured by real-time PCR and correlated to the differentially expressed miRNAs, showing a strong positive correlation with expression of miR-194, a regular positive correlation with miR-371 expression, and a moderate negative correlation with miR-150 expression in PBMC. These findings suggest that Leishmania infection interferes with miRNAs expression in PBMC, and their correlation with parasite load may help in the identification of therapeutic targets in Canine Visceral Leishmaniasis (CVL).
Collapse
Affiliation(s)
- Jaqueline Poleto Bragato
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Larissa Martins Melo
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Gabriela Lovizutto Venturin
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Gabriela Torres Rebech
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Leandro Encarnação Garcia
- Department of Support, Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Flavia Lombardi Lopes
- Department of Support, Production and Animal Health, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
| | - Valéria Marçal Felix de Lima
- Department of Animal Clinic, Surgery and Reproduction, São Paulo State University (Unesp), School of Veterinary Medicine, Araçatuba, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
42
|
Upadhyay S, Mittal E, Philips JA. Tuberculosis and the art of macrophage manipulation. Pathog Dis 2018; 76:4970761. [PMID: 29762680 DOI: 10.1093/femspd/fty037] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/04/2018] [Indexed: 12/24/2022] Open
Abstract
Macrophages are first-line responders against microbes. The success of Mycobacterium tuberculosis (Mtb) rests upon its ability to convert these antimicrobial cells into a permissive cellular niche. This is a remarkable accomplishment, as the antimicrobial arsenal of macrophages is extensive. Normally bacteria are delivered to an acidic, degradative lysosome through one of several trafficking pathways, including LC3-associated phagocytosis (LAP) and autophagy. Once phagocytozed, the bacilli are subjected to reactive oxygen and nitrogen species, and they induce the expression of proinflammatory cytokines, which serve to augment host responses. However, Mtb hijacks these host defense mechanisms, manipulating host cellular trafficking, innate immune responses, and cell death pathways to its benefit. The complex series of measures and countermeasures between host and pathogen ultimately determines the outcome of infection. In this review, we focus on the diverse effectors that Mtb uses in its multipronged effort to subvert the innate immune responses of macrophages. We highlight recent advances in understanding the molecular interface of the Mtb-macrophage interaction.
Collapse
Affiliation(s)
- S Upadhyay
- Division of Infectious Diseases, Department of Medicine, Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - E Mittal
- Division of Infectious Diseases, Department of Medicine, Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - J A Philips
- Division of Infectious Diseases, Department of Medicine, Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
43
|
Zhang W, Lu Q, Dong Y, Yue Y, Xiong S. Rv3033, as an Emerging Anti-apoptosis Factor, Facilitates Mycobacteria Survival via Inhibiting Macrophage Intrinsic Apoptosis. Front Immunol 2018; 9:2136. [PMID: 30319611 PMCID: PMC6168788 DOI: 10.3389/fimmu.2018.02136] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022] Open
Abstract
Apoptosis inhibition is a critical strategy of mycobacteria facilitating its survival in macrophages, but the underlying mechanism is not completely understood. In this study, we found that Rv3033, a secreted virulence factor of mycobacteria, played an important role in bacillary survival within macrophages. Forced over-expressed of Rv3033 in macrophages could efficiently resist mycobacteria-induced early and late apoptosis, accompanied with the obvious increased cellular bacterial burden. By exploring the underlying mechanism, we found that Rv3033 efficiently repressed the intrinsic (caspase-9 meditated), but not the extrinsic (caspase-8 mediated) apoptotic pathway in mycobacteria-infected macrophages. And this repression relied on the orchestrating blockade of both mitochondrial cytochrome c release and endoplasmic reticulum (ER) stress PERK branch activation. Our study uncovered a novel function of mycobacterial virulence factor Rv3033 as an anti-apoptotic protein, which may provide a new target for tuberculosis (TB) treatment.
