1
|
Wu Q, He M, Wang J, Tong T, Yang D, Tang H. The therapeutic mechanism of Chebulae Fructus in the treatment of immunosuppression in Chinese yellow quail on the basis of network pharmacology. Front Vet Sci 2023; 10:1123449. [PMID: 37275616 PMCID: PMC10235497 DOI: 10.3389/fvets.2023.1123449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/04/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction Chebulae Fructus (Terminalia chebula Retz.) is a well-known traditional Chinese medicine (TCM), one of the family Combretaceae, whose immature fruit is called Fructus Chebulae Immaturus or Zangqingguo. This present study aimed at detecting the target and therapeutic mechanism of Chebulae Fructus against immunosuppression through network analysis and experimental validation. Methods Effective components and potential targets of Chebulae Fructus were Search and filtered through the Chinese herbal medicine pharmacology data and analysis platform. A variety of known disease target databases were employed to screen the therapeutic target proteins against immunosuppression and thus constructing a protein-protein interaction network. Hub genes and key pathways in this study were identified by continuous project enrichment analysis. Further, the core targets and therapeutic mechanism of Chebulae Fructus against immunosuppression in Chinese yellow quail through animal experiment. Results Seventy-five identifiable major candidate targets of Chebulae Fructus were found and thus constructing a drug-compound-target-disease network. Targets derived from gene enrichment analysis play pivotal roles in lipid and atherosclerosis, fluid shear stress and atherosclerosis, and the hepatitis B pathway. Height of plicate and areas of lymphoid follicle were both increased and the expression of GATA-3 and T-bet was upregulated in Chinese yellow quail fed with Chebulae Fructus in animal experiment. Conclusion Chebulae Fructus may be a helpful Chinese medicine with immunosuppressive effect and prospective applications in future. Further research is also needed to understand the mechanisms of immunosuppression and the mechanism of action of immunomodulators.
Collapse
Affiliation(s)
- Qiang Wu
- Agricultural College, Yibin Vocational and Technical College, Yibin, China
| | - Min He
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jing Wang
- Agricultural College, Yibin Vocational and Technical College, Yibin, China
| | - TieJin Tong
- Agricultural College, Yibin Vocational and Technical College, Yibin, China
| | - Dan Yang
- Agricultural College, Yibin Vocational and Technical College, Yibin, China
| | - Huaqiao Tang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
2
|
Wang Y, Tian Q, Ye L. The Differentiation and Maintenance of SARS-CoV-2-Specific Follicular Helper T Cells. Front Cell Infect Microbiol 2022; 12:953022. [PMID: 35909969 PMCID: PMC9329515 DOI: 10.3389/fcimb.2022.953022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Upon acute viral infection, virus-specific CD4+ T cells differentiate into either TH1 cells or follicular helper T (TFH) cells. The molecular pathways governing such bimodal cell fate commitment remain elusive. Additionally, effector virus-specific TFH cells further differentiate into corresponding memory population, which confer long-term protection against re-infection of same viruses by providing immediate help to virus-specific memory B cells. Currently, the molecular mechanisms underlying the long-term maintenance of memory TFH cells are largely unknown. In this review, we discuss current understanding of early differentiation of virus-specific effector TFH cells and long-term maintenance of virus-specific memory TFH cells in mouse models of viral infection and patients of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection.
Collapse
Affiliation(s)
- Yifei Wang
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qin Tian
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Lilin Ye
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
- *Correspondence: Lilin Ye,
| |
Collapse
|
3
|
Zhou L, Ge M, Zhang Y, Wu X, Leng M, Gan C, Mou Y, Zhou J, Valencia CA, Hao Q, Zhu B, Dong B, Dong B. Centenarians Alleviate Inflammaging by Changing the Ratio and Secretory Phenotypes of T Helper 17 and Regulatory T Cells. Front Pharmacol 2022; 13:877709. [PMID: 35721185 PMCID: PMC9203077 DOI: 10.3389/fphar.2022.877709] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/29/2022] [Indexed: 02/05/2023] Open
Abstract
The immune system of centenarians remains active and young to prevent cancer and infections. Aging is associated with inflammaging, a persistent low-grade inflammatory state in which CD4+ T cells play a role. However, there are few studies that have been done on the CD4+ T cell subsets in centenarians. Herein, the changes in CD4+ T cell subsets were investigated in centenarians. It was found that with aging, the old adults had higher levels of proinflammatory cytokines and lower levels of anti-inflammatory cytokines in plasma. The levels of CRP, IL-12, TNF-α, IFN-γ, IL-6 and IL-10 were further increased in centenarians compared to old adults. While the levels of IL-17A, IL-1β, IL-23 and TGF-β in centenarians were closer to those in young adults. The total CD4+, CD8+, Th17 and Treg cells from peripheral blood mononuclear cells (PBMCs) were similar among the three groups. It was observed that the ratio of Th17/Treg cells was elevated in old adults compared to young adults. The ratio was not further elevated in centenarians but rather decreased. In addition, the ex vivo PBMCs differentiation assay showed that increased Th17 cells in centenarians tended to secrete fewer proinflammatory cytokines, while decreased Treg cells in centenarians were prone to secrete more anti-inflammatory cytokines. These observations suggested centenarians alleviated inflammaging by decreasing the ratio of Th17/Treg cells and changing them into anti-inflammatory secretory phenotypes, which provided a novel mechanism for anti-aging research.
