1
|
Rangaraj S, Agarwal A, Banerjee S. Bird's Eye View on Mycobacterium tuberculosis-HIV Coinfection: Understanding the Molecular Synergism, Challenges, and New Approaches to Therapeutics. ACS Infect Dis 2025. [PMID: 40229972 DOI: 10.1021/acsinfecdis.4c00870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M.tb), is the most common secondary infection in the Human Immunodeficiency Virus (HIV) infected population, accounting for more than one-fourth of deaths in people living with HIV (PLWH). Reciprocally, HIV infection increases the susceptibility to primary TB or reactivation of latent TB by several folds. The synergistic interactions between M.tb and HIV not only potentiate their deleterious impact but also complicate the clinical management of both the diseases. M.tb-HIV coinfected patients have a high risk of failure of accurate diagnosis, treatment inefficiency for both TB and HIV, concurrent nontuberculous mycobacterial infections, other comorbidities such as diabetes mellitus, severe cytotoxicity due to drug overburden, and immune reconstitution inflammatory syndrome (IRIS). The need of the hour is to understand M.tb-HIV coinfection biology and their collective impact on the host immunocompetence and to think of out-of-the-box treatment perspectives, including host-directed therapy under the rising view of homeostatic medicines. This review aims to highlight the molecular players, both from the pathogens and host, that facilitate the synergistic interactions and host-associated proteins/enzymes regulating immunometabolism, underlining potential targets for designing and screening chemical inhibitors to reduce the burden of both pathogens concomitantly during M.tb-HIV coinfection. To appreciate the necessity of revisiting therapeutic approaches and research priorities, we provide a glimpse of anti-TB and antiretroviral drug-drug interactions, project the gaps in our understanding of coinfection biology, and also enlist some key research initiatives that will help us deal with the synergistic epidemic of M.tb-HIV coinfection.
Collapse
Affiliation(s)
- Siranjeevi Rangaraj
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Anushka Agarwal
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Sharmistha Banerjee
- Laboratory of Molecular Pathogenesis, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| |
Collapse
|
2
|
Dawi J, Affa S, Kafaja K, Misakyan Y, Kades S, Dayal S, Fardeheb S, Narasimhan A, Tumanyan K, Venketaraman V. The Role of Ferroptosis and Cuproptosis in Tuberculosis Pathogenesis: Implications for Therapeutic Strategies. Curr Issues Mol Biol 2025; 47:99. [PMID: 39996820 PMCID: PMC11853893 DOI: 10.3390/cimb47020099] [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: 12/31/2024] [Revised: 01/28/2025] [Accepted: 02/01/2025] [Indexed: 02/26/2025] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis (M.tb) remains a global health crisis, with over 10 million people affected annually. Despite advancements in treatment, M.tb has developed mechanisms to evade host immune responses, complicating efforts to eradicate the disease. Two emerging cell death pathways, ferroptosis and cuproptosis, have been linked to TB pathogenesis. Ferroptosis, an iron-dependent form of cell death, is driven by lipid peroxidation and reactive oxygen species (ROS) accumulation. This process can limit M.tb replication by depleting intracellular iron and inducing macrophage necrosis. However, excessive ferroptosis may lead to tissue damage and aid bacterial dissemination. Cuproptosis, triggered by copper accumulation, disrupts mitochondrial metabolism, leading to protein aggregation and cell death. M.tb exploits both iron and copper metabolism to survive within macrophages, manipulating these processes to resist oxidative stress and immune responses. This review examines the roles of ferroptosis and cuproptosis in TB, discussing how M.tb manipulates these pathways for survival. While therapeutic strategies targeting these processes, such as ferroptosis inducers (Erastin, RSL3) and inhibitors (Ferrostatin-1) and copper ionophores (Disulfiram, Elesclomol) and chelators, show promise, the limited understanding of these pathways and potential off-target effects remains a significant challenge. Further exploration of these pathways may provide insights into the development of targeted therapies aimed at controlling M.tb infection while minimizing host tissue damage. By elucidating the complex interactions between ferroptosis, cuproptosis, and TB, future therapies could better address bacterial resistance and improve clinical outcomes.
Collapse
Affiliation(s)
- John Dawi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (J.D.); (K.K.); (Y.M.); (S.K.); (S.D.); (S.F.); (A.N.)
| | - Stephen Affa
- Department of Chemistry, Physics, and Engineering, Los Angeles Valley College, Valley Glen, CA 91401, USA;
| | - Kevin Kafaja
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (J.D.); (K.K.); (Y.M.); (S.K.); (S.D.); (S.F.); (A.N.)
| | - Yura Misakyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (J.D.); (K.K.); (Y.M.); (S.K.); (S.D.); (S.F.); (A.N.)
| | - Samuel Kades
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (J.D.); (K.K.); (Y.M.); (S.K.); (S.D.); (S.F.); (A.N.)
| | - Surbi Dayal
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (J.D.); (K.K.); (Y.M.); (S.K.); (S.D.); (S.F.); (A.N.)
| | - Sabrina Fardeheb
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (J.D.); (K.K.); (Y.M.); (S.K.); (S.D.); (S.F.); (A.N.)
| | - Ananya Narasimhan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (J.D.); (K.K.); (Y.M.); (S.K.); (S.D.); (S.F.); (A.N.)
| | - Kevin Tumanyan
- College of Podiatric Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (J.D.); (K.K.); (Y.M.); (S.K.); (S.D.); (S.F.); (A.N.)
| |
Collapse
|
3
|
Tang S, Qiu Z, Liu F, Li C, Peng G. Lipopolysaccharide aggravating anaphylactoid reactions caused by traditional Chinese Medicine injections via p38/ERK/NF-κB signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118946. [PMID: 39419299 DOI: 10.1016/j.jep.2024.118946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/12/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Currently, adverse reactions limit the development of traditional Chinese medicine injections (TCMI), and severe anaphylactoid shock is one of the serious adverse reactions, which presents a significant challenge. The presence of abnormal inflammatory mediators before the administration of TCMI will most likely result in severe anaphylactoid reactions. Not only that, the lack of clinically relevant safety evaluations impedes the widespread use of TCMI, and there is an urgent need for studies to reveal the mechanisms of anaphylactoid shock caused by TCMI. AIM OF THE STUDY To investigate the effects and underlying mechanisms of lipopolysaccharide (LPS)-induced inflammation, which aggravates anaphylactoid reactions caused by TCMI, utilizing a guinea pig model. METHODS The dose and duration of LPS administration and different doses of compound 48/80 (C48/80) were examined by using guinea pigs as a model. Shuanghuanglian (SHLI) and Qingkailing (QKLI) injections, these two representative TCMI, were used for validation. The plasma biochemical indices, including histamine, 5-hydroxytryptamine, tumor necrosis factor-α, interleukin 6, immunoglobulin E, C5a, tryptase, and platelet activating factor, as well as the pathological characteristics of the lung, were analyzed. Futhermore, plasma metabolomics was employed to reveal changes in metabolic pathways in vivo when inflammation co-exists with TCMI. In addition, Western blot analysis was conducted to assess the expression of critical signaling pathways. RESULTS Stimulation with 2 mg/kg of LPS for 12 h induced inflammatory responses in the guinea pig model. C48/80 (3.0 mg/kg) in combination with LPS resulted in an increase in anaphylactoid-related indicators in the plasma. High doses of SHLI and QKLI aggravated plasma indices and lung histological injury caused by LPS-induced inflammation. A total of 36 and 63 differential metabolites were significantly altered after LPS stimulation in the SHLI-and QKLI-treated guinea pig groups, respectively. The associated metabolic pathways include central carbon metabolism in cancer, the tricarboxylic acid cycle, glyoxylate and dicarboxylate metabolism. The p38/ERK/NF-κB signal pathway may be significantly affected by TCMI in vivo after LPS stimulation. CONCLUSION LPS-induced inflammation aggravated anaphylactoid reactions caused by SHLI and QKLI, with a correlation to dosage. After the presence of LPS, the administration of TCMI interferes with the immune response by over-activating the p38/ERK/NF-κB signaling pathway. This activation leads to an excessive release of inflammatory factors and anaphylactoid mediators. These results present a new direction for mitigating adverse clinical reactions associated with TCMI.
