1
|
Araújo-Pereira M, Andrade BB. Oxidative battles in tuberculosis: walking the ferroptotic tightrope. Trends Immunol 2025; 46:338-351. [PMID: 40122726 DOI: 10.1016/j.it.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains one of the leading causes of death worldwide. TB pathogenesis is shaped by a complex interaction between the pathogen and host immune responses, particularly through mechanisms such as oxidative stress and ferroptosis; a form of regulated necrotic cell death driven by iron-dependent lipid peroxidation. This Review highlights recent insights into how Mtb modulates oxidative stress pathways and thus triggers ferroptosis in host cells. Understanding the interplay between oxidative stress responses and cellular and tissue necrosis opens new avenues for therapeutic interventions of TB by controlling bacterial growth and preventing host tissue damage.
Collapse
Affiliation(s)
- Mariana Araújo-Pereira
- Laboratório de Pesquisa Clínica e Translacional (LPCT), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia 40296-710, Brazil; Instituto de Pesquisa Clínica e Translacional (IPCT), Medicina Zarns, Clariens Educação, Salvador, Bahia 41720-200, Brazil; Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Institute, Salvador, Bahia 41810-710, Brazil.
| | - Bruno B Andrade
- Laboratório de Pesquisa Clínica e Translacional (LPCT), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia 40296-710, Brazil; Instituto de Pesquisa Clínica e Translacional (IPCT), Medicina Zarns, Clariens Educação, Salvador, Bahia 41720-200, Brazil; Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Institute, Salvador, Bahia 41810-710, Brazil.
| |
Collapse
|
2
|
Shankar G, Akhter Y. Stealing survival: Iron acquisition strategies of Mycobacteriumtuberculosis. Biochimie 2024; 227:37-60. [PMID: 38901792 DOI: 10.1016/j.biochi.2024.06.006] [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: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), faces iron scarcity within the host due to immune defenses. This review explores the importance of iron for Mtb and its strategies to overcome iron restriction. We discuss how the host limits iron as an innate immune response and how Mtb utilizes various iron acquisition systems, particularly the siderophore-mediated pathway. The review illustrates the structure and biosynthesis of mycobactin, a key siderophore in Mtb, and the regulation of its production. We explore the potential of targeting siderophore biosynthesis and uptake as a novel therapeutic approach for TB. Finally, we summarize current knowledge on Mtb's iron acquisition and highlight promising directions for future research to exploit this pathway for developing new TB interventions.
Collapse
Affiliation(s)
- Gauri Shankar
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226 025, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226 025, India.
| |
Collapse
|
3
|
Zhang M, Adroub S, Ummels R, Asaad M, Song L, Pain A, Bitter W, Guan Q, Abdallah AM. Comprehensive pan-genome analysis of Mycobacterium marinum: insights into genomic diversity, evolution, and pathogenicity. Sci Rep 2024; 14:27723. [PMID: 39532890 PMCID: PMC11557581 DOI: 10.1038/s41598-024-75228-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Mycobacteria is a diverse genus that includes both innocuous environmental species and serious pathogens like Mycobacterium tuberculosis, Mycobacterium leprae, and Mycobacterium ulcerans, the causative agents of tuberculosis, leprosy, and Buruli ulcer, respectively. This study focuses on Mycobacterium marinum, a closely related species known for its larger genome and ability to infect ectothermic species and cooler human extremities. Utilizing whole-genome sequencing, we conducted a comprehensive pan-genome analysis of 100 M. marinum strains, exploring genetic diversity and its impact on pathogenesis and host specificity. Our findings highlight significant genomic diversity, with clear distinctions in core, dispensable, and unique genes among the isolates. Phylogenetic analysis revealed a broad distribution of genetic lineages, challenging previous classifications into distinct clusters. Additionally, we examined the synteny and diversity of the virulence factor CpnT, noting a wide range of C-terminal domain variations across strains, which points to potential adaptations in pathogenic mechanisms. This study enhances our understanding of M. marinum's genomic architecture and its evolutionary relationship with other mycobacterial pathogens, providing insights that could inform disease control strategies for M. tuberculosis and other mycobacteria.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Sabir Adroub
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 23955-6900, Jeddah, Makkah, Saudi Arabia
| | - Roy Ummels
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center, Section Molecular Microbiology, Amsterdam, The Netherlands
| | - Mohammed Asaad
- College of Medicine, Qatar University (QU) Health, Qatar University, Doha, Qatar
| | - Lei Song
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Arnab Pain
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 23955-6900, Jeddah, Makkah, Saudi Arabia
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center, Section Molecular Microbiology, Amsterdam, The Netherlands
| | - Qingtian Guan
- Bioinformatics Laboratory, The First Hospital of Jilin University, Changchun, China.
| | - Abdallah M Abdallah
- College of Medicine, Qatar University (QU) Health, Qatar University, Doha, Qatar.
| |
Collapse
|
4
|
Choi SR, Hassan MA, Britigan BE, Narayanasamy P. Antimicrobial Activity of Gallium(III) Compounds: Pathogen-Dependent Targeting of Multiple Iron/Heme-Dependent Biological Processes. Curr Issues Mol Biol 2024; 46:9149-9161. [PMID: 39194758 DOI: 10.3390/cimb46080541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Metals play vital roles in biological systems, with iron/heme being essential for cellular and metabolic functions necessary for survival and/or virulence in many bacterial pathogens. Given the rise of bacterial resistance to current antibiotics, there is an urgent need for the development of non-toxic and novel antibiotics that do not contribute to resistance to other antibiotics. Gallium, which mimics iron, has emerged as a promising antimicrobial agent, offering a novel approach to combat bacterial infections. Gallium does not have any known functions in biological systems. Gallium exerts its effects primarily by replacing iron in redox enzymes, effectively inhibiting bacterial growth by targeting multiple iron/heme-dependent biological processes and suppressing the development of drug resistance. The aim of this review is to highlight recent findings on the mechanisms of action of gallium and provide further insights into the development of gallium-based compounds. Understanding the mechanisms underlying gallium's biological activities is crucial for designing drugs that enhance their therapeutic therapies while minimizing side effects, offering promising avenues for the treatment of infectious diseases.
Collapse
Affiliation(s)
- Seoung-Ryoung Choi
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mohammed A Hassan
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bradley E Britigan
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Prabagaran Narayanasamy
- Department of Pathology, Microbiology and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
5
|
He P, Zhao B, He W, Song Z, Pei S, Liu D, Xia H, Wang S, Ou X, Zheng Y, Zhou Y, Song Y, Wang Y, Cao X, Xing R, Zhao Y. Impact of MSMEG5257 Deletion on Mycolicibacterium smegmatis Growth. Microorganisms 2024; 12:770. [PMID: 38674714 PMCID: PMC11052289 DOI: 10.3390/microorganisms12040770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Mycobacterial membrane proteins play a pivotal role in the bacterial invasion of host cells; however, the precise mechanisms underlying certain membrane proteins remain elusive. Mycolicibacterium smegmatis (Ms) msmeg5257 is a hemolysin III family protein that is homologous to Mycobacterium tuberculosis (Mtb) Rv1085c, but it has an unclear function in growth. To address this issue, we utilized the CRISPR/Cas9 gene editor to construct Δmsmeg5257 strains and combined RNA transcription and LC-MS/MS protein profiling to determine the functional role of msmeg5257 in Ms growth. The correlative analysis showed that the deletion of msmeg5257 inhibits ABC transporters in the cytomembrane and inhibits the biosynthesis of amino acids in the cell wall. Corresponding to these results, we confirmed that MSMEG5257 localizes in the cytomembrane via subcellular fractionation and also plays a role in facilitating the transport of iron ions in environments with low iron levels. Our data provide insights that msmeg5257 plays a role in maintaining Ms metabolic homeostasis, and the deletion of msmeg5257 significantly impacts the growth rate of Ms. Furthermore, msmeg5257, a promising drug target, offers a direction for the development of novel therapeutic strategies against mycobacterial diseases.