Collapse
Affiliation(s)
- Lixing Zhou
- National Clinical Research Center for Geriatrics and Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Meiling Ge
- National Clinical Research Center for Geriatrics and Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics and Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaochu Wu
- National Clinical Research Center for Geriatrics and Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Mi Leng
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chunmei Gan
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Mou
- Geroscience and Chronic Disease Department, The 8th Municipal Hospital for the People, Chengdu, China
| | - Jiao Zhou
- National Clinical Research Center for Geriatrics and Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - C Alexander Valencia
- National Clinical Research Center for Geriatrics and Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Interpath Laboratory, Pendleton, OR, United States.,Department of Preclinical Education, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | - Qiukui Hao
- National Clinical Research Center for Geriatrics and Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Zhu
- Geroscience and Chronic Disease Department, The 8th Municipal Hospital for the People, Chengdu, China
| | - Biao Dong
- National Clinical Research Center for Geriatrics and Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Birong Dong
- National Clinical Research Center for Geriatrics and Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Santoro A, Bientinesi E, Monti D. Immunosenescence and inflammaging in the aging process: age-related diseases or longevity? Ageing Res Rev 2021; 71:101422. [PMID: 34391943 DOI: 10.1016/j.arr.2021.101422] [Citation(s) in RCA: 213] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022]
Abstract
During aging the immune system (IS) undergoes remarkable changes that collectively are known as immunosenescence. It is a multifactorial and dynamic phenomenon that affects both natural and acquired immunity and plays a critical role in most chronic diseases in older people. For a long time, immunosenescence has been considered detrimental because it may lead to a low-grade, sterile chronic inflammation we proposed to call "inflammaging" and a progressive reduction in the ability to trigger effective antibody and cellular responses against infections and vaccinations. Recently, many scientists revised this negative meaning because it can be considered an essential adaptation/remodeling resulting from the lifelong immunological biography of single individuals from an evolutionary perspective. Inflammaging can be considered an adaptive process because it can trigger an anti-inflammatory response to counteract the age-related pro-inflammatory environment. Centenarians represent a valuable model to study the beneficial changes occurring in the IS with age. These extraordinary individuals reached the extreme limits of human life by slowing down the aging process and, in most cases, delaying, avoiding or surviving the major age-associated diseases. They indeed show a complex and heterogeneous phenotype determined by an improved ability to adapt and remodel in response to harmful stimuli. This review aims to point out the intimate relationship between immunosenescence and inflammaging and how these processes impact unsuccessful aging rather than longevity. We also describe the gut microbiota age-related changes as one of the significant triggers of inflammaging and the sex/gender differences in the immune system of the elderly, contributing to the sex/gender disparity in terms of epidemiology, pathophysiology, symptoms and severity of age-related diseases. Finally, we discuss how these phenomena could influence the susceptibility to COVID-19 infection.
Collapse
|
5
|
Callender LA, Schroth J, Carroll EC, Garrod-Ketchley C, Romano LEL, Hendy E, Kelly A, Lavender P, Akbar AN, Chapple JP, Henson SM. GATA3 induces mitochondrial biogenesis in primary human CD4 + T cells during DNA damage. Nat Commun 2021; 12:3379. [PMID: 34099719 PMCID: PMC8184923 DOI: 10.1038/s41467-021-23715-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/06/2021] [Indexed: 11/09/2022] Open
Abstract
GATA3 is as a lineage-specific transcription factor that drives the differentiation of CD4+ T helper 2 (Th2) cells, but is also involved in a variety of processes such as immune regulation, proliferation and maintenance in other T cell and non-T cell lineages. Here we show a mechanism utilised by CD4+ T cells to increase mitochondrial mass in response to DNA damage through the actions of GATA3 and AMPK. Activated AMPK increases expression of PPARG coactivator 1 alpha (PPARGC1A or PGC1α protein) at the level of transcription and GATA3 at the level of translation, while DNA damage enhances expression of nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2). PGC1α, GATA3 and NRF2 complex together with the ATR to promote mitochondrial biogenesis. These findings extend the pleotropic interactions of GATA3 and highlight the potential for GATA3-targeted cell manipulation for intervention in CD4+ T cell viability and function after DNA damage. GATA3 has been considered to be primarily associated with CD4+ Th2 cell function. Using CD4+ effector memory that re-express CD45RA (EMRA) T cells the authors show that in response to DNA damage GATA3 can regulate increase of mitochondrial mass and biogenesis involving AMPK.