Collapse
Affiliation(s)
- Shuwan Tang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zichao Qiu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fangmei Liu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cunyu Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China.
| | - Guoping Peng
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China.
| |
Collapse
|
4
|
Jain M, Vyas R. Unveiling the silent defenders: mycobacterial stress sensors at the forefront to combat tuberculosis. Crit Rev Biotechnol 2025:1-19. [PMID: 39880585 DOI: 10.1080/07388551.2024.2449367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/12/2024] [Accepted: 09/14/2024] [Indexed: 01/31/2025]
Abstract
The global escalation in tuberculosis (TB) cases accompanied by the emergence of multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains of Mycobacterium tuberculosis (M.tb) emphasizes the critical requirement for novel potent drugs. The M.tb demonstrates extraordinary adaptability, thriving in diverse conditions, and always finds itself in win-win situations regardless of whether the environment is favorable or unfavorable; no matter the magnitude of the challenge, it can endure and survive. This review aims to uncover the role of multiple stress sensors of M.tb that assist bacteria in remaining viable within the host for years against various physiological stresses offered by the host. M.tb is an exceptionally triumphant pathogen, primarily due to its adeptness in developing defense mechanisms against stressful situations. The recent advances emphasize the significance of M.tb stress sensors, including chaperones, proteases, transcription factors, riboswitches, inteins, etc., employed in responding to a spectrum of physiological stresses imposed by the host, encompassing surface stress, host immune responses, osmotic stress, oxidative and nitrosative stresses, cell envelope stress, environmental stress, reductive stress, and drug pressure. These sensors act as silent defenders orchestrating adaptive strategies, with limited comprehensive information in current literature, necessitating a focused review. The M.tb strategies utilizing these stress sensors to mitigate the impact of traumatic conditions demand attention to neutralize this pathogen effectively. Moreover, the intricacies of these stress sensors provide potential targets to design an effective TB drug using structure-based drug design against this formidable global health threat.
Collapse
Affiliation(s)
- Manya Jain
- Department of Life Sciences, Shiv Nadar Institution of Eminence (Deemed to be University), Gautam Buddha Nagar, Uttar Pradesh, India
| | - Rajan Vyas
- Department of Life Sciences, Shiv Nadar Institution of Eminence (Deemed to be University), Gautam Buddha Nagar, Uttar Pradesh, India
| |
Collapse
|
5
|
Wang L, Wang Q, Liu Y, Chen Y, Bao S, Zhang X, Wang C. Proteomics Reveals the Response Mechanism of Embryonic Bovine Lung Cells to Mycoplasma bovis Infection. Int J Mol Sci 2025; 26:823. [PMID: 39859536 PMCID: PMC11765741 DOI: 10.3390/ijms26020823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Mycoplasma bovis (M. bovis) has caused huge economic losses to the cattle industry. The interaction between M. bovis and host cells is elucidated by screening and identifying the target protein of M. bovis adhesin on the surface of the host cell membrane. However, the response mechanism of embryonic bovine lung (EBL) cells to M. bovis infection is not yet fully understood. Additionally, it is necessary to further explore whether infection with M. bovis induces oxidative stress and mitochondrial damage in EBL cells. In this study, oxidation reaction, mitochondrial membrane potential, mitochondrial structure, and apoptosis ability of EBL cells infected with M. bovis were assessed at different times (12, 24, 48 h post-infection; hpi). Then, the differential proteomic analysis of M. bovis-infected EBL cells at 12 h and 24 h was performed with uninfected cells as the control. The results showed that M. bovis infection reduced the antioxidant capacity of EBL cells, increased ROS levels, and decreased mitochondrial membrane potential. The mitochondrial membrane of EBL cells was damaged, and the ridge arrangement was disordered after infection by transmission electron microscopy. With the increase in infection time, the mitochondrial matrix partially dissolved and spilled. The apoptosis rate of EBL cells increased with the increase in infection time of M. bovis. Furthermore, proteomic analysis identified 268 and 2061 differentially expressed proteins (DEPs) at 12 hpi and 24 hpi, respectively, compared with the uninfected cells. According to GO analysis, these DEPs were involved in the mitosis and negative regulation of cell growth. Additionally, the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis indicated the following pathways were linked to mitochondrial damage or cell growth regulation, including glycolysis/gluconeogenesis, pentose phosphate pathway, oxidative phosphorylation, AMPK, cGMP-PKG, cAMP, calcium, Wnt, Phospholipase D, apoptosis, MAPK, cell cycle, Ras, PI3K-Akt, mTOR, HIF-1. PPI results indicated that YWHAZ, PIK3CA, HSP90AB1, RAP1A, TXN, RAF1, MAPK1, PKM, PGK1, and GAPDH might be involved in mitochondrial pathway apoptosis induced by M. bovis infection. This study offers helpful data toward understanding the response of mitochondria of EBL cells to M. bovis infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoli Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (L.W.); (Q.W.); (Y.L.); (Y.C.); (S.B.)
| | - Chuan Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (L.W.); (Q.W.); (Y.L.); (Y.C.); (S.B.)
| |
Collapse
|
6
|
Patel A, Nguyen L, Shea C, Singh S, Venketaraman V. The Role of mTOR in Mycobacterium tuberculosis Infection. Biomedicines 2024; 12:2238. [PMID: 39457551 PMCID: PMC11505195 DOI: 10.3390/biomedicines12102238] [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: 08/12/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Mycobacterium tuberculosis (M. tb) is a pathogen that causes tuberculosis (TB), an extremely infectious disease which is responsible for millions of deaths worldwide. The severity of this pathogen is further amplified with the emergence of multidrug-resistant strains that are becoming more prevalent at an alarming rate, and novel treatments are needed. Methods: In this paper, we discuss the pathology M. tb infection. We review the literature on the role that mTOR plays in autophagy and the immune system as well as its impact on M. tb infection. Lastly, we discuss the current therapies targeting mTOR and potential routes to explore for future treatments. Results: The mTOR protein acts as a negative regulator of the autophagy pathway and presents as a potent target to establish new treatments for TB. M. tb survival is affected by mTOR, the PI3K/mTOR/AKT pathway, and autophagy. M. tb evades destruction by manipulating host cellular mechanisms, which increases resistance and complicates treatment. Conclusions: Targeting mTOR can enhance autophagy and increase M. tb clearance. Existing drugs such as everolimus, rapamycin + CC214-2, and bazedoxifene are all being currently studied for effectiveness and show positive results. Alternative therapies, including Chinese herbs, baicalin, BTLA, glutathione, and precision medicine can modulate the PI3K/mTOR/AKT pathway and the host's immune response, resulting in increased M. tb clearance, and these may be the future treatments for M. tb infection.
Collapse
Affiliation(s)
| | | | | | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (A.P.); (L.N.); (C.S.); (S.S.)
| |
Collapse
|
7
|
Priyanka, Sharma S, Varma-Basil M, Sharma M. C-terminal region of Rv1039c (PPE15) protein of Mycobacterium tuberculosis targets host mitochondria to induce macrophage apoptosis. Apoptosis 2024; 29:1757-1779. [PMID: 38615303 DOI: 10.1007/s10495-024-01965-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/15/2024]
Abstract
Mycobacterium tuberculosis (Mtb) genome possesses a unique family called Proline-Glutamate/Proline-Proline-Glutamate (PE/PPE) gene family, exclusive to pathogenic mycobacterium. Some of these proteins are known to play role in virulence and immune response modulation, but many are still uncharacterized. This study investigated the role of C-terminal region of Rv1039c (PPE15) in inducing mitochondrial perturbations and macrophage apoptosis. Our in-silico studies revealed the disordered, coiled, and hydrophobic C-terminal region in Rv1039c has similarity with C-terminal of mitochondria-targeting pro-apoptotic host proteins. Wild type Rv1039c and C-terminal deleted Rv1039c (Rv1039c-/-Cterm) recombinant proteins were purified and their M. smegmatis knock-in strains were constructed which were used for in-vitro experiments. Confocal microscopy showed localization of Rv1039c to mitochondria of PMA-differentiated THP1 macrophages; and reduced mitochondrial membrane depolarization and production of mitochondrial superoxides were observed in response to Rv1039c-/-Cterm in comparison to full-length Rv1039c. The C-terminal region of Rv1039c was found to activate caspases 3, 7 and 9 along with upregulated expression of pro-apoptotic genes like Bax and Bim. Rv1039c-/-Cterm also reduced the Cytochrome-C release from the mitochondria and the expression of AnnexinV/PI positive and TUNEL positive cells as compared to Rv1039c. Additionally, Rv1039c was observed to upregulate the TLR4-NF-κB-TNF-α signalling whereas the same was downregulated in response to Rv1039c-/-Cterm. These findings suggested that the C-terminal region of Rv1039c is a molecular mimic of pro-apoptotic host proteins which induce mitochondria-dependent macrophage apoptosis and evoke host immune response. These observations enhance our understanding about the role of PE/PPE proteins at host-pathogen interface.