Collapse
Affiliation(s)
- Ping He
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Bing Zhao
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Wencong He
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Zexuan Song
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Shaojun Pei
- School of Public Health, Peking University, Haidian District, Beijing 100871, China;
| | - Dongxin Liu
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Hui Xia
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Shengfen Wang
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Xichao Ou
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Yang Zheng
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Yang Zhou
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Yuanyuan Song
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Yiting Wang
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Xiaolong Cao
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Ruida Xing
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| | - Yanlin Zhao
- Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China; (P.H.); (B.Z.); (W.H.); (Z.S.); (D.L.); (H.X.); (S.W.); (X.O.); (Y.Z.); (Y.Z.); (Y.S.); (Y.W.); (X.C.); (R.X.)
| |
Collapse
|
6
|
Sao Emani C, Reiling N. The efflux pumps Rv1877 and Rv0191 play differential roles in the protection of Mycobacterium tuberculosis against chemical stress. Front Microbiol 2024; 15:1359188. [PMID: 38516013 PMCID: PMC10956863 DOI: 10.3389/fmicb.2024.1359188] [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: 12/20/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
Background It was previously shown that GlnA3sc enabled Streptomyces coelicolor to survive in excess polyamines. However, subsequent studies revealed that Rv1878, the corresponding Mycobacterium tuberculosis (M.tb) ortholog, was not essential for the detoxification of spermine (Spm), in M.tb. On the other hand, the multi-drug efflux pump Rv1877 was previously shown to enable export of a wide range of compounds, while Rv0191 was shown to be more specific to chloramphenicol. Rationale Therefore, we first wanted to determine if detoxification of Spm by efflux can be achieved by any efflux pump, or if that was dependent upon the function of the pump. Next, since Rv1878 was found not to be essential for the detoxification of Spm, we sought to follow-up on the investigation of the physiological role of Rv1878 along with Rv1877 and Rv0191. Approach To evaluate the specificity of efflux pumps in the mycobacterial tolerance to Spm, we generated unmarked ∆rv1877 and ∆rv0191 M.tb mutants and evaluated their susceptibility to Spm. To follow up on the investigation of any other physiological roles they may have, we characterized them along with the ∆rv1878 M.tb mutant. Results The ∆rv1877 mutant was sensitive to Spm stress, while the ∆rv0191 mutant was not. On the other hand, the ∆rv1878 mutant grew better than the wild-type during iron starvation yet was sensitive to cell wall stress. The proteins Rv1877 and Rv1878 seemed to play physiological roles during hypoxia and acidic stress. Lastly, the ∆rv0191 mutant was the only mutant that was sensitive to oxidative stress. Conclusion The multidrug MFS-type efflux pump Rv1877 is required for Spm detoxification, as opposed to Rv0191 which seems to play a more specific role. Moreover, Rv1878 seems to play a role in the regulation of iron homeostasis and the reconstitution of the cell wall of M.tb. On the other hand, the sensitivity of the ∆rv0191 mutant to oxidative stress, suggests that Rv0191 may be responsible for the transport of low molecular weight thiols.
Collapse
Affiliation(s)
- Carine Sao Emani
- Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Norbert Reiling
- Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
| |
Collapse
|
7
|
Zhu X, Lu Q, Li Y, Long Q, Zhang X, Long X, Cao D. Contraction and expansion dynamics: deciphering genomic underpinnings of growth rate and pathogenicity in Mycobacterium. Front Microbiol 2023; 14:1292897. [PMID: 38075891 PMCID: PMC10701892 DOI: 10.3389/fmicb.2023.1292897] [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: 09/12/2023] [Accepted: 11/01/2023] [Indexed: 10/16/2024] Open
Abstract
BACKGROUND Mycobacterium bacteria, encompassing both slow growth (SGM) and rapid growth mycobacteria (RGM), along with true pathogenic (TP), opportunistic pathogenic (OP), and non-pathogenic (NP) types, exhibit diverse phenotypes. Yet, the genetic underpinnings of these variations remain elusive. METHODS Here, We conducted a comprehensive comparative genomics study involving 53 Mycobacterium species to unveil the genomic drivers behind growth rate and pathogenicity disparities. RESULTS Our core/pan-genome analysis highlighted 1,307 shared gene families, revealing an open pan-genome structure. A phylogenetic tree highlighted clear boundaries between SGM and RGM, as well as TP and other species. Gene family contraction emerged as the primary alteration associated with growth and pathogenicity transitions. Specifically, ABC transporters for amino acids and inorganic ions, along with quorum sensing genes, exhibited significant contractions in SGM species, potentially influencing their distinct traits. Conversely, TP strains displayed contraction in lipid and secondary metabolite biosynthesis and metabolism-related genes. Across the 53 species, we identified 26 core and 64 accessory virulence factors. Remarkably, TP and OP strains stood out for their expanded mycobactin biosynthesis and type VII secretion system gene families, pivotal for their pathogenicity. CONCLUSION Our findings underscore the importance of gene family contraction in nucleic acids, ions, and substance metabolism for host adaptation, while emphasizing the significance of virulence gene family expansion, including type VII secretion systems and mycobactin biosynthesis, in driving mycobacterial pathogenicity.
Collapse
Affiliation(s)
- Xiaoying Zhu
- Clinical Pathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Medical College, Guangxi University, Nanning, Guangxi, China
| | - Qunfeng Lu
- Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi, China
- School of Medical Laboratory Sciences, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Yulei Li
- Clinical Pathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Qinqin Long
- Clinical Pathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xinyu Zhang
- Clinical Pathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xidai Long
- Clinical Pathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Medical College, Guangxi University, Nanning, Guangxi, China
| | - Demin Cao
- Clinical Pathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| |
Collapse
|
8
|
Anand PK, Kaur G, Saini V, Kaur J, Kaur J. N-terminal PPE domain plays an integral role in extracellular transportation and stability of the immunomodulatory Rv3539 protein of the Mycobacterium tuberculosis. Biochimie 2023; 213:30-40. [PMID: 37156406 DOI: 10.1016/j.biochi.2023.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/31/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Multigene PE/PPE family is exclusively present in mycobacterium species. Only few selected genes of this family have been characterized till date. Rv3539 was annotated as PPE63 with conserved PPE domain at N-terminal and PE-PPE at C-terminal. An α/β hydrolase structural fold, characteristic of lipase/esterase, was present in the PE-PPE domain. To assign the biochemical function to Rv3539, the corresponding gene was cloned in pET-32a (+) as full-length, PPE, and PE-PPE domains individually, followed by expression in E. Coli C41 (DE3). All three proteins demonstrated esterase activity. However, the enzyme activity in the N-terminal PPE domain was very low. The enzyme activity of Rv3539 and PE-PPE proteins was approximately same with the pNP-C4 as optimum substrate at 40 °C and pH 8.0. The loss of enzyme activity after mutating the predicted catalytic triad (Ser296Ala, Asp369Ala, and His395Ala) found only in the PE-PPE domain, confirmed the candidature of the bioinformatically predicted active site residue. The optimal activity and thermostability of the Rv3539 protein was altered by removing the PPE domain. CD-spectroscopy analysis confirmed the role of PPE domain to the thermostability of Rv3539 by maintaining the structural integrity at higher temperatures. The presence of the N-terminal PPE domain directed the Rv3539 protein to the cell membrane/wall and the extracellular compartment. The Rv3539 protein could generate humoral response in TB patients. Therefore, results demonstrated that Rv3539 demonstrated esterase activity. PE-PPE domain of Rv3539 is functionally automated, however, N-terminus domain played a role in protein stabilization and its transportation. Both domains participated in immunomodulation.