Collapse
Affiliation(s)
- Lauren A Callender
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Translational Science, Achilles Therapeutics Ltd, Stevenage Bioscience Catalyst, Stevenage, UK
| | - Johannes Schroth
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Elizabeth C Carroll
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Life Sciences, Institute of Technology Sligo, Sligo, Ireland
| | - Conor Garrod-Ketchley
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Lisa E L Romano
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eleanor Hendy
- Peter Gorer Department of Immunobiology and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Audrey Kelly
- Peter Gorer Department of Immunobiology and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Paul Lavender
- Peter Gorer Department of Immunobiology and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Arne N Akbar
- Division of Infection and Immunity, Department of Immunology, University College London, London, UK
| | - J Paul Chapple
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sian M Henson
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
6
|
Ganaie AA, Mansini AP, Hussain T, Rao A, Siddique HR, Shabaneh A, Ferrari MG, Murugan P, Klingelhöfer J, Wang J, Ambartsumian N, Warlick CA, Konety BR, Saleem M. Anti-S100A4 Antibody Therapy Is Efficient in Treating Aggressive Prostate Cancer and Reversing Immunosuppression: Serum and Biopsy S100A4 as a Clinical Predictor. Mol Cancer Ther 2020; 19:2598-2611. [PMID: 32999046 DOI: 10.1158/1535-7163.mct-20-0410] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/27/2020] [Accepted: 09/16/2020] [Indexed: 11/16/2022]
Abstract
S100A4 oncoprotein plays a critical role during prostate cancer progression and induces immunosuppression in host tissues. We hypothesized that S100A4-regulated oncogenic activity in immunosuppressed prostate tumors promotes growth of neoplastic cells, which are likely to become aggressive. In the current study, we investigated whether biopsy-S100A4 gene alteration independently predicts the outcome of disease in patients and circulatory-S100A4 is druggable target for treating immunosuppressive prostate cancer. Aided by DECIPHER-genomic test, we show biopsy-S100A4 overexpression as predictive of (i) poor ADT response and (ii) high risk of mortality in 228 radical prostatectomy-treated patients. Furthermore, analysis of tumor genome data of more than 1,000 patients with prostate cancer (PRAD/SU2C/FHCRC studies) validated the association of S100A4-alteration to poor survival and metastasis. We show that increased serum-S100A4 levels are associated to the prostate cancer progression in patients. The prerequisite for metastasis is the escape of tumor cells via vascular system. We show that extracellular-S100A4 protein as a growth factor induces vascular transmigration of prostate cancer cells and bone demineralization thus forms an ideal target for therapies for treating prostate cancer. By employing surface plasmon resonance and isothermal titration calorimetry, we show that mab6B12 antibody interacts with and neutralizes S100A4 protein. When tested for therapeutic efficacy, the mab6B12 therapy reduced the (i) osteoblastic demineralization of bone-derived MSCs, (ii) S100A4-target (NFκB/MMP9/VEGF) levels in prostate cancer cells, and (iii) tumor growth in a TRAMPC2 syngeneic mouse model. The immuno-profile analysis showed that mAb6B12-therapy (i) shifted Th1/Th2 balance (increased Stat4+/T-bet+ and decreased GATA2+/CD68+/CD45+/CD206+ cells); (ii) modulated cytokine levels in CD4+ T cells; and (iii) decreased levels of IL5/6/12/13, sTNFR1, and serum-RANTES. We suggest that S100A4-antibody therapy has clinical applicability in treating immunosuppressive prostate cancer in patients.