Collapse
Affiliation(s)
- Priyanka
- DSKC BioDiscovery Laboratory, Department of Zoology, Miranda House, University of Delhi, Delhi, India
| | - Sadhna Sharma
- DSKC BioDiscovery Laboratory, Department of Zoology, Miranda House, University of Delhi, Delhi, India
| | - Mandira Varma-Basil
- Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Monika Sharma
- DSKC BioDiscovery Laboratory, Department of Zoology, Miranda House, University of Delhi, Delhi, India.
| |
Collapse
|
8
|
Medikonda J, Wankar N, Asalla S, Raja SO, Yandrapally S, Jindal H, Agarwal A, Pant C, Kalivendi SV, Kumar Dubey H, Mohareer K, Gulyani A, Banerjee S. Rv0547c, a functional oxidoreductase, supports Mycobacterium tuberculosis persistence by reprogramming host mitochondrial fatty acid metabolism. Mitochondrion 2024; 78:101931. [PMID: 38986924 DOI: 10.1016/j.mito.2024.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 07/12/2024]
Abstract
Mycobacterium tuberculosis (Mtb) successfully thrives in the host by adjusting its metabolism and manipulating the host environment. In this study, we investigated the role of Rv0547c, a protein that carries mitochondria-targeting sequence (MTS), in mycobacterial persistence. We show that Rv0547c is a functional oxidoreductase that targets host-cell mitochondria. Interestingly, the localization of Rv0547c to mitochondria was independent of the predicted MTS but depended on specific arginine residues at the N- and C-terminals. As compared to the mitochondria-localization defective mutant, Rv0547c-2SDM, wild-type Rv0547c increased mitochondrial membrane fluidity and spare respiratory capacity. To comprehend the possible reason, comparative lipidomics was performed that revealed a reduced variability of long-chain and very long-chain fatty acids as well as altered levels of phosphatidylcholine and phosphatidylinositol class of lipids upon expression of Rv0547c, explaining the increased membrane fluidity. Additionally, the over representation of propionate metabolism and β-oxidation intermediates in Rv0547c-targeted mitochondrial fractions indicated altered fatty acid metabolism, which corroborated with changes in oxygen consumption rate (OCR) upon etomoxir treatment in HEK293T cells transiently expressing Rv0547c, resulting in enhanced mitochondrial fatty acid oxidation capacity. Furthermore, Mycobacterium smegmatis over expressing Rv0547c showed increased persistence during infection of THP-1 macrophages, which correlated with its increased expression in Mtb during oxidative and nutrient starvation stresses. This study identified for the first time an Mtb protein that alters mitochondrial metabolism and aids in survival in host macrophages by altering fatty acid metabolism to its benefit and, at the same time increases mitochondrial spare respiratory capacity to mitigate infection stresses and maintain cell viability.
Collapse
Affiliation(s)
- Jayashankar Medikonda
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Nandini Wankar
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Suman Asalla
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Sufi O Raja
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Sriram Yandrapally
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Haneesh Jindal
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Anushka Agarwal
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Chitrakshi Pant
- CSIR-Indian Institute of Chemical Technology (IICT), Uppal Road, Hyderabad, India 500007
| | - Shasi V Kalivendi
- CSIR-Indian Institute of Chemical Technology (IICT), Uppal Road, Hyderabad, India 500007
| | - Harish Kumar Dubey
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Krishnaveni Mohareer
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Akash Gulyani
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046
| | - Sharmistha Banerjee
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India 500046.
| |
Collapse
|
9
|
Priyanka, Sharma S, Sharma M. Role of PE/PPE proteins of Mycobacterium tuberculosis in triad of host mitochondria, oxidative stress and cell death. Microb Pathog 2024; 193:106757. [PMID: 38908454 DOI: 10.1016/j.micpath.2024.106757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The PE and PPE family proteins of Mycobacterium tuberculosis (Mtb) is exclusively found in pathogenic Mycobacterium species, comprising approximately 8-10 % of the Mtb genome. These emerging virulent factors have been observed to play pivotal roles in Mtb pathogenesis and immune evasion through various strategies. These immunogenic proteins are known to modulate the host immune response and cell-death pathways by targeting the powerhouse of the cell, the mitochondria to support Mtb survival. In this article, we are focused on how PE/PPE family proteins target host mitochondria to induce mitochondrial perturbations, modulate the levels of cellular ROS (Reactive oxygen species) and control cell death pathways. We observed that the time of expression of these proteins at different stages of infection is crucial for elucidating their impact on the cell death pathways and eventually on the outcome of infection. This article focuses on understanding the contributions of the PE/PPE proteins by unravelling the triad of host mitochondria, oxidative stress and cell death pathways that facilitate the Mtb persistence. Understanding the role of these proteins in host cellular pathways and the intricate mechanisms paves the way for the development of novel therapeutic strategies to combat TB infections.
Collapse
Affiliation(s)
- Priyanka
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Sadhna Sharma
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Monika Sharma
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
10
|
Vu A, Glassman I, Campbell G, Yeganyan S, Nguyen J, Shin A, Venketaraman V. Host Cell Death and Modulation of Immune Response against Mycobacterium tuberculosis Infection. Int J Mol Sci 2024; 25:6255. [PMID: 38892443 PMCID: PMC11172987 DOI: 10.3390/ijms25116255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a prevalent infectious disease affecting populations worldwide. A classic trait of TB pathology is the formation of granulomas, which wall off the pathogen, via the innate and adaptive immune systems. Some key players involved include tumor necrosis factor-alpha (TNF-α), foamy macrophages, type I interferons (IFNs), and reactive oxygen species, which may also show overlap with cell death pathways. Additionally, host cell death is a primary method for combating and controlling Mtb within the body, a process which is influenced by both host and bacterial factors. These cell death modalities have distinct molecular mechanisms and pathways. Programmed cell death (PCD), encompassing apoptosis and autophagy, typically confers a protective response against Mtb by containing the bacteria within dead macrophages, facilitating their phagocytosis by uninfected or neighboring cells, whereas necrotic cell death benefits the pathogen, leading to the release of bacteria extracellularly. Apoptosis is triggered via intrinsic and extrinsic caspase-dependent pathways as well as caspase-independent pathways. Necrosis is induced via various pathways, including necroptosis, pyroptosis, and ferroptosis. Given the pivotal role of host cell death pathways in host defense against Mtb, therapeutic agents targeting cell death signaling have been investigated for TB treatment. This review provides an overview of the diverse mechanisms underlying Mtb-induced host cell death, examining their implications for host immunity. Furthermore, it discusses the potential of targeting host cell death pathways as therapeutic and preventive strategies against Mtb infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (G.C.); (A.S.)