Collapse
Affiliation(s)
- Pradeep Kumar Anand
- Department of Biotechnology, BMS Block-1, South Campus, Panjab University, Chandigarh, 160014, India.
| | - Gagandeep Kaur
- Department of Biotechnology, BMS Block-1, South Campus, Panjab University, Chandigarh, 160014, India.
| | - Varinder Saini
- Department of Pulmonary Medicine, Government Medical College and Hospital, Chandigarh, India.
| | - Jasbinder Kaur
- Department of Biochemistry, Government Medical College and Hospital, Chandigarh, India.
| | - Jagdeep Kaur
- Department of Biotechnology, BMS Block-1, South Campus, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
9
|
Guo F, Wei J, Song Y, Li B, Qian Z, Wang X, Wang H, Xu T. Immunological effects of the PE/PPE family proteins of Mycobacterium tuberculosis and related vaccines. Front Immunol 2023; 14:1255920. [PMID: 37841250 PMCID: PMC10569470 DOI: 10.3389/fimmu.2023.1255920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/25/2023] [Indexed: 10/17/2023] Open
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (Mtb), and its incidence and mortality are increasing. The BCG vaccine was developed in the early 20th century. As the most widely administered vaccine in the world, approximately 100 million newborns are vaccinated with BCG every year, which has saved tens of millions of lives. However, due to differences in region and race, the average protective rate of BCG in preventing tuberculosis in children is still not high in some areas. Moreover, because the immune memory induced by BCG will weaken with the increase of age, it is slightly inferior in preventing adult tuberculosis, and BCG revaccination cannot reduce the incidence of tuberculosis again. Research on the mechanism of Mtb and the development of new vaccines against TB are the main strategies for preventing and treating TB. In recent years, Pro-Glu motif-containing (PE) and Pro-Pro-Glu motif-containing (PPE) family proteins have been found to have an increasingly important role in the pathogenesis and chronic protracted infection observed in TB. The development and clinical trials of vaccines based on Mtb antigens are in progress. Herein, we review the immunological effects of PE/PPE proteins and the development of common PE/PPE vaccines.
Collapse
Affiliation(s)
- Fangzheng Guo
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
| | - Jing Wei
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
| | - Yamin Song
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
| | - Baiqing Li
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical College, Bengbu, China
| | - Zhongqing Qian
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical College, Bengbu, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Bengbu Medical College, Bengbu, China
| | - Hongtao Wang
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical College, Bengbu, China
| | - Tao Xu
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical College, Bengbu, China
| |
Collapse
|
10
|
de Miranda R, Cuthbert BJ, Klevorn T, Chao A, Mendoza J, Arbing M, Sieminski PJ, Papavinasasundaram K, Abdul-Hafiz S, Chan S, Sassetti CM, Ehrt S, Goulding CW. Differentiating the roles of Mycobacterium tuberculosis substrate binding proteins, FecB and FecB2, in iron uptake. PLoS Pathog 2023; 19:e1011650. [PMID: 37747938 PMCID: PMC10553834 DOI: 10.1371/journal.ppat.1011650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 10/05/2023] [Accepted: 08/31/2023] [Indexed: 09/27/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, poses a great threat to human health. With the emergence of drug resistant Mtb strains, new therapeutics are desperately needed. As iron is critical to the growth and survival of Mtb, mechanisms through which Mtb acquires host iron represent attractive therapeutic targets. Mtb scavenges host iron via Mtb siderophore-dependent and heme iron uptake pathways. While multiple studies describe the import of heme and ferric-siderophores and the export of apo-siderophores across the inner membrane, little is known about their transport across the periplasm and cell-wall environments. Mtb FecB and FecB2 are predicted periplasmic binding proteins implicated in host iron acquisition; however, their precise roles are not well understood. This study sought to differentiate the roles FecB and FecB2 play in Mtb iron acquisition. The crystallographic structures of Mtb FecB and FecB2 were determined to 2.0 Å and 2.2 Å resolution, respectively, and show distinct ligand binding pockets. In vitro ligand binding experiments for FecB and FecB2 were performed with heme and bacterial siderophores from Mtb and other species, revealing that both FecB and FecB2 bind heme, while only FecB binds the Mtb sideophore ferric-carboxymycobactin (Fe-cMB). Subsequent structure-guided mutagenesis of FecB identified a single glutamate residue-Glu339-that significantly contributes to Fe-cMB binding. A role for FecB in the Mtb siderophore-mediated iron acquisition pathway was corroborated by Mycobacterium smegmatis and Mtb pull-down assays, which revealed interactions between FecB and members of the mycobacterial siderophore export and import machinery. Similarly, pull-down assays with FecB2 confirms its role in heme uptake revealing interactions with a potential inner membrane heme importer. Due to ligand preference and protein partners, our data suggest that Mtb FecB plays a role in siderophore-dependent iron and heme acquisition pathways; in addition, we confirm that Mtb FecB2 is involved in heme uptake.
Collapse
Affiliation(s)
- Rodger de Miranda
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Bonnie J. Cuthbert
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Thaís Klevorn
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Alex Chao
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Jessica Mendoza
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Mark Arbing
- UCLA-DOE Institute, UCLA, Los Angeles, Calofornia, United States of America
| | - Paul J. Sieminski
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Kadamba Papavinasasundaram
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - Sumer Abdul-Hafiz
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Sum Chan
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Christopher M. Sassetti
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - Sabine Ehrt
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Celia W. Goulding
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, Califiornia, United States of America
| |
Collapse
|
11
|
Li F, Guo X, Bi Y, Jia R, Pitt ME, Pan S, Li S, Gasser RB, Coin LJ, Song J. Digerati - A multipath parallel hybrid deep learning framework for the identification of mycobacterial PE/PPE proteins. Comput Biol Med 2023; 163:107155. [PMID: 37356289 DOI: 10.1016/j.compbiomed.2023.107155] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/27/2023]
Abstract
The genome of Mycobacterium tuberculosis contains a relatively high percentage (10%) of genes that are poorly characterised because of their highly repetitive nature and high GC content. Some of these genes encode proteins of the PE/PPE family, which are thought to be involved in host-pathogen interactions, virulence, and disease pathogenicity. Members of this family are genetically divergent and challenging to both identify and classify using conventional computational tools. Thus, advanced in silico methods are needed to identify proteins of this family for subsequent functional annotation efficiently. In this study, we developed the first deep learning-based approach, termed Digerati, for the rapid and accurate identification of PE and PPE family proteins. Digerati was built upon a multipath parallel hybrid deep learning framework, which equips multi-layer convolutional neural networks with bidirectional, long short-term memory, equipped with a self-attention module to effectively learn the higher-order feature representations of PE/PPE proteins. Empirical studies demonstrated that Digerati achieved a significantly better performance (∼18-20%) than alignment-based approaches, including BLASTP, PHMMER, and HHsuite, in both prediction accuracy and speed. Digerati is anticipated to facilitate community-wide efforts to conduct high-throughput identification and analysis of PE/PPE family members. The webserver and source codes of Digerati are publicly available at http://web.unimelb-bioinfortools.cloud.edu.au/Digerati/.
Collapse
Affiliation(s)
- Fuyi Li
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, 792 Elizabeth Street, Melbourne, Victoria, 3000, Australia.
| | - Xudong Guo
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
| | - Yue Bi
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, 3800, Australia
| | - Runchang Jia
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
| | - Miranda E Pitt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, 792 Elizabeth Street, Melbourne, Victoria, 3000, Australia
| | - Shirui Pan
- School of Information and Communication Technology, Griffith University, QLD, 4222, Australia
| | - Shuqin Li
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
| | - Robin B Gasser
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, VIC, 3010, Australia
| | - Lachlan Jm Coin
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, 792 Elizabeth Street, Melbourne, Victoria, 3000, Australia.