Collapse
Affiliation(s)
- Arsheed A Ganaie
- Department of Urology, Medical School, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Adrian P Mansini
- Department of Urology, Medical School, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Tabish Hussain
- Department of Urology, Medical School, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Arpit Rao
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Hifzur R Siddique
- Department of Urology, Medical School, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Ashraf Shabaneh
- Institute for Health Informatics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Marina G Ferrari
- Department of Urology, Medical School, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Paari Murugan
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Jörg Klingelhöfer
- Danish Cancer Society Research Center, Copenhagen, Denmark.,Laboratory of Neural Plasticity, Copenhagen University, Copenhagen, Denmark
| | - Jinhua Wang
- Institute for Health Informatics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Noona Ambartsumian
- Danish Cancer Society Research Center, Copenhagen, Denmark.,Laboratory of Neural Plasticity, Copenhagen University, Copenhagen, Denmark
| | - Christopher A Warlick
- Department of Urology, Medical School, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Badrinath R Konety
- Department of Urology, Medical School, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Rush Medical College, Rush University, Chicago, Illinois
| | - Mohammad Saleem
- Department of Urology, Medical School, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
7
|
Modulation of T helper 1 and T helper 2 immune balance in a murine stress model during Chlamydia muridarum genital infection. PLoS One 2020; 15:e0226539. [PMID: 32413046 PMCID: PMC7228091 DOI: 10.1371/journal.pone.0226539] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/22/2020] [Indexed: 02/08/2023] Open
Abstract
A murine model to study the effect of cold-induced stress (CIS) on Chlamydia muridarum genital infection and immune response has been developed in our laboratory. Previous results in the lab show that CIS increases the intensity of chlamydia genital infection, but little is known about the effects and mechanisms of CIS on the differentiation and activities of CD4+ T cell subpopulations and bone marrow-derived dendritic cells (BMDCs). The factors that regulate the production of T helper 1 (Th1) or T helper 2 (Th2) cytokines are not well defined. In this study, we examined whether CIS modulates the expressions of beta-adrenergic receptor (β-AR), transcription factors, hallmark cytokines of Th1 and Th2, and differentiation of BMDCs during C. muridarum genital infection in the murine model. Our results show that the mRNA level of the beta2-adrenergic receptor (β2-AR) compared to β1-AR and β3-AR was high in the mixed populations of CD4+ T cells and BMDCs. Furthermore, we observed decreased expression of T-bet, low level of Interferon-gamma (IFN-γ) production, increased expression of GATA-3, and Interleukin-4 (IL-4) production in CD4+ T cells of stressed mice. Exposure of BMDCs to Fenoterol, β2-AR agonist, or ICI118,551, β2-AR antagonist, revealed significant β2-AR stimulation or inhibition, respectively, in stressed mice. Moreover, co-culturing of mature BMDCs and naïve CD4+ T cells increased the production of IL-4, IL-10, L-17, and IL-23 cytokines, suggesting that stimulation of β2-AR leads to the increased production of Th2 cytokines. Overall, our results show for the first time that CIS promotes the switching from a Th1 to Th2 cytokine environment. This was evidenced in the murine stress model by the overexpression of GATA-3 concurrent with elevated IL-4 production, reduced T-bet expression, and IFN-γ secretion.
Collapse
|
8
|
Yang Q, Qu J, Jin C, Feng Y, Xie S, Zhu J, Liu G, Xie H, Qiu H, Qi Y, Mu J, Huang J. Schistosoma japonicum Infection Promotes the Response of Tfh Cells Through Down-Regulation of Caspase-3-Mediating Apoptosis. Front Immunol 2019; 10:2154. [PMID: 31572373 PMCID: PMC6753327 DOI: 10.3389/fimmu.2019.02154] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 08/28/2019] [Indexed: 01/13/2023] Open
Abstract
CD4+ T follicular helper (Tfh) cells, a new subset of immune cells, have been demonstrated to be involved in granulomatous responses to Schistosoma japonicum (S. japonicum) infection. However, the role and underlying mechanisms of Tfh cell aggregation in S. japonicum infection remain incompletely understood. In this study, we provide evidence that S. japonicum infection enhances the accumulation of Tfh cells in the spleen, lymph nodes, and peripheral blood of C57BL/6 mice. Infection-induced Tfh cells exhibited more potent effects directly on B cell responses than the control Tfh cells (P < 0.05). Furthermore, reduced apoptosis of Tfh cells was found both in S. japonicum infected mice and in soluble egg antigen (SEA) treated Tfh cells (P < 0.05). Mechanistic studies reveal that caspase-3 is the primary drivers of down-regulated apoptotic Tfh cell death in S. japonicum infection. In summary, this study demonstrates that Tfh cell accumulation might have an impact on the generation of immune responses in S. japonicum infection, and caspase-3 signaling mediated apoptosis down-regulation might responsible for the accumulation of Tfh cell in this course.