| |
Collapse
|
11
|
Zhang L, Cai M, Su B, Ma Y, Zhang Y. Mitochondrial Metabolism in Alveolar Macrophages of Patients Infected with HIV, Tuberculosis, and HIV/Tuberculosis. AIDS Res Hum Retroviruses 2024; 40:148-157. [PMID: 37885217 DOI: 10.1089/aid.2023.0064] [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] [Indexed: 10/28/2023] Open
Abstract
Tuberculosis (TB) is one of the most common opportunistic infections and is a leading cause of mortality in patients with HIV and AIDS. HIV infection causes serious defects in the host immune system and increases the risk of active TB. TB infection promotes HIV replication and aggravates host damage in patients with HIV/AIDS. Alveolar macrophages (AMs) are essential immune cells during TB and HIV infections. AMs undergo a shift in mitochondrial metabolism during TB or HIV infection, that is, metabolic reprogramming, allowing them to act in the form of classical activated macrophages (M1) and alternative activated macrophages (M2) at different stages of infection. We reviewed the alterations in the mitochondrial energy metabolism of AMs in patients with HIV, TB, and HIV/TB to provide ideas for further research on the role of metabolic reprogramming by AMs in the pathogeneses of HIV, TB, and HIV/TB coinfection.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Miaotian Cai
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| |
Collapse
|
12
|
Dawrs SN, Virdi R, Islam MN, Hasan NA, Norton GJ, Crooks JL, Parr J, Heinz D, Cool CD, Belisle JT, Chan ED, Honda JR. Immunological and metabolic characterization of environmental Mycobacterium chimaera infection in a murine model. Microbes Infect 2023; 25:105184. [PMID: 37453489 DOI: 10.1016/j.micinf.2023.105184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 06/22/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
Mycobacterium chimaera causes pulmonary disease, but little is known of gradations in isolate virulence. Previously, 17 M. chimaera isolates were screened for survival in THP1 macrophages. "M. chimaera 1" was categorized as "more virulent" because it showed the greatest survival in macrophages, whereas "M. chimaera 2" was categorized as "less virulent" with reduced survival. Herein, we infected C3HeB/FeJ mice to compare the in vivo immune responses to M. chimaera 1 and 2. Unlike macrophages, significantly lower M. chimaera 1 counts were recovered from mouse lung tissue and BAL cells with less lung histopathologic changes compared to M. chimaera 2. Compared to M. chimaera 2, significantly more IL-1β, IL-6, and TNFα was produced early after M. chimaera 1 infection. LC-MS metabolomics analyses of BAL fluid revealed divergence in sphingolipid, phospholipid metabolism between M. chimaera 1 versus M. chimaera 2 mice. From pan-GWAS analyses, virulence and organizing DNA/molecular structure genes were associated with more virulent M. chimaera isolates. Vigorous lung-specific immune responses to M. chimaera 1 may influence effective bacterial control, but for a different isolate M. chimaera 2, subvert immune control. Continued studies of the gradations in virulence among the same NTM species will advance our understanding of NTM pathogenesis.
Collapse
Affiliation(s)
- Stephanie N Dawrs
- Center for Genes, Environment, and Health, National Jewish Health, CO, United States.
| | - Ravleen Virdi
- Center for Genes, Environment, and Health, National Jewish Health, CO, United States.
| | - M Nurul Islam
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Nabeeh A Hasan
- Center for Genes, Environment, and Health, National Jewish Health, CO, United States
| | - Grant J Norton
- Center for Genes, Environment, and Health, National Jewish Health, CO, United States.
| | - James L Crooks
- Division of Biostatistics and Bioinformatics, National Jewish Health, Colorado, United States; Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, United States
| | - Jane Parr
- Division of Pathology and Department of Medicine, National Jewish Health, CO, United States
| | - David Heinz
- Division of Pathology and Department of Medicine, National Jewish Health, CO, United States
| | - Carlyne D Cool
- Division of Pathology and Department of Medicine, National Jewish Health, CO, United States; Department of Pathology, University of Colorado Anschutz Medical Campus, CO, United States
| | - John T Belisle
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Edward D Chan
- Department of Medicine and Academic Affairs, National Jewish Health, Denver, CO, United States; Division of Pulmonary Science and Critical Care Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Denver, CO, United States
| | - Jennifer R Honda
- Department of Cellular and Molecular Biology, School of Medicine, University of Texas Health Science Center at Tyler, TX, United States.
| |
Collapse
|
13
|
Yandrapally S, Agarwal A, Chatterjee A, Sarkar S, Mohareer K, Banerjee S. Mycobacterium tuberculosis EspR modulates Th1-Th2 shift by transcriptionally regulating IL-4, steering increased mycobacterial persistence and HIV propagation during co-infection. Front Immunol 2023; 14:1276817. [PMID: 37928551 PMCID: PMC10621737 DOI: 10.3389/fimmu.2023.1276817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) and HIV are known to mutually support each other during co-infection by multiple mechanisms. This synergistic influence could be either by direct interactions or indirectly through secreted host or pathogen factors that work in trans. Mtb secretes several virulence factors to modulate the host cellular environment for its persistence and escaping cell-intrinsic immune responses. We hypothesized that secreted Mtb transcription factors that target the host nucleus can directly interact with host DNA element(s) or HIV LTR during co-infection, thereby modulating immune gene expression, or driving HIV transcription, helping the synergistic existence of Mtb and HIV. Here, we show that the Mtb-secreted protein, EspR, a transcription regulator, increased mycobacterial persistence and HIV propagation during co-infection. Mechanistically, EspR localizes to the nucleus of the host cells during infection, binds to its putative cognate motif on the promoter region of the host IL-4 gene, activating IL-4 gene expression, causing high IL-4 titers that induce a Th2-type microenvironment, shifting the macrophage polarization to an M2 state as evident from CD206 dominant population over CD64. This compromised the clearance of the intracellular mycobacteria and enhanced HIV propagation. It was interesting to note that EspR did not bind to HIV LTR, although its transient expression increased viral propagation. This is the first report of an Mtb transcription factor directly regulating a host cytokine gene. This augments our understanding of the evolution of Mtb immune evasion strategies and unveils how Mtb aggravates comorbidities, such as HIV co-infection, by modulating the immune microenvironment.
Collapse
|
14
|
Lee J, Lee SA, Son SH, Choi JA, Nguyen TD, Kim J, Son D, Song CH. Impaired mitophagy induces antimicrobial responses in macrophages infected with Mycobacterium tuberculosis. Cell Biosci 2023; 13:158. [PMID: 37649112 PMCID: PMC10470153 DOI: 10.1186/s13578-023-01107-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 08/18/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Mitophagy, mitochondrial selective autophagy, plays a pivotal role in the maintenance of cellular homeostasis in response to cellular stress. However, the role of mitophagy in macrophages during infection has not been elucidated. To determine whether mitophagy regulates intracellular pathogen survival, macrophages were infected with Mycobacterium tuberculosis (Mtb), an intracellular bacterium. RESULTS We showed that Mtb-infected macrophages induced mitophagy through BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3) activation. In contrast, BNIP3-deficient macrophages failed to induce mitophagy, resulting in reduced mitochondrial membrane potential in response to Mtb infection. Moreover, the accumulation of damaged mitochondria due to BNIP3 deficiency generated higher levels of mitochondrial reactive oxygen species (mROS) compared to the control, suppressing the intracellular survival of Mtb. We observed that siBNIP3 suppressed intracellular Mtb in mice lungs. CONCLUSION We found that BNIP3 plays a critical role in the regulation of mitophagy during Mtb infection. The inhibition of mitophagy suppresses Mtb growth in macrophages through increased mROS production. Therefore, BNIP3 might be a novel therapeutic target for tuberculosis treatment.
Collapse
Affiliation(s)
- Junghwan Lee
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, 34134, South Korea
| | - Seong-Ahn Lee
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Sang-Hun Son
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Ji-Ae Choi
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, 34134, South Korea
| | - Tam Doan Nguyen
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Jaewhan Kim
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Doyi Son
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Chang-Hwa Song
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea.
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea.
- Translational Immunology Institute, Chungnam National University, Daejeon, 34134, South Korea.
| |
Collapse
|
15
|
Medha, Priyanka, Sharma S, Sharma M. PE_PGRS45 (Rv2615c) protein of Mycobacterium tuberculosis perturbs mitochondria of macrophages. Immunol Cell Biol 2023. [PMID: 37565603 DOI: 10.1111/imcb.12677] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/23/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023]
Abstract
The PE_PGRS proteins have coevolved with the antigenic ESX-V secretory system and are abundant in pathogenic Mycobacterium. Only a few PE_PGRS proteins have been characterized, and research suggests their role in organelle targeting, cell death pathways, calcium (Ca2+ ) homeostasis and disease pathogenesis. The PE_PGRS45 (Rv2615c) protein was predicted to contain mitochondria targeting sequences by in silico evaluation. Therefore, we investigated the targeting of the Rv2615c protein to host mitochondria and its effect on mitochondrial functions. In vitro experiments showed the Rv2615c protein colocalized with the mitochondria and led to morphological mitochondrial perturbations. Recombinant Rv2615c was observed to cause increased levels of intracellular reactive oxygen species and the adenosine diphosphate-to-adenosine triphosphate ratio. The Rv2615c protein also induced mitochondrial membrane depolarization and the generation of mitochondrial superoxide. We observed the release of cytochrome C into the cytoplasm and increased expression of proapoptotic genes Bax and Bim with no significant change in anti-apoptotic Bcl2 in Rv2615c-stimulated THP1 macrophages. Ca2+ is a key signaling molecule in tuberculosis pathogenesis, modulating host cell responses. As reported for other PE_PGRS proteins, Rv2615c also has Ca2+ -binding motifs and thus can modulate calcium homeostasis in the host. We also observed a high level of Ca2+ influx in THP1 macrophages stimulated with Rv2615c. Based on these findings, we suggest that Rv2615c may be an effector protein that could contribute to disease pathogenesis by targeting host mitochondria.
