51
|
An P, Sezgin E, Kirk GD, Duggal P, Binns-Roemer E, Nelson G, Limou S, Van Natta ML, Jabs DA, Estrella M, Kopp JB, Winkler CA. APOL1 variant alleles associate with reduced risk for opportunistic infections in HIV infection. Commun Biol 2021; 4:284. [PMID: 33674766 PMCID: PMC7977062 DOI: 10.1038/s42003-021-01812-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/08/2021] [Indexed: 11/10/2022] Open
Abstract
Apolipoprotein L1 (APOL1), an innate immune factor against African trypanosoma brucei, inhibits HIV-1 in vitro. The impact of APOL1 G1-G2 variants on HIV-1-associated opportunistic infections (OIs) is unknown. Here, we report findings from a metaanalysis of four HIV/AIDS prospective cohorts (ALIVE, LSOCA, MACS, and WIHS) including 2066 African American participants. Using a global test combining all four cohorts, carriage of two APOL1 variant alleles is associated with a 50% reduction in odds of OI (combined OR 0.50, 95% CI 0.33-0.76). Subgroup analysis of OI etiological categories (viral, parasitic, fungal and Mycobacterial) suggests the possibility of specific protection from fungal infections (OR 0.54. 95% CI 0.32-0.93; PBonferroni corrected = 0.08). We observe an association of APOL1 variant alleles with host protection against OI in HIV-positive individuals. The study suggests a broader role of APOL1 variant alleles in innate immunity in vivo.
Collapse
Affiliation(s)
- Ping An
- Basic Research Laboratory, Molecular Genetic Epidemiology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| | - Efe Sezgin
- Basic Research Laboratory, Molecular Genetic Epidemiology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Department of Epidemiology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Laboratory of Nutrigenomics and Epidemiology, Izmir Institute of Technology, Izmir, Turkey
| | - Gregory D Kirk
- Department of Epidemiology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Priya Duggal
- Department of Epidemiology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth Binns-Roemer
- Basic Research Laboratory, Molecular Genetic Epidemiology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - George Nelson
- Center for Cancer Research Informatics Core, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Sophie Limou
- Basic Research Laboratory, Molecular Genetic Epidemiology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- CRTI UMR1064, Inserm, Université de Nantes & ITUN, CHU Nantes, Nantes, France
- Ecole Centrale de Nantes, Nantes, France
| | - Mark L Van Natta
- Department of Epidemiology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Douglas A Jabs
- Department of Epidemiology, the Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Ophthalmology, the Wilmer Eye Institute, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michelle Estrella
- Kidney Health Research Collaborative, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- San Francisco VA Health Care System, San Francisco, CA, USA
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Cheryl A Winkler
- Basic Research Laboratory, Molecular Genetic Epidemiology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
52
|
Dong R, Zhang B, Tan B, Lin N. Long non-coding RNAs as the regulators and targets of macrophage M2 polarization. Life Sci 2021; 266:118895. [PMID: 33310042 DOI: 10.1016/j.lfs.2020.118895] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/16/2020] [Accepted: 12/04/2020] [Indexed: 12/30/2022]
Abstract
Macrophages are immune cells with high heterogeneity and plasticity. M2 polarization is one extreme of the well-established phenotypes of macrophage polarization, and involves in diverse biological processes. The polarization process is initiated at the command of numerous components. Long non-coding RNAs (lncRNAs) are RNAs longer than 200 nucleotides with limited protein-coding capacity. Recent studies have revealed a newly found subset of lncRNAs engaged in the M2 polarization and their potent and multifunctional roles in developing diseases. By interfering with specific signaling pathways and altering the active mode, acting as the sponges of microRNAs or decoys of transcription factors, lncRNAs prompted macrophages to an M2 phenotype. Further, lncRNAs can bind to the genome to regulate the chromatin dynamics or work as a platform for protein complexes tether. Exosomal lncRNAs can also orchestrate the polarization in a paracrine way. To make it easier to interpret the roles of lncRNAs in the M2 polarization, we review the reported lncRNAs according to the underlying mechanisms. Moreover, we discuss the possibilities of targeting macrophages' M2 polarization using the oligonucleotides drugs or clustered regularly interspaced palindromic repeats (CRISPR) technologies to provoke wisdom on the therapeutic strategies.