Collapse
Affiliation(s)
- Quan Yang
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiale Qu
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chenxi Jin
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuanfa Feng
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shihao Xie
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jinxin Zhu
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Gaoshen Liu
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hongyan Xie
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huaina Qiu
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yanwei Qi
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jun Huang
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Phelan JJ, Basdeo SA, Tazoll SC, McGivern S, Saborido JR, Keane J. Modulating Iron for Metabolic Support of TB Host Defense. Front Immunol 2018; 9:2296. [PMID: 30374347 PMCID: PMC6196273 DOI: 10.3389/fimmu.2018.02296] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/17/2018] [Indexed: 02/05/2023] Open
Abstract
Tuberculosis (TB) is the world's biggest infectious disease killer. The increasing prevalence of multidrug-resistant and extensively drug-resistant TB demonstrates that current treatments are inadequate and there is an urgent need for novel therapies. Research is now focused on the development of host-directed therapies (HDTs) which can be used in combination with existing antimicrobials, with a special focus on promoting host defense. Immunometabolic reprogramming is integral to TB host defense, therefore, understanding and supporting the immunometabolic pathways that are altered after infection will be important for the development of new HDTs. Moreover, TB pathophysiology is interconnected with iron metabolism. Iron is essential for the survival of Mycobacterium tuberculosis (Mtb), the bacteria that causes TB disease. Mtb struggles to replicate and persist in low iron environments. Iron chelation has therefore been suggested as a HDT. In addition to its direct effects on iron availability, iron chelators modulate immunometabolism through the stabilization of HIF1α. This review examines immunometabolism in the context of Mtb and its links to iron metabolism. We suggest that iron chelation, and subsequent stabilization of HIF1α, will have multifaceted effects on immunometabolic function and holds potential to be utilized as a HDT to boost the host immune response to Mtb infection.
Collapse
Affiliation(s)
- James J Phelan
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Sharee A Basdeo
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Simone C Tazoll
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Sadhbh McGivern
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Judit R Saborido
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| |
Collapse
|
10
|
Fulop T, Larbi A, Dupuis G, Le Page A, Frost EH, Cohen AA, Witkowski JM, Franceschi C. Immunosenescence and Inflamm-Aging As Two Sides of the Same Coin: Friends or Foes? Front Immunol 2018; 8:1960. [PMID: 29375577 PMCID: PMC5767595 DOI: 10.3389/fimmu.2017.01960] [Citation(s) in RCA: 791] [Impact Index Per Article: 131.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022] Open
Abstract
The immune system is the most important protective physiological system of the organism. It has many connections with other systems and is, in fact, often considered as part of the larger neuro-endocrine-immune axis. Most experimental data on immune changes with aging show a decline in many immune parameters when compared to young healthy subjects. The bulk of these changes is termed immunosenescence. Immunosenescence has been considered for some time as detrimental because it often leads to subclinical accumulation of pro-inflammatory factors and inflamm-aging. Together, immunosenescence and inflamm-aging are suggested to stand at the origin of most of the diseases of the elderly, such as infections, cancer, autoimmune disorders, and chronic inflammatory diseases. However, an increasing number of immune-gerontologists have challenged this negative interpretation of immunosenescence with respect to its significance in aging-related alterations of the immune system. If one considers these changes from an evolutionary perspective, they can be viewed preferably as adaptive or remodeling rather than solely detrimental. Whereas it is conceivable that global immune changes may lead to various diseases, it is also obvious that these changes may be needed for extended survival/longevity. Recent cumulative data suggest that, without the existence of the immunosenescence/inflamm-aging duo (representing two sides of the same phenomenon), human longevity would be greatly shortened. This review summarizes recent data on the dynamic reassessment of immune changes with aging. Accordingly, attempts to intervene on the aging immune system by targeting its rejuvenation, it may be more suitable to aim to maintain general homeostasis and function by appropriately improving immune-inflammatory-functions.
Collapse
Affiliation(s)
- Tamas Fulop
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Biopolis, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Gilles Dupuis
- Department of Biochemistry, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Aurélie Le Page
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Eric H. Frost
- Department of Infectious Diseases and Microbiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Alan A. Cohen
- Department of Family Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Jacek M. Witkowski
- Department of Pathophysiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Claudio Franceschi
- Italian National Research Center on Aging, Department of Experimental Pathology, University of Bologna, Bologna, Italy
| |
Collapse
|