Collapse
Affiliation(s)
- Medha
- DSKC BioDiscovery Laboratory, Department of Zoology, Miranda House, University of Delhi, Delhi, India
| | - Priyanka
- DSKC BioDiscovery Laboratory, Department of Zoology, Miranda House, University of Delhi, Delhi, India
| | - Sadhna Sharma
- DSKC BioDiscovery Laboratory, Department of Zoology, Miranda House, University of Delhi, Delhi, India
| | - Monika Sharma
- DSKC BioDiscovery Laboratory, Department of Zoology, Miranda House, University of Delhi, Delhi, India
| |
Collapse
|
16
|
Fan M, Shi Y, Zhao J, Li L. Cancer stem cell fate determination: mito-nuclear communication. Cell Commun Signal 2023; 21:159. [PMID: 37370081 PMCID: PMC10294499 DOI: 10.1186/s12964-023-01160-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/06/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer stem cells (CSCs) are considered to be responsible for tumor recurrence and metastasis. Therefore, clarification of the mechanisms involved in CSC stemness maintenance and cell fate determination would provide a new strategy for cancer therapy. Unregulated cellular energetics has been accepted as one of the hallmarks of cancer cells, but recent studies have revealed that mitochondrial metabolism can also actively determine CSC fate by affecting nuclear stemness gene expression. Herein, from the perspective of mito-nuclear communication, we review recent progress on the influence of mitochondria on CSC potential from four aspects: metabolism, dynamics, mitochondrial homeostasis, and reactive oxygen species (ROS). Video Abstract.
Collapse
Affiliation(s)
- Mengchen Fan
- School of Basic Medical Sciences, Medical College of Yan’an University, Yanan, 716000 China
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi’an, 710032 China
| | - Ying Shi
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi’an, 710032 China
| | - Jumei Zhao
- School of Basic Medical Sciences, Medical College of Yan’an University, Yanan, 716000 China
| | - Ling Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi’an, 710032 China
| |
Collapse
|
17
|
Martin M, deVisch A, Boudehen YM, Barthe P, Gutierrez C, Turapov O, Aydogan T, Heriaud L, Gracy J, Neyrolles O, Mukamolova GV, Letourneur F, Cohen-Gonsaud M. A Mycobacterium tuberculosis Effector Targets Mitochondrion, Controls Energy Metabolism, and Limits Cytochrome c Exit. Microbiol Spectr 2023; 11:e0106623. [PMID: 37036353 PMCID: PMC10269737 DOI: 10.1128/spectrum.01066-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 04/11/2023] Open
Abstract
Host metabolism reprogramming is a key feature of Mycobacterium tuberculosis (Mtb) infection that enables the survival of this pathogen within phagocytic cells and modulates the immune response facilitating the spread of the tuberculosis disease. Here, we demonstrate that a previously uncharacterized secreted protein from Mtb, Rv1813c, manipulates the host metabolism by targeting mitochondria. When expressed in eukaryotic cells, the protein is delivered to the mitochondrial intermembrane space and promotes the enhancement of host ATP production by boosting the oxidative phosphorylation metabolic pathway. Furthermore, the release of cytochrome c from mitochondria, an early apoptotic event in response to short-term oxidative stress, is delayed in Rv1813c-expressing cells. This study reveals a novel class of mitochondria targeting effectors from Mtb that might participate in host cell metabolic reprogramming and apoptosis control during Mtb infections. IMPORTANCE In this article, using a combination of techniques (bioinformatics, structural biology, and cell biology), we identified and characterized a new class of effectors present only in intracellular mycobacteria. These proteins specifically target host cell mitochondria when ectopically expressed in cells. We showed that one member of this family (Rv1813c) affects mitochondria metabolism in a way that might twist the immune response. This effector also inhibits the cytochrome c exit from mitochondria, suggesting that it might alter normal host cell apoptotic capacities, one of the first defenses of immune cells against Mtb infection.
Collapse
Affiliation(s)
- Marianne Martin
- Laboratory of Pathogen Host Interactions, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Angelique deVisch
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Yves-Marie Boudehen
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse CNRS, UPS, Toulouse, France
| | - Philippe Barthe
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Claude Gutierrez
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse CNRS, UPS, Toulouse, France
| | - Obolbek Turapov
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Talip Aydogan
- Laboratory of Pathogen Host Interactions, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Laurène Heriaud
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Jerome Gracy
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse CNRS, UPS, Toulouse, France
| | - Galina V. Mukamolova
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - François Letourneur
- Laboratory of Pathogen Host Interactions, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Martin Cohen-Gonsaud
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| |
Collapse
|
18
|
Role of mitochondria in regulating immune response during bacterial infection. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 374:159-200. [PMID: 36858655 DOI: 10.1016/bs.ircmb.2022.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mitochondria are dynamic organelles of eukaryotes involved in energy production and fatty acid oxidation. Besides maintaining ATP production, calcium signaling, cellular apoptosis, and fatty acid synthesis, mitochondria are also known as the central hub of the immune system as it regulates the innate immune pathway during infection. Mitochondria mediated immune functions mainly involve regulation of reactive oxygen species production, inflammasome activation, cytokine secretion, and apoptosis of infected cells. Recent findings indicate that cellular mitochondria undergo constant biogenesis, fission, fusion and degradation, and these dynamics regulate cellular immuno-metabolism. Several intracellular pathogens target and modulate these normal functions of mitochondria to facilitate their own survival and growth. De-regulation of mitochondrial functions and dynamics favors bacterial infection and pathogens are able to protect themselves from mitochondria mediated immune responses. Here, we will discuss how mitochondria mediated anti-bacterial immune pathways help the host to evade pathogenic insult. In addition, examples of bacterial pathogens modulating mitochondrial metabolism and dynamics will also be elaborated. Study of these interactions between the mitochondria and bacterial pathogens during infection will lead to a better understanding of the mitochondrial metabolism pathways and dynamics important for the establishment of bacterial diseases. In conclusion, detailed studies on how mitochondria regulate the immune response during bacterial infection can open up new avenues to develop mitochondria centric anti-bacterial therapeutics.
Collapse
|
19
|
Role of C-terminal domain of Mycobacterium tuberculosis PE6 (Rv0335c) protein in host mitochondrial stress and macrophage apoptosis. Apoptosis 2023; 28:136-165. [PMID: 36258102 PMCID: PMC9579591 DOI: 10.1007/s10495-022-01778-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2022] [Indexed: 11/02/2022]
Abstract
PE/PPE proteins of Mycobacterium tuberculosis (Mtb) target the host organelles to dictate the outcome of infection. This study investigated the significance of PE6/Rv0335c protein's unique C-terminal in causing host mitochondrial perturbations and apoptosis. In-silico analysis revealed that similar to eukaryotic apoptotic Bcl2 proteins, Rv0335c had disordered, hydrophobic C-terminal and two BH3-like motifs in which one was located at C-terminal. Also, Rv0335c's N terminal had mitochondrial targeting sequence. Since, C-terminal of Bcl2 proteins are crucial for mitochondria targeting and apoptosis; it became relevant to evaluate the role of Rv0335c's C-terminal domain in modulating host mitochondrial functions and apoptosis. To confirm this, in-vitro experiments were conducted with Rv0335c whole protein and Rv0335c∆Cterm (C-terminal domain deleted Rv0335c) protein. Rv0335c∆Cterm caused significant reduction in mitochondrial perturbations and Caspase-mediated apoptosis of THP1 macrophages in comparison to Rv0335c. However, the deletion of C-terminal domain didn't affect Rv0335c's ability to localize to mitochondria. Nine Ca2+ binding residues were predicted within Rv0335c and four of them were at the C-terminal. In-vitro studies confirmed that Rv0335c caused significant increase in intracellular calcium influx whereas Rv0335c∆Cterm had insignificant effect on Ca2+ influx. Rv0335c has been reported to be a TLR4 agonist and, we observed a significant reduction in the expression of TLR4-HLA-DR-TNF-α in response to Rv0335c∆Cterm protein also suggesting the role of Rv0335c's C-terminal domain in host-pathogen interaction. These findings indicate the possibility of Rv0335c as a molecular mimic of eukaryotic Bcl2 proteins which equips it to cause host mitochondrial perturbations and apoptosis that may facilitate pathogen persistence.