Collapse
Affiliation(s)
- Rong Dong
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Bo Zhang
- Translational Medicine Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 31006, China
| | - Biqin Tan
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Nengming Lin
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Translational Medicine Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 31006, China.
| |
Collapse
|
53
|
Huang XH, Ma Y, Lou H, Chen N, Zhang T, Wu LY, Chen YJ, Zheng MM, Lou YL, Xie DL. The Role of TSC1 in the Macrophages Against Vibrio vulnificus Infection. Front Cell Infect Microbiol 2021; 10:596609. [PMID: 33585271 PMCID: PMC7873526 DOI: 10.3389/fcimb.2020.596609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/08/2020] [Indexed: 12/19/2022] Open
Abstract
Vibrio vulnificus (V. vulnificus) is an estuarine bacterium that is capable of causing rapidly fatal infection in humans. Proper polarization and bactericidal activity of macrophages play essential roles in defending against invading pathogens. How macrophages limit V. vulnificus infection remains not well understood. Here we report that tuberous sclerosis complex 1 (TSC1) is crucial for the regulation of V. vulnificus-induced macrophage polarization, bacterial clearance, and cell death. Mice with myeloid-specific deletion of TSC1 exhibit a significant reduction of survival time after V. vulnificus infection. V. vulnificus infection induces both M1 and M2 polarization. However, TSC1 deficient macrophages show enhanced M1 response to V. vulnificus infection. Interestedly, the absence of TSC1 in myeloid cells results in impaired bacterial clearance both in vivo and in vitro after V. vulnificus infection. Inhibition of the mammalian target of rapamycin (mTOR) activity significantly reverses V. vulnificus-induced hypersensitive M1 response and resistant bactericidal activity both in wild-type and TSC1-deficient macrophages. Moreover, V. vulnificus infection causes cell death of macrophages, possibly contributes to defective of bacterial clearance, which also exhibits in a mTORC1-dependent manner. These findings highlight an essential role for the TSC1-mTOR signaling in the regulation of innate immunity against V. vulnificus infection.
Collapse
Affiliation(s)
- Xian-Hui Huang
- Department of Microbiology and Immunology, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou, China.,Department of Infection and Immunity, Wenzhou Key Laboratory of Sanitary Microbiology, Wenzhou, China
| | - Yao Ma
- Department of Microbiology and Immunology, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou, China.,Department of Laboratory Medicine, Dong Yang People's Hospital, Jinhua, China
| | - Han Lou
- Department of Pathology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Na Chen
- Department of Microbiology and Immunology, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou, China
| | - Ting Zhang
- Department of Laboratory Medicine, Jinshan Hospital of Fudan University, Shanghai, China
| | - Liu-Ying Wu
- Department of Microbiology and Immunology, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou, China
| | - Yi-Ju Chen
- Department of Microbiology and Immunology, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou, China
| | - Meng-Meng Zheng
- Department of Microbiology and Immunology, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou, China
| | - Yong-Liang Lou
- Department of Microbiology and Immunology, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou, China.,Department of Infection and Immunity, Wenzhou Key Laboratory of Sanitary Microbiology, Wenzhou, China
| | - Dan-Li Xie
- Department of Microbiology and Immunology, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou, China.,Department of Infection and Immunity, Wenzhou Key Laboratory of Sanitary Microbiology, Wenzhou, China
| |
Collapse
|
54
|
Ali H, Caballero R, Dong SXM, Gajnayaka N, Vranjkovic A, Ahmed D, Iqbal S, Crawley AM, Angel JB, Cassol E, Kumar A. Selective killing of human M1 macrophages by Smac mimetics alone and M2 macrophages by Smac mimetics and caspase inhibition. J Leukoc Biol 2021; 110:693-710. [PMID: 33404106 DOI: 10.1002/jlb.4a0220-114rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022] Open
Abstract
The inflammatory and anti-inflammatory Mϕs have been implicated in many diseases including rheumatoid arthritis, multiple sclerosis, and leprosy. Recent studies suggest targeting Mϕ function and activation may represent a potential target to treat these diseases. Herein, we investigated the effect of second mitochondria-derived activator of caspases (SMAC) mimetics (SMs), the inhibitors of apoptosis (IAPs) proteins, on the killing of human pro- and anti-inflammatory Mϕ subsets. We have shown previously that human monocytes are highly susceptible whereas differentiated Mϕs (M0) are highly resistant to the cytocidal abilities of SMs. To determine whether human Mϕ subsets are resistant to the cytotoxic effects of SMs, we show that M1 Mϕs are highly susceptible to SM-induced cell death whereas M2a, M2b, and M2c differentiated subsets are resistant, with M2c being the most resistant. SM-induced cell death in M1 Mϕs was mediated by apoptosis as well as necroptosis, activated both extrinsic and intrinsic pathways of apoptosis, and was attributed to the IFN-γ-mediated differentiation. In contrast, M2c and M0 Mϕs experienced cell death through necroptosis following simultaneous blockage of the IAPs and the caspase pathways. Overall, the results suggest that survival of human Mϕs is critically linked to the activation of the IAPs pathways. Moreover, agents blocking the cellular IAP1/2 and/or caspases can be exploited therapeutically to address inflammation-related diseases.