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, 3800, Australia.
| |
Collapse
|
12
|
Sankey N, Merrick H, Singh P, Rogers J, Reddi A, Hartson SD, Mitra A. Role of the Mycobacterium tuberculosis ESX-4 Secretion System in Heme Iron Utilization and Pore Formation by PPE Proteins. mSphere 2023; 8:e0057322. [PMID: 36749044 PMCID: PMC10117145 DOI: 10.1128/msphere.00573-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/16/2023] [Indexed: 02/08/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is transmitted through aerosols and primarily colonizes within the lung. The World Health Organization estimates that Mtb kills ~1.4 million people every year. A key aspect that makes Mtb such a successful pathogen is its ability to overcome iron limitation mounted by the host immune response. In our previous studies, we have shown that Mtb can utilize iron from heme, the largest source of iron in the human host, and that it uses two redundant heme utilization pathways. In this study, we show that the ESX-4 type VII secretion system (T7SS) is necessary for extracellular heme uptake into the Mtb cell through both heme utilization pathways. ESX-4 influences the secretion of the culture filtrate proteins Rv0125 and Rv1085c, which are also necessary for efficient heme utilization. We also discovered that deletion of the alternative sigma factor SigM significantly reduced Mtb heme utilization through both pathways and predict that SigM is a global positive regulator of core heme utilization genes of both pathways. Finally, we present the first direct evidence that some mycobacterial PPE (proline-proline-glutamate motif) proteins of the PPE protein family are pore-forming membrane proteins. Altogether, we identified core components of both Mtb Heme utilization pathways that were previously unknown and identified a novel channel-forming membrane protein of Mtb. IMPORTANCE M. tuberculosis (Mtb) is completely dependent on iron acquisition in the host to cause disease. The largest source of iron for Mtb in the human host is heme. Here, we show that the ancestral ESX-4 type VII secretion system is required for the efficient utilization of heme as a source of iron, which is an essential nutrient. This is another biological function identified for ESX-4 in Mtb, whose contribution to Mtb physiology is poorly understood. A most exciting finding is that some mycobacterial PPE (proline-proline-glutamate motif) proteins that have been implicated in the nutrient acquisition are membrane proteins that can form channels in a lipid bilayer. These observations have far-reaching implications because they support an emerging theme that PPE proteins can function as channel proteins in the outer mycomembrane for nutrient acquisition. Mtb has evolved a heme uptake system that is drastically different from all other known bacterial heme acquisition systems.
Collapse
Affiliation(s)
- November Sankey
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Haley Merrick
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Padam Singh
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Janet Rogers
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Amit Reddi
- School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Steven D. Hartson
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Avishek Mitra
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
13
|
D'Souza C, Kishore U, Tsolaki AG. The PE-PPE Family of Mycobacterium tuberculosis: Proteins in Disguise. Immunobiology 2023; 228:152321. [PMID: 36805109 DOI: 10.1016/j.imbio.2022.152321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
Mycobacterium tuberculosis has thrived in parallel with humans for millennia, and despite our efforts, M. tuberculosis continues to plague us, currently infecting a third of the world's population. The success of M. tuberculosis has recently been attributed, in part, to the PE-PPE family; a unique collection of 168 proteins fundamentally involved in the pathogenesis of M. tuberculosis. The PE-PPE family proteins have been at the forefront of intense research efforts since their discovery in 1998 and whilst our knowledge and understanding has significantly advanced over the last two decades, many important questions remain to be elucidated. This review consolidates and examines the vast body of existing literature regarding the PE-PPE family proteins, with respect to the latest developments in elucidating their evolution, structure, subcellular localisation, function, and immunogenicity. This review also highlights significant inconsistencies and contradictions within the field. Additionally, possible explanations for these knowledge gaps are explored. Lastly, this review poses many important questions, which need to be addressed to complete our understanding of the PE-PPE family, as well as highlighting the challenges associated with studying this enigmatic family of proteins. Further research into the PE-PPE family, together with technological advancements in genomics and proteomics, will undoubtedly improve our understanding of the pathogenesis of M. tuberculosis, as well as identify key targets/candidates for the development of novel drugs, diagnostics, and vaccines.
Collapse
Affiliation(s)
- Christopher D'Souza
- Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, United Kingdom
| | - Uday Kishore
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Anthony G Tsolaki
- Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, United Kingdom.
| |
Collapse
|
14
|
Donegan RK, Fu Y, Copeland J, Idga S, Brown G, Hale OF, Mitra A, Yang H, Dailey HA, Niederweis M, Jain P, Reddi AR. Exogenously Scavenged and Endogenously Synthesized Heme Are Differentially Utilized by Mycobacterium tuberculosis. Microbiol Spectr 2022; 10:e0360422. [PMID: 36169423 PMCID: PMC9604157 DOI: 10.1128/spectrum.03604-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 01/12/2023] Open
Abstract
Heme is both an essential cofactor and an abundant source of nutritional iron for the human pathogen Mycobacterium tuberculosis. While heme is required for M. tuberculosis survival and virulence, it is also potentially cytotoxic. Since M. tuberculosis can both synthesize and take up heme, the de novo synthesis of heme and its acquisition from the host may need to be coordinated in order to mitigate heme toxicity. However, the mechanisms employed by M. tuberculosis to regulate heme uptake, synthesis, and bioavailability are poorly understood. By integrating ratiometric heme sensors with mycobacterial genetics, cell biology, and biochemistry, we determined that de novo-synthesized heme is more bioavailable than exogenously scavenged heme, and heme availability signals the downregulation of heme biosynthetic enzyme gene expression. Ablation of heme synthesis does not result in the upregulation of known heme import proteins. Moreover, we found that de novo heme synthesis is critical for survival from macrophage assault. Altogether, our data suggest that mycobacteria utilize heme from endogenous and exogenous sources differently and that targeting heme synthesis may be an effective therapeutic strategy to treat mycobacterial infections. IMPORTANCE Mycobacterium tuberculosis infects ~25% of the world's population and causes tuberculosis (TB), the second leading cause of death from infectious disease. Heme is an essential metabolite for M. tuberculosis, and targeting the unique heme biosynthetic pathway of M. tuberculosis could serve as an effective therapeutic strategy. However, since M. tuberculosis can both synthesize and scavenge heme, it was unclear if inhibiting heme synthesis alone could serve as a viable approach to suppress M. tuberculosis growth and virulence. The importance of this work lies in the development and application of genetically encoded fluorescent heme sensors to probe bioavailable heme in M. tuberculosis and the discovery that endogenously synthesized heme is more bioavailable than exogenously scavenged heme. Moreover, it was found that heme synthesis protected M. tuberculosis from macrophage killing, and bioavailable heme in M. tuberculosis is diminished during macrophage infection. Altogether, these findings suggest that targeting M. tuberculosis heme synthesis is an effective approach to combat M. tuberculosis infections.