Collapse
|
20
|
Jeon SM, Kim YJ, Nguyen TQ, Cui J, Thi Bich Hanh B, Silwal P, Kim JK, Kim JM, Oh DC, Jang J, Jo EK. Ohmyungsamycin Promotes M1-like Inflammatory Responses to Enhance Host Defense against Mycobacteroides abscessus Infections. Virulence 2022; 13:1966-1984. [PMID: 36271707 DOI: 10.1080/21505594.2022.2138009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Ohmyungsamycin A (OMS) is a newly identified cyclic peptide that exerts antimicrobial effects against Mycobacterium tuberculosis. However, its role in nontuberculous mycobacteria (NTMs) infections has not been clarified. Mycobacteroides abscessus (Mabc) is a rapidly growing NTM that has emerged as a human pathogen in both immunocompetent and immunosuppressed individuals. In this study, we demonstrated that OMS had significant antimicrobial effects against Mabc infection in both immunocompetent and immunodeficient mice, and in macrophages. OMS treatment amplified Mabc-induced expression of M1-related proinflammatory cytokines and inducible nitric oxide synthase, and significantly downregulated arginase-1 expression in murine macrophages. In addition, OMS augmented Mabc-mediated production of mitochondrial reactive oxygen species (mtROS), which promoted M1-like proinflammatory responses in Mabc-infected macrophages. OMS-induced production of mtROS and nitric oxide was critical for OMS-mediated antimicrobial responses during Mabc infections. Notably, the combination of OMS and rifabutin had a synergistic effect on the antimicrobial responses against Mabc infections in vitro, in murine macrophages, and in zebrafish models in vivo. Collectively, these data strongly suggest that OMS may be an effective M1-like adjunctive therapeutic against Mabc infections, either alone or in combination with antibiotics.
Collapse
Affiliation(s)
- Sang Min Jeon
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Thanh Quang Nguyen
- Division of Applied Life Science (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Jinsheng Cui
- Department of Microbiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Bui Thi Bich Hanh
- Division of Applied Life Science (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Jin-Man Kim
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Pathology, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Dong-Chan Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University,Jinju, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| |
Collapse
|
21
|
Pagán AJ, Lee LJ, Edwards-Hicks J, Moens CB, Tobin DM, Busch-Nentwich EM, Pearce EL, Ramakrishnan L. mTOR-regulated mitochondrial metabolism limits mycobacterium-induced cytotoxicity. Cell 2022; 185:3720-3738.e13. [PMID: 36103894 PMCID: PMC9596383 DOI: 10.1016/j.cell.2022.08.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/17/2022] [Accepted: 08/16/2022] [Indexed: 02/01/2023]
Abstract
Necrosis of macrophages in the granuloma, the hallmark immunological structure of tuberculosis, is a major pathogenic event that increases host susceptibility. Through a zebrafish forward genetic screen, we identified the mTOR kinase, a master regulator of metabolism, as an early host resistance factor in tuberculosis. We found that mTOR complex 1 protects macrophages from mycobacterium-induced death by enabling infection-induced increases in mitochondrial energy metabolism fueled by glycolysis. These metabolic adaptations are required to prevent mitochondrial damage and death caused by the secreted mycobacterial virulence determinant ESAT-6. Thus, the host can effectively counter this early critical mycobacterial virulence mechanism simply by regulating energy metabolism, thereby allowing pathogen-specific immune mechanisms time to develop. Our findings may explain why Mycobacterium tuberculosis, albeit humanity's most lethal pathogen, is successful in only a minority of infected individuals.
Collapse
Affiliation(s)
- Antonio J. Pagán
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK,Department of Microbiology, University of Washington, Seattle, WA 98195, USA,Corresponding author
| | - Lauren J. Lee
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Joy Edwards-Hicks
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Cecilia B. Moens
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - David M. Tobin
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Elisabeth M. Busch-Nentwich
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Erika L. Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK,Department of Microbiology, University of Washington, Seattle, WA 98195, USA,Corresponding author
| |
Collapse
|
22
|
de Souza FG, Cavalcante GC. Mitochondria in Mycobacterium Infection: From the Immune System to Mitochondrial Haplogroups. Int J Mol Sci 2022; 23:ijms23179511. [PMID: 36076909 PMCID: PMC9455157 DOI: 10.3390/ijms23179511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/23/2022] Open
Abstract
In humans, mitochondria play key roles in the regulation of cellular functions, such as the regulation of the innate immune response and are targets of several pathogenic viruses and bacteria. Mycobacteria are intracellular pathogens that infect cells important to the immune system of organisms and target mitochondria to meet their energy demands. In this review, we discuss the main mechanisms by which mitochondria regulate the innate immune response of humans to mycobacterial infection, especially those that cause tuberculosis and leprosy. Notably, the importance of mitochondrial haplogroups and ancestry studies for mycobacterial diseases is also discussed.
Collapse
|
23
|
Wigger GW, Bouton TC, Jacobson KR, Auld SC, Yeligar SM, Staitieh BS. The Impact of Alcohol Use Disorder on Tuberculosis: A Review of the Epidemiology and Potential Immunologic Mechanisms. Front Immunol 2022; 13:864817. [PMID: 35432348 PMCID: PMC9009367 DOI: 10.3389/fimmu.2022.864817] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Globally, an estimated 107 million people have an alcohol use disorder (AUD) leading to 2.8 million premature deaths each year. Tuberculosis (TB) is one of the leading causes of death globally and over 8% of global TB cases are estimated to be attributable to AUD. Social determinants of health such as poverty and undernutrition are often shared among those with AUD and TB and could explain the epidemiologic association between them. However, recent studies suggest that these shared risk factors do not fully account for the increased risk of TB in people with AUD. In fact, AUD has been shown to be an independent risk factor for TB, with a linear increase in the risk for TB with increasing alcohol consumption. While few studies have focused on potential biological mechanisms underlying the link between AUD and TB, substantial overlap exists between the effects of alcohol on lung immunity and the mechanisms exploited by Mycobacterium tuberculosis (Mtb) to establish infection. Alcohol misuse impairs the immune functions of the alveolar macrophage, the resident innate immune effector in the lung and the first line of defense against Mtb in the lower respiratory tract. Chronic alcohol ingestion also increases oxidative stress in the alveolar space, which could in turn facilitate Mtb growth. In this manuscript, we review the epidemiologic data that links AUD to TB. We discuss the existing literature on the potential mechanisms by which alcohol increases the risk of TB and review the known effects of alcohol ingestion on lung immunity to elucidate other mechanisms that Mtb may exploit. A more in-depth understanding of the link between AUD and TB will facilitate the development of dual-disease interventions and host-directed therapies to improve lung health and long-term outcomes of TB.
Collapse
Affiliation(s)
- Gregory W Wigger
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Tara C Bouton
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Karen R Jacobson
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Sara C Auld
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Atlanta VA Medical Center, Atlanta, GA, United States
| | - Bashar S Staitieh
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
24
|
Scarim CB, Pavan FR. Recent advancement in drug development of nitro(NO 2 )-heterocyclic compounds as lead scaffolds for the treatment of Mycobacterium tuberculosis. Drug Dev Res 2022; 83:842-858. [PMID: 35106801 DOI: 10.1002/ddr.21921] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/26/2021] [Accepted: 01/13/2022] [Indexed: 11/11/2022]
Abstract
Tuberculosis (TB) is an infectious disease caused predominantly by Mycobacterium tuberculosis (Mtb). It was responsible for approximately 1.4 million deaths worldwide in 2019. The lack of new drugs to treat drug-resistant strains is a principal factor for the slow rise in TB infections. Our aim is to aid the development of new TB treatments by describing improvements (last decade, 2011-2021) to nitro(NO2 )-based compounds that have shown activity or pharmacological properties (e.g., anti-proliferative, anti-kinetoplastid) against Mtb. For all compounds, we have included final correlations of minimum inhibitory concentrations against Mtb (H37 Rv).