Collapse
Affiliation(s)
- Hamza Ali
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada.,Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Ramon Caballero
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Simon X M Dong
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Niranjala Gajnayaka
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Agatha Vranjkovic
- Chronic Disease Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Duale Ahmed
- Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Salma Iqbal
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Angela M Crawley
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada.,Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Chronic Disease Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Jonathan B Angel
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada.,Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Chronic Disease Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Ashok Kumar
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada.,Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada.,Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| |
Collapse
|
55
|
Liu G, Yan D, Yang L, Sun Y, Zhan L, Lu L, Jin Z, Zhang C, Long P, Chen J, Yuan Q. The effect of miR-471-3p on macrophage polarization in the development of diabetic cardiomyopathy. Life Sci 2021; 268:118989. [PMID: 33417962 DOI: 10.1016/j.lfs.2020.118989] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 12/26/2022]
Abstract
AIMS The imbalance of M1/M2 macrophage ratio promotes the occurrence of diabetic cardiomyopathy (DCM), but the precise mechanisms are not fully understood. The aim of this study was to investigate whether miR-471-3p/silent information regulator 1 (SIRT1) pathway is involved in the macrophage polarization during the development of DCM. METHODS Immunohistochemical staining was used to detect M1 and M2 macrophages infiltration in the heart tissue. Flow cytometry was used to detect the proportion of M1 and M2 macrophages. Expression of miR-471-3p was quantified by real time quantitative-PCR. Transfection of miRNA inhibitor into RAW264.7 cells was performed to investigate the underlying mechanisms. Bioinformatics methods and western blotting were used to explore the target gene of miR-471-3p and further confirmed by dual luciferase reporter assay. KEY FINDINGS We observed that M1 macrophages infiltration in the heart of tissue in DCM while M2 type was decreased. M1/M2 ratio was increased significantly in bone marrow-derived macrophages (BMDMs) from db/db mice and in RAW264.7 cells treated with advanced glycation end products (AGEs). Meanwhile, miR-471-3p was significantly upregulated in RAW264.7 cells induced by AGEs and inhibition of miR-471-3p could reduce the inflammatory polarization of macrophages. Bioinformatics analysis identified SIRT1 as a target of miR-471-3p. Both dual luciferase reporter assay and western blotting verified that miR-471-3p negatively regulated SIRT1 expression. SIRT1 agonist resveratrol could downregulate the increased proportion of M1 macrophages induced by AGEs. CONCLUSION Our results indicated that the development of DCM was related to AGEs-induced macrophage polarized to M1 type through a mechanism involving the miR-471-3p/SIRT1 pathway.
Collapse
Affiliation(s)
- Guangqi Liu
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan Yan
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Liu Yang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, China
| | - Yunwei Sun
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Lin Zhan
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Lili Lu
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhigang Jin
- China Resource & WISCO General Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Chunxiang Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Ping Long
- China Resource & WISCO General Hospital, Wuhan University of Science and Technology, Wuhan, China.
| | - Jinhua Chen
- Department of Pharmacy, Wuhan Asia Heart Hospital, Wuhan University of Science and Technology, Wuhan, Hubei Province, China.