Collapse
Affiliation(s)
- Rebecca K. Donegan
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA
- Department of Chemistry, Barnard College, New York, New York, USA
| | - Yibo Fu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Jacqueline Copeland
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Stanzin Idga
- Department of Pathology, Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, New York, USA
| | - Gabriel Brown
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Owen F. Hale
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Avishek Mitra
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hui Yang
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Harry A. Dailey
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Michael Niederweis
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Paras Jain
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Cell Therapy and Cell Engineering Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Amit R. Reddi
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
15
|
Donegan RK. The role of host heme in bacterial infection. Biol Chem 2022; 403:1017-1029. [PMID: 36228088 DOI: 10.1515/hsz-2022-0192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/14/2022] [Indexed: 11/15/2022]
Abstract
Heme is an indispensable cofactor for almost all aerobic life, including the human host and many bacterial pathogens. During infection, heme and hemoproteins are the largest source of bioavailable iron, and pathogens have evolved various heme acquisition pathways to satisfy their need for iron and heme. Many of these pathways are regulated transcriptionally by intracellular iron levels, however, host heme availability and intracellular heme levels have also been found to regulate heme uptake in some species. Knowledge of these pathways has helped to uncover not only how these bacteria incorporate host heme into their metabolism but also provided insight into the importance of host heme as a nutrient source during infection. Within this review is covered multiple aspects of the role of heme at the host pathogen interface, including the various routes of heme biosynthesis, how heme is sequestered by the host, and how heme is scavenged by bacterial pathogens. Also discussed is how heme and hemoproteins alter the behavior of the host immune system and bacterial pathogens. Finally, some unanswered questions about the regulation of heme uptake and how host heme is integrated into bacterial metabolism are highlighted.
Collapse
Affiliation(s)
- Rebecca K Donegan
- Department of Chemistry, Barnard College, 3009 Broadway, New York, NY, 10027, USA
| |
Collapse
|
16
|
Ahmad F, Rani A, Alam A, Zarin S, Pandey S, Singh H, Hasnain SE, Ehtesham NZ. Macrophage: A Cell With Many Faces and Functions in Tuberculosis. Front Immunol 2022; 13:747799. [PMID: 35603185 PMCID: PMC9122124 DOI: 10.3389/fimmu.2022.747799] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 03/30/2022] [Indexed: 01/16/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of human tuberculosis (TB) which primarily infects the macrophages. Nearly a quarter of the world's population is infected latently by Mtb. Only around 5%-10% of those infected develop active TB disease, particularly during suppressed host immune conditions or comorbidity such as HIV, hinting toward the heterogeneity of Mtb infection. The aerosolized Mtb first reaches the lungs, and the resident alveolar macrophages (AMs) are among the first cells to encounter the Mtb infection. Evidence suggests that early clearance of Mtb infection is associated with robust innate immune responses in resident macrophages. In addition to lung-resident macrophage subsets, the recruited monocytes and monocyte-derived macrophages (MDMs) have been suggested to have a protective role during Mtb infection. Mtb, by virtue of its unique cell surface lipids and secreted protein effectors, can evade killing by the innate immune cells and preferentially establish a niche within the AMs. Continuous efforts to delineate the determinants of host defense mechanisms have brought to the center stage the crucial role of macrophage phenotypical variations for functional adaptations in TB. The morphological and functional heterogeneity and plasticity of the macrophages aid in confining the dissemination of Mtb. However, during a suppressed or hyperactivated immune state, the Mtb virulence factors can affect macrophage homeostasis which may skew to favor pathogen growth, causing active TB. This mini-review is aimed at summarizing the interplay of Mtb pathomechanisms in the macrophages and the implications of macrophage heterogeneity and plasticity during Mtb infection.
Collapse
Affiliation(s)
- Faraz Ahmad
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Anshu Rani
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
| | - Anwar Alam
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Sheeba Zarin
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Saurabh Pandey
- Department of Biochemistry, Jamia Hamdard, New Delhi, India
| | - Hina Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, India
| | - Nasreen Zafar Ehtesham
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| |
Collapse
|
17
|
Chengalroyen MD, Mason MK, Borsellini A, Tassoni R, Abrahams GL, Lynch S, Ahn YM, Ambler J, Young K, Crowley BM, Olsen DB, Warner DF, Barry III CE, Boshoff HIM, Lamers MH, Mizrahi V. DNA-Dependent Binding of Nargenicin to DnaE1 Inhibits Replication in Mycobacterium tuberculosis. ACS Infect Dis 2022; 8:612-625. [PMID: 35143160 PMCID: PMC8922275 DOI: 10.1021/acsinfecdis.1c00643] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Indexed: 12/15/2022]
Abstract
Natural products provide a rich source of potential antimicrobials for treating infectious diseases for which drug resistance has emerged. Foremost among these diseases is tuberculosis. Assessment of the antimycobacterial activity of nargenicin, a natural product that targets the replicative DNA polymerase of Staphylococcus aureus, revealed that it is a bactericidal genotoxin that induces a DNA damage response in Mycobacterium tuberculosis (Mtb) and inhibits growth by blocking the replicative DNA polymerase, DnaE1. Cryo-electron microscopy revealed that binding of nargenicin to Mtb DnaE1 requires the DNA substrate such that nargenicin is wedged between the terminal base pair and the polymerase and occupies the position of both the incoming nucleotide and templating base. Comparative analysis across three bacterial species suggests that the activity of nargenicin is partly attributable to the DNA binding affinity of the replicative polymerase. This work has laid the foundation for target-led drug discovery efforts focused on Mtb DnaE1.
Collapse
Affiliation(s)
- Melissa D. Chengalroyen
- SAMRC/NHLS/UCT
Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence
for Biomedical TB Research, Institute of Infectious Disease and Molecular
Medicine and Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, South Africa
| | - Mandy K. Mason
- SAMRC/NHLS/UCT
Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence
for Biomedical TB Research, Institute of Infectious Disease and Molecular
Medicine and Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, South Africa
| | - Alessandro Borsellini
- Cell
and Chemical Biology, Leiden University
Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Raffaella Tassoni
- Cell
and Chemical Biology, Leiden University
Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Garth L. Abrahams
- SAMRC/NHLS/UCT
Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence
for Biomedical TB Research, Institute of Infectious Disease and Molecular
Medicine and Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, South Africa
- Tuberculosis
Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease,
National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United
States
| | - Sasha Lynch
- SAMRC/NHLS/UCT
Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence
for Biomedical TB Research, Institute of Infectious Disease and Molecular
Medicine and Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, South Africa
| | - Yong-Mo Ahn
- Tuberculosis
Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease,
National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United
States
| | - Jon Ambler
- Wellcome
Centre for Infectious Diseases Research in Africa, University of Cape Town, Anzio Road, Observatory 7925, South Africa
| | - Katherine Young
- Infectious
Disease, Merck & Co. Inc., West Point, Pennsylvania 19446, United States
| | - Brendan M. Crowley
- Discovery
Chemistry, Merck & Co. Inc., West Point, Pennsylvania 19446, United States
| | - David B. Olsen
- Infectious
Disease, Merck & Co. Inc., West Point, Pennsylvania 19446, United States
| | - Digby F. Warner
- SAMRC/NHLS/UCT
Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence
for Biomedical TB Research, Institute of Infectious Disease and Molecular
Medicine and Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, South Africa
| | - Clifton E. Barry III
- Tuberculosis
Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease,
National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United
States
| | - Helena I. M. Boshoff
- Tuberculosis
Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease,
National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United
States
| | - Meindert H. Lamers
- Cell
and Chemical Biology, Leiden University
Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Valerie Mizrahi
- SAMRC/NHLS/UCT
Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence
for Biomedical TB Research, Institute of Infectious Disease and Molecular
Medicine and Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, South Africa
| |
Collapse
|
18
|
Peng Z, Yue Y, Xiong S. Mycobacterial PPE36 Modulates Host Inflammation by Promoting E3 Ligase Smurf1-Mediated MyD88 Degradation. Front Immunol 2022; 13:690667. [PMID: 35237255 PMCID: PMC8882603 DOI: 10.3389/fimmu.2022.690667] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 01/21/2022] [Indexed: 11/18/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) PPE36, a cell-wall-associated protein, is highly specific and conserved for the Mtb complex group. Although PPE36 has been proven essential for iron utilization, little is known about it in regulating host immune responses. Here we exhibited that PPE36 was preferentially enriched in Mtb virulent strains and could efficiently inhibit host inflammatory responses and increase bacterial loads in infected macrophages and mice. In exploring the underlying mechanisms, we found that PPE36 could robustly inhibit the activation of inflammatory NF-κB and MAPK (Erk, p38, and Jnk) pathways by promoting E3 ligase Smurf1-mediated ubiquitination and proteasomal degradation of MyD88 protein. Our research revealed a previously unknown function of PPE36 on modulating host immune responses and provided some clues to the development of novel tuberculosis treatment strategies based on immune regulation.