Collapse
Affiliation(s)
- Cauê Benito Scarim
- Department of Cell and Molecular Biology, University of Mississippi Medical Center (UMMC), Jackson, Mississippi, USA
| | - Fernando Rogério Pavan
- School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, Sao Paulo, Brazil
| |
Collapse
|
25
|
Vaughn B, Abu Kwaik Y. Idiosyncratic Biogenesis of Intracellular Pathogens-Containing Vacuoles. Front Cell Infect Microbiol 2021; 11:722433. [PMID: 34858868 PMCID: PMC8632064 DOI: 10.3389/fcimb.2021.722433] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
While most bacterial species taken up by macrophages are degraded through processing of the bacteria-containing vacuole through the endosomal-lysosomal degradation pathway, intravacuolar pathogens have evolved to evade degradation through the endosomal-lysosomal pathway. All intra-vacuolar pathogens possess specialized secretion systems (T3SS-T7SS) that inject effector proteins into the host cell cytosol to modulate myriad of host cell processes and remodel their vacuoles into proliferative niches. Although intravacuolar pathogens utilize similar secretion systems to interfere with their vacuole biogenesis, each pathogen has evolved a unique toolbox of protein effectors injected into the host cell to interact with, and modulate, distinct host cell targets. Thus, intravacuolar pathogens have evolved clear idiosyncrasies in their interference with their vacuole biogenesis to generate a unique intravacuolar niche suitable for their own proliferation. While there has been a quantum leap in our knowledge of modulation of phagosome biogenesis by intravacuolar pathogens, the detailed biochemical and cellular processes affected remain to be deciphered. Here we discuss how the intravacuolar bacterial pathogens Salmonella, Chlamydia, Mycobacteria, Legionella, Brucella, Coxiella, and Anaplasma utilize their unique set of effectors injected into the host cell to interfere with endocytic, exocytic, and ER-to-Golgi vesicle traffic. However, Coxiella is the main exception for a bacterial pathogen that proliferates within the hydrolytic lysosomal compartment, but its T4SS is essential for adaptation and proliferation within the lysosomal-like vacuole.
Collapse
Affiliation(s)
- Bethany Vaughn
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,Center for Predictive Medicine, College of Medicine, University of Louisville, Louisville, KY, United States
| |
Collapse
|
26
|
Mitochondria as a Cellular Hub in Infection and Inflammation. Int J Mol Sci 2021; 22:ijms222111338. [PMID: 34768767 PMCID: PMC8583510 DOI: 10.3390/ijms222111338] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are the energy center of the cell. They are found in the cell cytoplasm as dynamic networks where they adapt energy production based on the cell’s needs. They are also at the center of the proinflammatory response and have essential roles in the response against pathogenic infections. Mitochondria are a major site for production of Reactive Oxygen Species (ROS; or free radicals), which are essential to fight infection. However, excessive and uncontrolled production can become deleterious to the cell, leading to mitochondrial and tissue damage. Pathogens exploit the role of mitochondria during infection by affecting the oxidative phosphorylation mechanism (OXPHOS), mitochondrial network and disrupting the communication between the nucleus and the mitochondria. The role of mitochondria in these biological processes makes these organelle good targets for the development of therapeutic strategies. In this review, we presented a summary of the endosymbiotic origin of mitochondria and their involvement in the pathogen response, as well as the potential promising mitochondrial targets for the fight against infectious diseases and chronic inflammatory diseases.
Collapse
|
27
|
de Oliveira MF, Medeiros RCA, Mietto BS, Calvo TL, Mendonça APM, Rosa TLSA, da Silva DS, do Carmo de Vasconcelos KG, Pereira AMR, de Macedo CS, Pereira GMB, de Berrêdo Pinho Moreira M, Pessolani MCV, Moraes MO, Lara FA. Reduction of host cell mitochondrial activity as Mycobacterium leprae's strategy to evade host innate immunity. Immunol Rev 2021; 301:193-208. [PMID: 33913182 PMCID: PMC10084840 DOI: 10.1111/imr.12962] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
Leprosy is a much-feared incapacitating infectious disease caused by Mycobacterium leprae or M lepromatosis, annually affecting roughly 200,000 people worldwide. During host-pathogen interaction, M leprae subverts the immune response, leading to development of disease. Throughout the last few decades, the impact of energy metabolism on the control of intracellular pathogens and leukocytic differentiation has become more evident. Mitochondria play a key role in regulating newly-discovered immune signaling pathways by controlling redox metabolism and the flow of energy besides activating inflammasome, xenophagy, and apoptosis. Likewise, this organelle, whose origin is probably an alphaproteobacterium, directly controls the intracellular pathogens attempting to invade its niche, a feature conquered at the expense of billions of years of coevolution. In the present review, we discuss the role of reduced host cell mitochondrial activity during M leprae infection and the consequential fates of M leprae and host innate immunity. Conceivably, inhibition of mitochondrial energy metabolism emerges as an overlooked and novel mechanism developed by M leprae to evade xenophagy and the host immune response.
Collapse
Affiliation(s)
- Marcus Fernandes de Oliveira
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Bruno Siqueira Mietto
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Minas Gerais, Brazil
| | - Thyago Leal Calvo
- Laboratório de Hanseníase, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Ana Paula Miranda Mendonça
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | - Cristiana Santos de Macedo
- Laboratório de Microbiologia Celular, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
- Centro de Desenvolvimento Tecnológico em Saúde, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | - Flavio Alves Lara
- Laboratório de Microbiologia Celular, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
28
|
Ó Maoldomhnaigh C, Cox DJ, Phelan JJ, Malone FD, Keane J, Basdeo SA. The Warburg Effect Occurs Rapidly in Stimulated Human Adult but Not Umbilical Cord Blood Derived Macrophages. Front Immunol 2021; 12:657261. [PMID: 33927724 PMCID: PMC8076563 DOI: 10.3389/fimmu.2021.657261] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022] Open
Abstract
The Warburg effect, defined as increased glycolysis and decreased oxidative phosphorylation, occurs in murine macrophages following LPS stimulation and is required for activation. There are differences between human and murine macrophage metabolic responses to stimulation, with peak metabolite concentrations occurring earlier in humans than mice. Complex changes occur in the human immune system with age, resulting in the very young and the very old being more susceptible to infections. Anti-bacterial immune responses in umbilical cord immune cells are considered deficient but there is a paucity of data on the role that metabolism plays. We hypothesized that metabolic responses in human macrophages occur early during activation. In addition, we hypothesized that umbilical cord derived macrophages have an altered immunometabolic response compared with adult macrophages. We demonstrate that adult and cord blood monocyte derived macrophages (MDM) immediately increase glycolysis in response to stimulation with LPS or Mycobacterium tuberculosis (Mtb), however only adult MDM decrease oxidative phosphorylation. At 24 hours post stimulation, glycolysis remains elevated in both adult and cord blood MDM, oxidative phosphorylation remains unchanged in the cord blood MDM and has normalized in the adult MDM stimulated with Mtb. However, LPS stimulated adult MDM have increased oxidative phosphorylation at 24 hours, illustrating differences in metabolic responses to different stimuli, time-dependent variation in responses and differences in macrophage metabolism in adults compared with umbilical cord blood. We compared the phenotype and function of macrophages derived from adult or cord blood. Cord blood MDM secreted less TNF following Mtb stimulation and more IL-6 following LPS stimulation compared with adult MDM. Our findings demonstrate that whilst cord blood MDM exhibit an immediate increase in glycolytic flux in response to stimulation, similar to adult MDM, cord blood MDM do not concomitantly decrease oxygen consumption. This indicates that adult macrophages shift to Warburg metabolism immediately after stimulation, but cord blood macrophages do not. Understanding the differences in the metabolic profiles of macrophages over a human lifetime will enable the translation of immunometabolism into effective immuno-supportive therapies that could potentially be targeted at vulnerable populations, such as the very old and the very young.