Collapse
Affiliation(s)
- Wei Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Qian Lu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yuanshu Dong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yan Yue
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
44
|
Ma Y, Pei Q, Zhang L, Lu J, Shui T, Chen J, Shi C, Yang J, Smith M, Liu Y, Zhu J, Yang D. Live Mycobacterium leprae inhibits autophagy and apoptosis of infected macrophages and prevents engulfment of host cell by phagocytes. Am J Transl Res 2018; 10:2929-2939. [PMID: 30323879 PMCID: PMC6176229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 09/02/2018] [Indexed: 06/08/2023]
Abstract
Previous studies demonstrated that live Mycobacterium leprae (M. leprae) infection promoted macrophage differentiation toward the M2 type, with elevated interleukin (IL)-10 production. The underlying mechanism is not entirely clear. In this study, we treated macrophages with primary M. leprae strains isolated from both lepromatous leprosy (L-lep) and tuberculoid leprosy (T-lep) patients. We found that infection by live M. leprae, regardless of the primary strain, resulted in M2 skewing in the infected macrophage. This skewing was associated with downregulated IRGM expression, a core organizer protein in the autophagy assembly and reduced autophagosome formation, and with lower annexin V staining and lower caspase 3 and caspase 9 activity. Moreover, live M. leprae-infected macrophages prevented efficient phagocytosis by uninfected bystander macrophages. As a result, the phagocytes secreted less pro-inflammatory cytokines, and preferentially primed anti-inflammatory T cell responses. Together, these results suggested that live M. leprae could employ a strain-independent mechanism to suppress inflammation, possibly involving the inhibition of autophagy and apoptosis in the infected macrophages.
Collapse
Affiliation(s)
- Yuelong Ma
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| | - Qin Pei
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| | - Li Zhang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| | - Jie Lu
- Shiping County People’s HospitalYunnan 662200, China
| | - Tiejun Shui
- Yunnan Center for Disease Control and PreventionYunnan 650022, China
| | - Jia Chen
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| | - Chao Shi
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| | - Jun Yang
- Yunnan Center for Disease Control and PreventionYunnan 650022, China
| | - Michael Smith
- DICAT Biomedical Computation CentreBritish Columbia, Canada
| | - Yeqiang Liu
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| | - Jianyu Zhu
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| | - Degang Yang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of MedicineShanghai 200443, China
| |
Collapse
|
45
|
miRNAs reshape immunity and inflammatory responses in bacterial infection. Signal Transduct Target Ther 2018; 3:14. [PMID: 29844933 PMCID: PMC5968033 DOI: 10.1038/s41392-018-0006-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/20/2017] [Accepted: 12/10/2017] [Indexed: 12/15/2022] Open
Abstract
Pathogenic bacteria cause various infections worldwide, especially in immunocompromised and other susceptible individuals, and are also associated with high infant mortality rates in developing countries. MicroRNAs (miRNAs), small non-coding RNAs with evolutionarily conserved sequences, are expressed in various tissues and cells that play key part in various physiological and pathologic processes. Increasing evidence implies roles for miRNAs in bacterial infectious diseases by modulating inflammatory responses, cell penetration, tissue remodeling, and innate and adaptive immunity. This review highlights some recent intriguing findings, ranging from the correlation between aberrant expression of miRNAs with bacterial infection progression to their profound impact on host immune responses. Harnessing of dysregulated miRNAs in bacterial infection may be an approach to improving the diagnosis, prevention and therapy of infectious diseases. Changes in production of tiny cellular RNAs in response to bacterial infection could guide the development of better diagnostics and therapies. MicroRNAs regulate other genes by binding to messenger RNA strands and controlling their translation into proteins. Xikun Zhou, Min Wu and colleagues of the University of North Dakota have now reviewed current knowledge about how microRNA levels shift during infection with various bacterial pathogens. These microRNAs can modulate the immune response as well as pathways that influence metabolic activity and cell survival. Increasing studies have indicated that shifts in microRNA levels in response to different infections could provide a potential bacterial ‘fingerprint’ for achieving accurate diagnosis. With deeper insight into how different microRNAs influence infection, it might one day day become possible to target these molecules with ‘antisense’ or ‘agonist’ drugs that modulate their activity.