| | - Qiong Yuan
- Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
56
|
Mohamed Elashiry M, Tian F, Elashiry M, Zeitoun R, Elsayed R, Andrews ML, Bergeon BE, Cutler C, Tay F. Enterococcus faecalis shifts macrophage polarization toward M1-like phenotype with an altered cytokine profile. J Oral Microbiol 2021; 13:1868152. [PMID: 33488991 PMCID: PMC7801083 DOI: 10.1080/20002297.2020.1868152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: The macrophage is an innate immune defense cell involved in pathogen recognition and clearance. Aim: In view of the diversity of the macrophage phenotype and function, the present study investigated how Enterococcus faecalis infection affects the differentiation, phenotype and cytokine profile of macrophages. Methods: Murine bone marrow-derived stem cells were co-cultured with E. faecalis before and after differentiation. Macrophage M0 polarization towards M1 or M2 was initiated at day 6 by addition of LPS and INF-γ, or IL-4 and IL-13, respectively. Results: E. faecalis did not inhibit macrophage differentiation and were identified within macrophages. Viability of the macrophages infected with E. faecalis prior to differentiation was enhanced, evidenced by apoptosis inhibition, as was expression of CD38 and IRF5 proteins, indicators of M1-like polarization. These M1-like macrophages expressed an aberrant cytokine mRNA profile, with reduction in inflammatory cytokines IL-1β and IL-12 and increase in regulatory cytokine IL-10. No changes in TNF-α or TGF-β1 were detected, compared with the control groups. This atypical M1-like phenotype was retained even upon stimulation with growth factors that normally trigger their development into M2 macrophages. Conclusions: These findings suggested that E. faecalis infection of bone marrow-derived stem cells during differentiation into macrophages induces an atypical M1-like phenotype associated with intracellular bacterial survival.
Collapse
Affiliation(s)
- Mohamed Mohamed Elashiry
- Department of Endodontics, Faculty of Dentistry, Ain Shams University, Cairo, Egypt.,Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Fucong Tian
- Department of Endodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Mahmoud Elashiry
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Rana Zeitoun
- Department of Fixed Prosthodontics, Faculty of Dentistry, Ain Shams University, Cairo, Egypt.,Department of Oral Biology and Diagnostic Science, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Ranya Elsayed
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Matthew L Andrews
- Department of Endodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Brian E Bergeon
- Department of Endodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Christopher Cutler
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Franklin Tay
- Department of Endodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
57
|
Yasmin H, Varghese PM, Bhakta S, Kishore U. Pathogenesis and Host Immune Response in Leprosy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:155-177. [PMID: 34661895 DOI: 10.1007/978-3-030-67452-6_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Leprosy is an ancient insidious disease caused by Mycobacterium leprae, where the skin and peripheral nerves undergo chronic granulomatous infections, leading to sensory and motor impairment with characteristic deformities. Susceptibility to leprosy and its disease state are determined by the manifestation of innate immune resistance mediated by cells of monocyte lineage. Due to insufficient innate resistance, granulomatous infection is established, influencing the specific cellular immunity. The clinical presentation of leprosy ranges between two stable polar forms (tuberculoid to lepromatous) and three unstable borderline forms. The tuberculoid form involves Th1 response, characterized by a well demarcated granuloma, infiltrated by CD4+ T lymphocytes, containing epitheloid and multinucleated giant cells. In the lepromatous leprosy, there is no characteristic granuloma but only unstructured accumulation of ineffective macrophages containing engulfed pathogens. Th1 response, characterised by IFN-γ and IL-2 production, activates macrophages in order to kill intracellular pathogens. Conversely, a Th2 response, characterized by the production of IL-4, IL-5 and IL-10, helps in antibody production and consequently downregulates the cell-mediated immunity induced by the Th1 response. M. lepare has a long generation time and its inability to grow in culture under laboratory conditions makes its study challenging. The nine-banded armadillo still remains the best clinical and immunological model to study host-pathogen interaction in leprosy. In this chapter, we present cellular morphology and the genomic uniqueness of M. leprae, and how the pathogen shows tropism for Schwann cells, macrophages and dendritic cells.