Collapse
Affiliation(s)
- Zhangli Peng
- 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
|
19
|
Sharma S, Sharma M. Proline-Glutamate/Proline-Proline-Glutamate (PE/PPE) proteins of Mycobacterium tuberculosis: The multifaceted immune-modulators. Acta Trop 2021; 222:106035. [PMID: 34224720 DOI: 10.1016/j.actatropica.2021.106035] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 12/30/2022]
Abstract
The PE/PPE proteins encoded by seven percent (7%) of Mycobacterium tuberculosis (Mtb) genome are the chief constituents to pathogen's virulence reservoir. The fact that these genes have evolved along ESX secretory system in pathogenic Mtb strains make their investigation very intriguing. There is lot of speculation about the prominent role of these proteins at host pathogen interface and in disease pathogenesis. Nevertheless, the exact function of PE/PPE proteins still remains a mystery which calls for further research targeting these proteins. This article is an effort to document all the facts known so far with regard to these unique proteins which involves their origin, evolution, transcriptional control, and most important their role as host immune-modulators. Our understanding strongly points towards the versatile nature of these PE/PPE proteins as Mtb's host immune sensors and as decisive factors in shaping the outcome of infection. Further investigation on these proteins will surely pave way for newer and effective vaccines and therapeutics to control Tuberculosis (TB).
Collapse
Affiliation(s)
- Sadhna Sharma
- DS Kothari Central Interdisciplinary Research Centre and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| | - Monika Sharma
- DS Kothari Central Interdisciplinary Research Centre and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| |
Collapse
|
20
|
Jackson M, Stevens CM, Zhang L, Zgurskaya HI, Niederweis M. Transporters Involved in the Biogenesis and Functionalization of the Mycobacterial Cell Envelope. Chem Rev 2021; 121:5124-5157. [PMID: 33170669 PMCID: PMC8107195 DOI: 10.1021/acs.chemrev.0c00869] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The biology of mycobacteria is dominated by a complex cell envelope of unique composition and structure and of exceptionally low permeability. This cell envelope is the basis of many of the pathogenic features of mycobacteria and the site of susceptibility and resistance to many antibiotics and host defense mechanisms. This review is focused on the transporters that assemble and functionalize this complex structure. It highlights both the progress and the limits of our understanding of how (lipo)polysaccharides, (glyco)lipids, and other bacterial secretion products are translocated across the different layers of the cell envelope to their final extra-cytoplasmic location. It further describes some of the unique strategies evolved by mycobacteria to import nutrients and other products through this highly impermeable barrier.
Collapse
Affiliation(s)
- Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523-1682, USA
| | - Casey M. Stevens
- University of Oklahoma, Department of Chemistry and Biochemistry, 101 Stephenson Parkway, Norman, OK 73019, USA
| | - Lei Zhang
- Department of Microbiology, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL 35294, USA
| | - Helen I. Zgurskaya
- University of Oklahoma, Department of Chemistry and Biochemistry, 101 Stephenson Parkway, Norman, OK 73019, USA
| | - Michael Niederweis
- Department of Microbiology, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL 35294, USA
| |
Collapse
|
21
|
Nutritional immunity: the impact of metals on lung immune cells and the airway microbiome during chronic respiratory disease. Respir Res 2021; 22:133. [PMID: 33926483 PMCID: PMC8082489 DOI: 10.1186/s12931-021-01722-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Nutritional immunity is the sequestration of bioavailable trace metals such as iron, zinc and copper by the host to limit pathogenicity by invading microorganisms. As one of the most conserved activities of the innate immune system, limiting the availability of free trace metals by cells of the immune system serves not only to conceal these vital nutrients from invading bacteria but also operates to tightly regulate host immune cell responses and function. In the setting of chronic lung disease, the regulation of trace metals by the host is often disrupted, leading to the altered availability of these nutrients to commensal and invading opportunistic pathogenic microbes. Similarly, alterations in the uptake, secretion, turnover and redox activity of these vitally important metals has significant repercussions for immune cell function including the response to and resolution of infection. This review will discuss the intricate role of nutritional immunity in host immune cells of the lung and how changes in this fundamental process as a result of chronic lung disease may alter the airway microbiome, disease progression and the response to infection.
Collapse
|
22
|
Pisu D, Huang L, Grenier JK, Russell DG. Dual RNA-Seq of Mtb-Infected Macrophages In Vivo Reveals Ontologically Distinct Host-Pathogen Interactions. Cell Rep 2021; 30:335-350.e4. [PMID: 31940480 PMCID: PMC7032562 DOI: 10.1016/j.celrep.2019.12.033] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/31/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022] Open
Abstract
Dissecting the in vivo host-pathogen interplay is crucial to understanding the molecular mechanisms governing control or progression of intracellular infections. In this work, we explore the in vivo molecular dynamics of Mtb infection by performing dual RNA-seq on Mycobacterium tuberculosis-infected, ontogenetically distinct macrophage lineages isolated directly from murine lungs. We first define an in vivo signature of 180 genes specifically upregulated by Mtb in mouse lung macrophages, then we uncover a divergent transcriptional response of the bacteria between alveolar macrophages that appear to sustain Mtb growth through increased access to iron and fatty acids and interstitial macrophages that restrict Mtb growth through iron sequestration and higher levels of nitric oxide. We use an enrichment protocol for bacterial transcripts, which enables us to probe Mtb physiology at the host cell level in an in vivo environment, with broader application in understanding the infection dynamics of intracellular pathogens in general. In this study Pisu et al. performed dual RNA-seq on Mycobacterium tuberculosis-infected, ontogenetically distinct macrophage lineages isolated directly from infected murine lungs. The transcriptional response of host and bacteria diverged between alveolar macrophages that sustain Mtb growth and interstitial macrophages that restrict Mtb growth.
Collapse
Affiliation(s)
- Davide Pisu
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Lu Huang
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Jennifer K Grenier
- RNA Sequencing Core, Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - David G Russell
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
23
|
Qian J, Chen R, Wang H, Zhang X. Role of the PE/PPE Family in Host-Pathogen Interactions and Prospects for Anti-Tuberculosis Vaccine and Diagnostic Tool Design. Front Cell Infect Microbiol 2020; 10:594288. [PMID: 33324577 PMCID: PMC7726347 DOI: 10.3389/fcimb.2020.594288] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/31/2020] [Indexed: 12/13/2022] Open
Abstract
The pe/ppe genes are found in pathogenic, slow-growing Mycobacterium tuberculosis and other M. tuberculosis complex (MTBC) species. These genes are considered key factors in host-pathogen interactions. Although the function of most PE/PPE family proteins remains unclear, accumulating evidence suggests that this family is involved in M. tuberculosis infection. Here, we review the role of PE/PPE proteins, which are believed to be linked to the ESX system function. Further, we highlight the reported functions of PE/PPE proteins, including their roles in host cell interaction, immune response regulation, and cell fate determination during complex host-pathogen processes. Finally, we propose future directions for PE/PPE protein research and consider how the current knowledge might be applied to design more specific diagnostics and effective vaccines for global tuberculosis control.