Collapse
|
46
|
Sabir N, Hussain T, Shah SZA, Peramo A, Zhao D, Zhou X. miRNAs in Tuberculosis: New Avenues for Diagnosis and Host-Directed Therapy. Front Microbiol 2018; 9:602. [PMID: 29651283 PMCID: PMC5885483 DOI: 10.3389/fmicb.2018.00602] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/15/2018] [Indexed: 12/30/2022] Open
Abstract
Tuberculosis (TB) is one of the most fatal infectious diseases and a leading cause of mortality, with 95% of these deaths occurring in developing countries. The causative agent, Mycobacterium tuberculosis (Mtb), has a well-established ability to circumvent the host's immune system for its intracellular survival. microRNAs (miRNAs) are small, non-coding RNAs having an important function at the post-transcriptional level and are involved in shaping immunity by regulating the repertoire of genes expressed in immune cells. It has been established in recent studies that the innate immune response against TB is significantly regulated by miRNAs. Moreover, differential expression of miRNA in Mtb infection can reflect the disease progression and may help distinguish between active and latent TB infection (LTBI). These findings encouraged the application of miRNAs as potential biomarkers. Similarly, active participation of miRNAs in modulation of autophagy and apoptosis responses against Mtb opens an exciting avenue for the exploitation of miRNAs as host directed therapy (HDT) against TB. Nanoparticles mediated delivery of miRNAs to treat various diseases has been reported and this technology has a great potential to be used in TB. In reality, this exploitation of miRNAs as biomarkers and in HDT is still in its infancy stage, and more studies using animal models mimicking human TB are advocated to assess the role of miRNAs as biomarkers and therapeutic targets. In this review, we attempt to summarize the recent advancements in the role of miRNAs in TB as immune modulator, miRNAs' capability to distinguish between active and latent TB and, finally, usage of miRNAs as therapeutic targets against TB.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiangmei Zhou
- State Key Laboratories for Agrobiotechnology, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
47
|
Zheng W, Yu A. EZH2-mediated suppression of lncRNA-LET promotes cell apoptosis and inhibits the proliferation of post-burn skin fibroblasts. Int J Mol Med 2018; 41:1949-1957. [PMID: 29393360 PMCID: PMC5810232 DOI: 10.3892/ijmm.2018.3425] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/09/2017] [Indexed: 12/28/2022] Open
Abstract
Although the upregulation of enhancer of zeste homolog 2 (EZH2) expression and downregulation of long non-coding RNA (lncRNA) LET expression are known to be associated with cell apoptosis and proliferation, little is known about the interaction of EZH2 with lncRNA LET. The present study aimed to investigate the interaction of EZH2 and lncRNA LET, and the mechanism of human dermal fibroblast (HDF) proliferation and apoptosis. Tissue samples from 33 burn patients with second- and third-degree burns and 8 controls were collected. mRNA was extracted from the burn tissues for analysis. Isolated primary HDFs were treated with heat or transfected with LET overexpression vectors, and the cell functions and associated proteins in the HDFs were analyzed. Decreased lncRNA LET expression was detected in burn tissues compared with normal skin. Heat-treated HDFs exhibited a reduction in lncRNA LET expression and increase in EZH2 expression. LET gain-of-function experiments in primary HDFs revealed increases in cell proliferation, the proportion of cells in the S stage, and cyclin D1 and cyclin-dependent kinase 4 (CDK4) expression, and reductions in the percentage of apoptotic cells, the Bax/Bcl-2 ratio and caspase-3 expression. RNA immunoprecipitation and chromatin immunoprecipitation assays demonstrated the interaction of ZH2 with lncRNA LET, and of EZH2 with H3K27me3 in HDFs. Furthermore, a negative correlation between lncRNA LET and EZH2 expression was identified. It may be concluded that increased lncRNA-LET expression promoted cell proliferation and inhibited cell apoptosis via the cyclin D1-CDK4 and Bax/Bcl-2/caspase-3 signaling pathways, respectively. Furthermore, the inhibition of lncRNA LET may be regarded as an option for use in the healing of burns.
Collapse
Affiliation(s)
- Weicai Zheng
- Department of Burns and Plastic Surgery, The First Affiliated Hospital of Henan University of Science and Technology, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Aixiang Yu
- Department of Burns and Plastic Surgery, The First Affiliated Hospital of Henan University of Science and Technology, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
48
|
Mostoufi-Afshar S, Tabatabaei M, Ghahramani Seno MM. Mycobacterium avium subsp. paratuberculosis induces differential cytosine methylation at miR-21 transcription start site region. IRANIAN JOURNAL OF VETERINARY RESEARCH 2018; 19:262-269. [PMID: 30774666 PMCID: PMC6361596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/06/2018] [Accepted: 08/28/2018] [Indexed: 06/09/2023]
Abstract
Mycobacterium avium subspecies paratuberculosis (MAP), as an obligate intracellular bacterium, causes paratuberculosis (Johne's disease) in ruminants. Plus, MAP has consistently been isolated from Crohn's disease (CD) lesions in humans; a notion implying possible direct causative effect for MAP in CD development. Infections caused by MAP are refractory to treatment and in many cases the treatment does not easily resolve the infection. Studying the molecular mechanisms of host-pathogen interaction is helpful in identifying possible drug targets. In this line, it has already been shown that in macrophages infected with various bacteria, including mycobacteria, micro RNA 21 (miR-21) is upregulated, a change that results in diminished macrophages clearance ability and favours pathogens survival within the cells. However, the molecular mechanism(s) by which the intracellular bacteria induce miR-21 expression is not known. In order to verify possible effects from epigenetic changes induced by intracellular bacteria, we studied the cytosine methylation changes at the transcription start regions of miR-21 in THP-1 macrophages infected with MAP. For this purpose, genomic DNA was extracted from infected cells and the methylation status at the region of interest was evaluated by bisulfite conversion method. Our work showed that MAP directs de-methylation of the cystosines at CpG di-nucleotides in this region, while non-CpG cytosines of this region did not show significant changes. Interestingly, the CpG cytosines that were differentially methylated in the infected macrophages occur at the binding sites of the transcription factors already known to regulate miR-21 expression.