Collapse
Affiliation(s)
- Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Praveen Mathews Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK.,School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Sanjib Bhakta
- Department of Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck, University of London, London, UK
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| |
Collapse
|
58
|
Wei H, Liu S, Lian R, Huang C, Li Y, Chen L, Zeng Y. Abnormal Expression of Indoleamine 2, 3-Dioxygenase in Human Recurrent Miscarriage. Reprod Sci 2020; 27:1656-1664. [PMID: 32430712 DOI: 10.1007/s43032-020-00196-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Indoleamine 2, 3-dioxygenase (IDO), an immunosuppressive enzyme that mediates the conversion of tryptophan to kynurenine, was shown to play a key role in placental development during normal pregnancy. However, little is known about the pattern of IDO expression in the endometrium and its attendant functional significance in pregnancies complicated with recurrent miscarriage (RM). Immunohistochemical studies of IDO, Foxp3, CD56, and CD163 expression were performed in endometrial samples from women with RM and healthy fertile controls. Our study found that IDO was localized in glandular epithelial cells, surface epithelial cells, and a small number of cells within the stromal compartment (including stromal cells and leukocytes) in endometrium. Indoleamine 2, 3-dioxygenase expression in the RM group was significantly lower than control group. The Foxp3 and CD56 expression were significantly increased with the elevated IDO expression in controls but not in RM. The percentage of Foxp3 + Tregs was significantly correlated with the level of IDO expression in the control group. Comparatively, no correlation was found between the percentage of CD56 + cells, CD163 + cells, and the level of IDO expression, no matter in controls and RM patients. This study demonstrated that the downregulation of IDO expression and noncoordinated association between IDO and other endometrial immune cells were associated with RM. Our findings provide insights into the contribution of IDO in immune regulation to maintain normal pregnancy, which could be used to develop potential therapeutic methods for RM.
Collapse
Affiliation(s)
- Hongxia Wei
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, No. 1001 Fuqiang Road, Futian District, Shenzhen, 518045, China
| | - Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, No. 1001 Fuqiang Road, Futian District, Shenzhen, 518045, China
| | - Ruochun Lian
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, No. 1001 Fuqiang Road, Futian District, Shenzhen, 518045, China
| | - Chunyu Huang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, No. 1001 Fuqiang Road, Futian District, Shenzhen, 518045, China
| | - Yuye Li
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, No. 1001 Fuqiang Road, Futian District, Shenzhen, 518045, China
| | - Lanna Chen
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, No. 1001 Fuqiang Road, Futian District, Shenzhen, 518045, China
| | - Yong Zeng
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, No. 1001 Fuqiang Road, Futian District, Shenzhen, 518045, China.
| |
Collapse
|
59
|
Guo G, Fu R, Zhang L, Chao T, Jiang Z, Dong L, Huang R, Lu L, Yang W, Gu Y, Liang Y, Zheng Q. Gfi1 and Zc3h12c orchestrate a negative feedback loop that inhibits NF-kB activation during inflammation in macrophages. Mol Immunol 2020; 128:219-226. [DOI: 10.1016/j.molimm.2020.10.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/03/2020] [Accepted: 10/27/2020] [Indexed: 01/10/2023]
|
60
|
Yang X, Xie B, Peng H, Shi G, Sreenivas B, Guo J, Wang C, He Y. Eradicating intracellular MRSA via targeted delivery of lysostaphin and vancomycin with mannose-modified exosomes. J Control Release 2020; 329:454-467. [PMID: 33253805 DOI: 10.1016/j.jconrel.2020.11.045] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 10/17/2020] [Accepted: 11/23/2020] [Indexed: 02/08/2023]
Abstract
Intracellular methicillin-resistant Staphylococcus aureus (MRSA) is extremely difficult to remove by common antibiotics, leading to infection recurrence and resistance. Herein we report a novel exosome-based antibiotic delivery platform for eradicating intracellular MRSA, where mannosylated exosome (MExos) is employed as the drug carrier and preferentially taken up by macrophages, delivering lysostaphin (MExoL) and vancomycin (MExoV) to intracellular pathogens. Combination of MExoL and MExoV eradicated intracellular quiescent MRSA. Moreover, MExos rapidly accumulated in mouse liver and spleen, the target organs of intracellular MRSA, after intravenous (IV) administration. Thus, the MExos antibiotic delivery platform is a promising strategy for combating intracellular infection.
Collapse
Affiliation(s)
- Xiaohong Yang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Shuitu Technology Development Zone, Beibei, 400714 Chongqing, China; Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Beibei Xie
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Haibo Peng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Gongming Shi
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Banne Sreenivas
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Jian Guo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China
| | - Chenhui Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China.