Collapse
Affiliation(s)
- Jianing Qian
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Run Chen
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Honghai Wang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Xuelian Zhang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Choi SR, Switzer B, Britigan BE, Narayanasamy P. Gallium Porphyrin and Gallium Nitrate Synergistically Inhibit Mycobacterial Species by Targeting Different Aspects of Iron/Heme Metabolism. ACS Infect Dis 2020; 6:2582-2591. [PMID: 32845117 DOI: 10.1021/acsinfecdis.0c00113] [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: 12/19/2022]
Abstract
There is an urgent need for new effective and safe antibiotics active against pathogenic mycobacterial species. Gallium (Ga) nitrate (Ga(NO3)3) and Ga porphyrin (GaPP) have each been shown to inhibit the growth of a variety of mycobacterial species. The Ga(III) ion derived from Ga(NO3)3 has the potential to disrupt the mycobacterial Fe(III) uptake mechanisms and utilization, including replacing iron (Fe) in the active site of enzymes, resulting in the disruption of function. Similarly, noniron metalloporphyrins such as heme mimetics, which can be transported across the bacterial membrane via heme-uptake pathways, would potentially block the acquisition of iron-containing heme and bind to heme-utilizing proteins, making them nonfunctional. Given that they likely act on different aspects of mycobacterial Fe metabolism, the efficacy of combining Ga(NO3)3 and GaPP was studied in vitro against Mycobacterium avium, Mycobacterium abscessus, and Mycobacterium tuberculosis (M. tb). The combination was then assessed in vivo in a murine pulmonary infection model of M. abscessus. We observed that Ga(NO3)3 in combination with GaPP exhibited synergistic inhibitory activity against the growth of M. avium, M. tb, and M. abscessus, being most active against M. abscessus. Activity assays indicated that Ga(NO3)3 and GaPP inhibited both catalase and aconitase at high concentrations. However, the combination showed a synergistic effect on the aconitase activity of M. abscessus. The Ga(NO3)3/GaPP combination via intranasal administration showed significant antimicrobial activity in mice infected with M. abscessus. M. abscessus CFU from the lungs of the Ga(NO3)3/GaPP-treated mice was significantly less compared to that of nontreated or single Ga(III)-treated mice. These findings suggest that combinations of different Ga(III) compounds can synergistically target multiple iron/heme-utilizing mycobacterial enzymes. The results support the potential of combination Ga therapy for development against mycobacterial pathogens.
Collapse
Affiliation(s)
| | | | - Bradley E. Britigan
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center−Nebraska Western Iowa, Omaha, Nebraska 68105, United States
| | | |
Collapse
|
25
|
PPE51 Is Involved in the Uptake of Disaccharides by Mycobacterium tuberculosis. Cells 2020; 9:cells9030603. [PMID: 32138343 PMCID: PMC7140425 DOI: 10.3390/cells9030603] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 01/09/2023] Open
Abstract
We have recently found that selected thio-disaccharides possess bactericidal effects against Mycobacterium tuberculosis but not against Escherichia coli or Staphylococcus aureus. Here, we selected spontaneous mutants displaying resistance against the investigated thio-glycoside. According to next-generation sequencing, four of six analyzed mutants which were resistant to high concentrations of the tested chemical carried nonsynonymous mutations in the gene encoding the PPE51 protein. The complementation of these mutants with an intact ppe51 gene returned their sensitivity to the wild-type level. The uptake of tritiated thio-glycoside was significantly more abundant in wild-type Mycobacterium tuberculosis compared to the strain carrying the mutated ppe51 gene. The ppe51 mutations or CRISPR-Cas9-mediated downregulation of PPE51 expression affected the growth of mutant strains on minimal media supplemented with disaccharides (maltose or lactose) but not with glycerol or glucose as the sole carbon and energy source. Taking the above into account, we postulate that PPE51 participates in the uptake of disaccharides by tubercle bacilli.
Collapse
|
26
|
Zhang L, Hendrickson RC, Meikle V, Lefkowitz EJ, Ioerger TR, Niederweis M. Comprehensive analysis of iron utilization by Mycobacterium tuberculosis. PLoS Pathog 2020; 16:e1008337. [PMID: 32069330 PMCID: PMC7058343 DOI: 10.1371/journal.ppat.1008337] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/05/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022] Open
Abstract
Iron is essential for nearly all bacterial pathogens, including Mycobacterium tuberculosis (Mtb), but is severely limited in the human host. To meet its iron needs, Mtb secretes siderophores, small molecules with high affinity for iron, and takes up iron-loaded mycobactins (MBT) and carboxymycobactins (cMBT), from the environment. Mtb is also capable of utilizing heme and hemoglobin which contain more than 70% of the iron in the human body. However, many components of these iron acquisition pathways are still unknown. In this study, a high-density transposon mutagenesis coupled with deep sequencing (TnSeq) showed that Mtb exhibits nearly opposite requirements for 165 genes in the presence of heme and hemoglobin versus MBT and cMBT as iron sources. The ESX-3 secretion system was assessed as essential for siderophore-mediated iron uptake and, surprisingly, also for heme utilization by Mtb. Predictions derived from the TnSeq analysis were validated by growth experiments with isogenic Mtb mutants. These results showed that (i) the efflux pump MmpL5 plays a dominant role in siderophore secretion, (ii) the Rv2047c protein is essential for growth of Mtb in the presence of mycobactin, and (iii) the transcriptional repressor Zur is required for heme utilization by Mtb. The novel genetic determinants of iron utilization revealed in this study will stimulate further experiments in this important area of Mtb physiology.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - R. Curtis Hendrickson
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Virginia Meikle
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Elliot J. Lefkowitz
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Thomas R. Ioerger
- Department of Computer Science and Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Michael Niederweis
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
27
|
Ates LS. New insights into the mycobacterial PE and PPE proteins provide a framework for future research. Mol Microbiol 2020; 113:4-21. [PMID: 31661176 PMCID: PMC7028111 DOI: 10.1111/mmi.14409] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2019] [Indexed: 12/30/2022]
Abstract
The PE and PPE proteins of Mycobacterium tuberculosis have been studied with great interest since their discovery. Named after the conserved proline (P) and glutamic acid (E) residues in their N-terminal domains, these proteins are postulated to perform wide-ranging roles in virulence and immune modulation. However, technical challenges in studying these proteins and their encoding genes have hampered the elucidation of molecular mechanisms and leave many open questions regarding the biological functions mediated by these proteins. Here, I review the shared and unique characteristics of PE and PPE proteins from a molecular perspective linking this information to their functions in mycobacterial virulence. I discuss how the different subgroups (PE_PGRS, PPE-PPW, PPE-SVP and PPE-MPTR) are defined and why this classification of paramount importance to understand the PE and PPE proteins as individuals and or groups. The goal of this MicroReview is to summarize and structure the existing information on this gene family into a simplified framework of thinking about PE and PPE proteins and genes. Thereby, I hope to provide helpful starting points in studying these genes and proteins for researchers with different backgrounds. This has particular implications for the design and monitoring of novel vaccine candidates and in understanding the evolution of the M. tuberculosis complex.
Collapse
Affiliation(s)
- Louis S. Ates
- Department of Experimental ImmunologyAmsterdam Infection & Immunity InstituteAmsterdam UMCUniversity of AmsterdamMeibergdreef 9Amsterdamthe Netherlands
| |
Collapse
|
28
|
Matthews SJ, Pacholarz KJ, France AP, Jowitt TA, Hay S, Barran PE, Munro AW. MhuD from Mycobacterium tuberculosis: Probing a Dual Role in Heme Storage and Degradation. ACS Infect Dis 2019; 5:1855-1866. [PMID: 31480841 DOI: 10.1021/acsinfecdis.9b00181] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The Mycobacterium tuberculosis (Mtb) heme oxygenase MhuD liberates free iron by degrading heme to the linear tetrapyrrole mycobilin. The MhuD dimer binds up to two hemes within the active site of each monomer. Binding the first solvent-exposed heme allows heme degradation and releases free iron. Binding a second heme renders MhuD inactive, allowing heme storage. Native-mass spectrometry revealed little difference in binding affinity between solvent-exposed and solvent-protected hemes. Hence, diheme-MhuD is formed even when a large proportion of the MhuD population is in the apo form. Apomyoglobin heme transfer assays showed MhuD-diheme dissociation is far slower than monoheme dissociation at ∼0.12 min-1 and ∼0.25 s-1, respectively, indicating that MhuD has a strong affinity for diheme. MhuD has not evolved to preferentially occupy the monoheme form and, through formation of a diheme complex, it functions as part of a larger network to tightly regulate both heme and iron levels in Mtb.