Collapse
Affiliation(s)
- S. Mostoufi-Afshar
- Graduated from School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - M. Tabatabaei
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - M. M. Ghahramani Seno
- Department of Basic Sciences, and Division of Biotechnology, Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
49
|
Liu CH, Liu H, Ge B. Innate immunity in tuberculosis: host defense vs pathogen evasion. Cell Mol Immunol 2017; 14:963-975. [PMID: 28890547 PMCID: PMC5719146 DOI: 10.1038/cmi.2017.88] [Citation(s) in RCA: 353] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/16/2022] Open
Abstract
The major innate immune cell types involved in tuberculosis (TB) infection are macrophages, dendritic cells (DCs), neutrophils and natural killer (NK) cells. These immune cells recognize the TB-causing pathogen Mycobacterium tuberculosis (Mtb) through various pattern recognition receptors (PRRs), including but not limited to Toll-like receptors (TLRs), Nod-like receptors (NLRs) and C-type lectin receptors (CLRs). Upon infection by Mtb, the host orchestrates multiple signaling cascades via the PRRs to launch a variety of innate immune defense functions such as phagocytosis, autophagy, apoptosis and inflammasome activation. In contrast, Mtb utilizes numerous exquisite strategies to evade or circumvent host innate immunity. Here we discuss recent research on major host innate immune cells, PRR signaling, and the cellular functions involved in Mtb infection, with a specific focus on the host's innate immune defense and Mtb immune evasion. A better understanding of the molecular mechanisms underlying host-pathogen interactions could provide a rational basis for the development of effective anti-TB therapeutics.
Collapse
Affiliation(s)
- Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Haiying Liu
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100176, China
| | - Baoxue Ge
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
50
|
Xu GY, Tang XJ. Troxerutin (TXN) potentiated 5-Fluorouracil (5-Fu) treatment of human gastric cancer through suppressing STAT3/NF-κB and Bcl-2 signaling pathways. Biomed Pharmacother 2017; 92:95-107. [PMID: 28531805 DOI: 10.1016/j.biopha.2017.04.059] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/05/2017] [Accepted: 04/13/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer still presents a significant problem for public health worldwide. Troxerutin (TXN), a flavonoid present in tea, coffee, cereal grains, and a variety of fruits and vegetables, exhibits various pharmacological and biological activities in vitro and in vivo. We investigated the ability of TXN to reverse the in vitro and in vivo drug resistance of human gastric cancer cells, which were resistant to treatment of 5-Fluorouracil (5-FU). 5-Fu is a pyrimidine analog, which is widely used in the treatment of cancers. Here, we found the growth inhibitory effects of TXN on human gastric cancer cell, resistant to 5-FU. TXN and 5-FU co-treatment resulted in a dose-dependent suppression of the cell proliferation. Decreasing of phosphorylated signal transducers and activation of transcription 3 (STAT3) was included in suppression of p65 by TXN with 5-FU in combination. Additionally, the presence of TXN sensitized gastric cancer cells resistant to 5-FU to 5-FU-induced apoptosis by suppressing Bcl-2. The pro-apoptotic proteins of Bax and Bid were up-regulated, accompanied with Caspase cleavage, leading to apoptosis. Moreover, in mice xenograft models, the combined therapy inhibited tumor growth compared to the TXN or 5-FU treatment alone. Our data indicated a novel therapeutic strategy to potentiate 5-FU-induced anti-tumor effect in gastric cancer cells with resistance to 5-FU by TXN through suppression of p-STAT3/NF-κB (p65 and p50) and Bcl-2.
Collapse
Affiliation(s)
- Gui-Yun Xu
- Department of General Surgery, The Second People's Hospital of Huai'an, The Huai'an Hospital Affiliated with Xuzhou Medical College, Huai'an 223002, China; Department of General Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an 223002, China
| | - Xiao-Jun Tang
- Department of General Surgery, The Second People's Hospital of Huai'an, The Huai'an Hospital Affiliated with Xuzhou Medical College, Huai'an 223002, China; Department of General Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an 223002, China.
| |
Collapse
|