Collapse
Affiliation(s)
- Cilian Ó Maoldomhnaigh
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Donal J Cox
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - James J Phelan
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Fergal D Malone
- Obstetrics & Gynecology, Royal College of Surgeons in Ireland, Rotunda Hospital, Dublin, Ireland
| | - Joseph Keane
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Sharee A Basdeo
- TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| |
Collapse
|
29
|
TLRs in Mycobacterial Pathogenesis: Black and White or Shades of Gray. Curr Microbiol 2021; 78:2183-2193. [PMID: 33844035 DOI: 10.1007/s00284-021-02488-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 04/01/2021] [Indexed: 12/19/2022]
Abstract
Toll-like receptors (TLRs) play critical role in the innate recognition of pathogens besides orchestrating innate and adaptive immune responses. These receptors exhibit exquisite specificity for different pathogens or their products and, through a complex network of signalling, generate appropriate immune responses. TLRs induce both pro- and anti-inflammatory signals depending on interactions with the adapter molecules thereby impacting the outcome of infection. Hence, TLR signalling ought to be stringently regulated to avoid harmful effects on the host. Mycobacteria express antigens which are sensed by TLRs leading to activation of various signalling molecules important for initiating the death of infected cells and containment of pathogens. Conversely, it also utilizes TLRs for immune evasion and persistence. Due to the enormous diversity in the repertoire of virulence traits expressed by mycobacteria, genetic variations in TLRs often impair the host's ability to respond to mycobacterial-stress, affecting health and disease manifestations. Thus, understanding TLR signalling is of great importance for insights into host-mycobacterial interactions and designing effective measures for controlling the spread and persistence of the bacterium.
Collapse
|
30
|
Shariq M, Quadir N, Sharma N, Singh J, Sheikh JA, Khubaib M, Hasnain SE, Ehtesham NZ. Mycobacterium tuberculosis RipA Dampens TLR4-Mediated Host Protective Response Using a Multi-Pronged Approach Involving Autophagy, Apoptosis, Metabolic Repurposing, and Immune Modulation. Front Immunol 2021; 12:636644. [PMID: 33746976 PMCID: PMC7969667 DOI: 10.3389/fimmu.2021.636644] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/03/2021] [Indexed: 12/26/2022] Open
Abstract
Reductive evolution has endowed Mycobacterium tuberculosis (M. tb) with moonlighting in protein functions. We demonstrate that RipA (Rv1477), a peptidoglycan hydrolase, activates the NFκB signaling pathway and elicits the production of pro-inflammatory cytokines, TNF-α, IL-6, and IL-12, through the activation of an innate immune-receptor, toll-like receptor (TLR)4. RipA also induces an enhanced expression of macrophage activation markers MHC-II, CD80, and CD86, suggestive of M1 polarization. RipA harbors LC3 (Microtubule-associated protein 1A/1B-light chain 3) motifs known to be involved in autophagy regulation and indeed alters the levels of autophagy markers LC3BII and P62/SQSTM1 (Sequestosome-1), along with an increase in the ratio of P62/Beclin1, a hallmark of autophagy inhibition. The use of pharmacological agents, rapamycin and bafilomycin A1, reveals that RipA activates PI3K-AKT-mTORC1 signaling cascade that ultimately culminates in the inhibition of autophagy initiating kinase ULK1 (Unc-51 like autophagy activating kinase). This inhibition of autophagy translates into efficient intracellular survival, within macrophages, of recombinant Mycobacterium smegmatis expressing M. tb RipA. RipA, which also localizes into mitochondria, inhibits the production of oxidative phosphorylation enzymes to promote a Warburg-like phenotype in macrophages that favors bacterial replication. Furthermore, RipA also inhibited caspase-dependent programed cell death in macrophages, thus hindering an efficient innate antibacterial response. Collectively, our results highlight the role of an endopeptidase to create a permissive replication niche in host cells by inducing the repression of autophagy and apoptosis, along with metabolic reprogramming, and pointing to the role of RipA in disease pathogenesis.
Collapse
Affiliation(s)
- Mohd Shariq
- Indian Council of Medical Research-National Institute of Pathology, New Delhi, India
| | - Neha Quadir
- Indian Council of Medical Research-National Institute of Pathology, New Delhi, India.,Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Neha Sharma
- Indian Council of Medical Research-National Institute of Pathology, New Delhi, India.,Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Jasdeep Singh
- Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Javaid A Sheikh
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Khubaib
- Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Seyed E Hasnain
- Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India.,Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, India.,Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D) Hauz Khas, New Delhi, India
| | - Nasreen Z Ehtesham
- Indian Council of Medical Research-National Institute of Pathology, New Delhi, India
| |
Collapse
|
31
|
Abstract
Intracellular proliferation of Legionella pneumophila within a vacuole in human alveolar macrophages is essential for manifestation of Legionnaires’ pneumonia. Intravacuolar growth of the pathogen is totally dependent on remodeling the L. pneumophila-containing vacuole (LCV) by the ER and on its evasion of the endosomal-lysosomal degradation pathway. Diversion of the Legionella pneumophila-containing vacuole (LCV) from the host endosomal-lysosomal degradation pathway is one of the main virulence features essential for manifestation of Legionnaires’ pneumonia. Many of the ∼350 Dot/Icm-injected effectors identified in L. pneumophila have been shown to interfere with various host pathways and processes, but no L. pneumophila effector has ever been identified to be indispensable for lysosomal evasion. While most single effector mutants of L. pneumophila do not exhibit a defective phenotype within macrophages, we show that the MavE effector is essential for intracellular growth of L. pneumophila in human monocyte-derived macrophages (hMDMs) and amoebae and for intrapulmonary proliferation in mice. The mavE null mutant fails to remodel the LCV with endoplasmic reticulum (ER)-derived vesicles and is trafficked to the lysosomes where it is degraded, similar to formalin-killed bacteria. During infection of hMDMs, the MavE effector localizes to the poles of the LCV membrane. The crystal structure of MavE, resolved to 1.8 Å, reveals a C-terminal transmembrane helix, three copies of tyrosine-based sorting motifs, and an NPxY eukaryotic motif, which binds phosphotyrosine-binding domains present on signaling and adaptor eukaryotic proteins. Two point mutations within the NPxY motif result in attenuation of L. pneumophila in both hMDMs and amoeba. The substitution defects of P78 and D64 are associated with failure of vacuoles harboring the mutant to be remodeled by the ER and results in fusion of the vacuole to the lysosomes leading to bacterial degradation. Therefore, the MavE effector of L. pneumophila is indispensable for phagosome biogenesis and lysosomal evasion.
Collapse
|
32
|
Price CT, Abu Kwaik Y. Evolution and Adaptation of Legionella pneumophila to Manipulate the Ubiquitination Machinery of Its Amoebae and Mammalian Hosts. Biomolecules 2021; 11:biom11010112. [PMID: 33467718 PMCID: PMC7830128 DOI: 10.3390/biom11010112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
The ubiquitin pathway is highly conserved across the eukaryotic domain of life and plays an essential role in a plethora of cellular processes. It is not surprising that many intracellular bacterial pathogens often target the essential host ubiquitin pathway. The intracellular bacterial pathogen Legionella pneumophila injects into the host cell cytosol multiple classes of classical and novel ubiquitin-modifying enzymes that modulate diverse ubiquitin-related processes in the host cell. Most of these pathogen-injected proteins, designated as effectors, mimic known E3-ubiquitin ligases through harboring F-box or U-box domains. The classical F-box effector, AnkB targets host proteins for K48-linked polyubiquitination, which leads to excessive proteasomal degradation that is required to generate adequate supplies of amino acids for metabolism of the pathogen. In contrast, the SidC and SdcA effectors share no structural similarity to known eukaryotic ligases despite having E3-ubiquitin ligase activity, suggesting that the number of E3-ligases in eukaryotes is under-represented. L. pneumophila also injects into the host many novel ubiquitin-modifying enzymes, which are the SidE family of effectors that catalyze phosphoribosyl-ubiquitination of serine residue of target proteins, independently of the canonical E1-2-3 enzymatic cascade. Interestingly, the environmental bacterium, L. pneumophila, has evolved within a diverse range of amoebal species, which serve as the natural hosts, while accidental transmission through contaminated aerosols can cause pneumonia in humans. Therefore, it is likely that the novel ubiquitin-modifying enzymes of L. pneumophila were acquired by the pathogen through interkingdom gene transfer from the diverse natural amoebal hosts. Furthermore, conservation of the ubiquitin pathway across eukaryotes has enabled these novel ubiquitin-modifying enzymes to function similarly in mammalian cells. Studies on the biological functions of these effectors are likely to reveal further novel ubiquitin biology and shed further lights on the evolution of ubiquitin.
Collapse
Affiliation(s)
- Christopher T.D. Price
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA;
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA;
- Center for Predictive Medicine, College of Medicine, University of Louisville, Louisville, KY 40202, USA
- Correspondence:
| |
Collapse
|