Collapse
Affiliation(s)
- Sarah J. Matthews
- Manchester Institute of Biotechnology, School of Chemistry, Faculty of Science and Engineering, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Kamila J. Pacholarz
- Manchester Institute of Biotechnology, School of Chemistry, Faculty of Science and Engineering, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Aidan P. France
- Manchester Institute of Biotechnology, School of Chemistry, Faculty of Science and Engineering, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Thomas A. Jowitt
- The Biomolecular Analysis Facility, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Sam Hay
- Manchester Institute of Biotechnology, School of Chemistry, Faculty of Science and Engineering, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Perdita E. Barran
- Manchester Institute of Biotechnology, School of Chemistry, Faculty of Science and Engineering, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Andrew W. Munro
- Manchester Institute of Biotechnology, School of Chemistry, Faculty of Science and Engineering, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
29
|
Yu X, Feng J, Huang L, Gao H, Liu J, Bai S, Wu B, Xie J. Molecular Basis Underlying Host Immunity Subversion by Mycobacterium tuberculosis PE/PPE Family Molecules. DNA Cell Biol 2019; 38:1178-1187. [PMID: 31580738 DOI: 10.1089/dna.2019.4852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mycobacterium tuberculosis proline-glutamic acid (PE)/proline-proline-glutamic acid (PPE) family proteins, with >160 members, are crucial for virulence, cell wall, host cell fate, host Th1/Th2 balance, and CD8+ T cell recognition. Ca2+ signaling is involved in PE/PPE protein-mediated host-pathogen interaction. PE/PPE proteins also function in heme utilization and nitric oxide production. PE/PPE family proteins are intensively pursued as diagnosis biomarkers and vaccine components.
Collapse
Affiliation(s)
- Xiaowen Yu
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Jing Feng
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Lu Huang
- Department of Pathology, Xinqiao Hospital, Army Medical University, Chongqing, P.R. China
| | - Hongyan Gao
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Jinkun Liu
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Shutong Bai
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Bin Wu
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Jianping Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, P.R. China
| |
Collapse
|
30
|
Mitra A, Ko YH, Cingolani G, Niederweis M. Heme and hemoglobin utilization by Mycobacterium tuberculosis. Nat Commun 2019; 10:4260. [PMID: 31534126 PMCID: PMC6751184 DOI: 10.1038/s41467-019-12109-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/21/2019] [Indexed: 12/31/2022] Open
Abstract
Iron is essential for growth of Mycobacterium tuberculosis (Mtb), but most iron in the human body is stored in heme within hemoglobin. Here, we demonstrate that the substrate-binding protein DppA of the inner membrane Dpp transporter is required for heme and hemoglobin utilization by Mtb. The 1.27 Å crystal structure of DppA shows a tetrapeptide bound in the protein core and a large solvent-exposed crevice for heme binding. Mutation of arginine 179 in this cleft eliminates heme binding to DppA and prevents heme utilization by Mtb. The outer membrane proteins PPE36 and PPE62 are also required for heme and hemoglobin utilization, indicating that these pathways converge at the cell surface of Mtb. Albumin, the most abundant blood protein, binds heme specifically and bypasses the requirements for PPE36, PPE62 and Dpp. Thus, our study reveals albumin-dependent and -independent heme uptake pathways, highlighting the importance of iron acquisition from heme for Mtb.
Collapse
Affiliation(s)
- Avishek Mitra
- Department of Microbiology, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL, 35294, USA
| | - Ying-Hui Ko
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233S. 10th Street, Philadelphia, PA, 19107, USA
| | - Gino Cingolani
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233S. 10th Street, Philadelphia, PA, 19107, USA. .,Institute of Biomembranes and Bioenergetics, National Research Council, Via Amendola 165/A, 70126, Bari, Italy.
| | - Michael Niederweis
- Department of Microbiology, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL, 35294, USA.
| |
Collapse
|
31
|
Mashabela GT, de Wet TJ, Warner DF. Mycobacterium tuberculosis Metabolism. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0067-2019. [PMID: 31350832 PMCID: PMC10957194 DOI: 10.1128/microbiolspec.gpp3-0067-2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis is the cause of tuberculosis (TB), a disease which continues to overwhelm health systems in endemic regions despite the existence of effective combination chemotherapy and the widespread use of a neonatal anti-TB vaccine. For a professional pathogen, M. tuberculosis retains a surprisingly large proportion of the metabolic repertoire found in nonpathogenic mycobacteria with very different lifestyles. Moreover, evidence that additional functions were acquired during the early evolution of the M. tuberculosis complex suggests the organism has adapted (and augmented) the metabolic pathways of its environmental ancestor to persistence and propagation within its obligate human host. A better understanding of M. tuberculosis pathogenicity, however, requires the elucidation of metabolic functions under disease-relevant conditions, a challenge complicated by limited knowledge of the microenvironments occupied and nutrients accessed by bacilli during host infection, as well as the reliance in experimental mycobacteriology on a restricted number of experimental models with variable relevance to clinical disease. Here, we consider M. tuberculosis metabolism within the framework of an intimate host-pathogen coevolution. Focusing on recent advances in our understanding of mycobacterial metabolic function, we highlight unusual adaptations or departures from the better-characterized model intracellular pathogens. We also discuss the impact of these mycobacterial "innovations" on the susceptibility of M. tuberculosis to existing and experimental anti-TB drugs, as well as strategies for targeting metabolic pathways. Finally, we offer some perspectives on the key gaps in the current knowledge of fundamental mycobacterial metabolism and the lessons which might be learned from other systems.
Collapse
Affiliation(s)
- Gabriel T Mashabela
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Current address: Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, University of Stellenbosch, South Africa
| | - Timothy J de Wet
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, South Africa
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Wellcome Centre for Infectious Disease Research in Africa, University of Cape Town, South Africa
| |
Collapse
|
32
|
Iron Supplementation Therapy, A Friend and Foe of Mycobacterial Infections? Pharmaceuticals (Basel) 2019; 12:ph12020075. [PMID: 31108902 PMCID: PMC6630247 DOI: 10.3390/ph12020075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/21/2022] Open
Abstract
Iron is an essential element that is required for oxygen transfer, redox, and metabolic activities in mammals and bacteria. Mycobacteria, some of the most prevalent infectious agents in the world, require iron as growth factor. Mycobacterial-infected hosts set up a series of defense mechanisms, including systemic iron restriction and cellular iron distribution, whereas mycobacteria have developed sophisticated strategies to acquire iron from their hosts and to protect themselves from iron’s harmful effects. Therefore, it is assumed that host iron and iron-binding proteins, and natural or synthetic chelators would be keys targets to inhibit mycobacterial proliferation and may have a therapeutic potential. Beyond this hypothesis, recent evidence indicates a host protective effect of iron against mycobacterial infections likely through promoting remodeled immune response. In this review, we discuss experimental procedures and clinical observations that highlight the role of the immune response against mycobacteria under various iron availability conditions. In addition, we discuss the clinical relevance of our knowledge regarding host susceptibility to mycobacteria in the context of iron availability and suggest future directions for research on the relationship between host iron and the immune response and the use of iron as a therapeutic agent.
Collapse
|