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, Shapingba, 401331 Chongqing, China.
| |
Collapse
|
61
|
Chen L, Christian DA, Kochanowsky JA, Phan AT, Clark JT, Wang S, Berry C, Oh J, Chen X, Roos DS, Beiting DP, Koshy AA, Hunter CA. The Toxoplasma gondii virulence factor ROP16 acts in cis and trans, and suppresses T cell responses. J Exp Med 2020; 217:133618. [PMID: 31961916 PMCID: PMC7062521 DOI: 10.1084/jem.20181757] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/10/2019] [Accepted: 11/25/2019] [Indexed: 01/19/2023] Open
Abstract
The ability of Toxoplasma gondii to inject the rhoptry kinase ROP16 into host cells results in the activation of the transcription factors STAT3 and STAT6, but it is unclear how these events impact infection. Here, parasites that inject Cre-recombinase with rhoptry proteins were used to distinguish infected macrophages from those only injected with parasite proteins. Transcriptional profiling revealed that injection of rhoptry proteins alone was sufficient to induce an M2 phenotype that is dependent on STAT3 and STAT6, but only infected cells displayed reduced expression of genes associated with antimicrobial activity and protective immunity. In vivo, the absence of STAT3 or STAT6 improved parasite control, while the loss of ROP16 resulted in a marked reduction in parasite numbers and heightened parasite-specific T cell responses. Thus, ROP16 is a virulence factor that can act in cis and trans to promote M2 programs and which limits the magnitude of parasite-specific T cell responses.
Collapse
Affiliation(s)
- Longfei Chen
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joshua A Kochanowsky
- Department of Neurology and Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, AZ
| | - Anthony T Phan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joseph T Clark
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shuai Wang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Corbett Berry
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jung Oh
- Department of Biology, University of Pennsylvania, Philadelphia, PA
| | - Xiaoguang Chen
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - David S Roos
- Department of Biology, University of Pennsylvania, Philadelphia, PA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Anita A Koshy
- Department of Neurology and Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, AZ
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
62
|
Thiriot JD, Martinez-Martinez YB, Endsley JJ, Torres AG. Hacking the host: exploitation of macrophage polarization by intracellular bacterial pathogens. Pathog Dis 2020; 78:5739920. [PMID: 32068828 DOI: 10.1093/femspd/ftaa009] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
Macrophages play an integral role in host defenses against intracellular bacterial pathogens. A remarkable plasticity allows for adaptation to the needs of the host to orchestrate versatile innate immune responses to a variety of microbial threats. Several bacterial pathogens have adapted to macrophage plasticity and modulate the classical (M1) or alternative (M2) activation bias towards a polarization state that increases fitness for intracellular survival. Here, we summarize the current understanding of the host macrophage and intracellular bacterial interface; highlighting the roles of M1/M2 polarization in host defense and the mechanisms employed by several important intracellular pathogens to modulate macrophage polarization to favor persistence or proliferation. Understanding macrophage polarization in the context of disease caused by different bacterial pathogens is important for the identification of targets for therapeutic intervention.
Collapse
Affiliation(s)
- Joseph D Thiriot
- Department of Microbiology and Immunology , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA
| | - Yazmin B Martinez-Martinez
- Department of Microbiology and Immunology , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA
| | - Janice J Endsley
- Department of Microbiology and Immunology , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA
| | - Alfredo G Torres
- Department of Microbiology and Immunology , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA.,Department of Pathology, University of Texas Medical Branch , University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555 USA
| |
Collapse
|
63
|
Nwizu N, Wactawski-Wende J, Genco RJ. Periodontal disease and cancer: Epidemiologic studies and possible mechanisms. Periodontol 2000 2020; 83:213-233. [PMID: 32385885 PMCID: PMC7328760 DOI: 10.1111/prd.12329] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epidemiologic and cancer control studies on the association of periodontal disease and cancer risk mostly suggest a positive association with overall cancer risk and certain specific types of cancer. These findings are generally consistent among cross‐sectional and longitudinal studies. In this paper, we review epidemiologic studies and current knowledge on periodontal disease and cancer, with a focus on those studies conducted in the years following the Joint European Federation of Periodontology/American Academy of Periodontology Workshop on “Periodontitis and Systemic Diseases” in November 2012. This review also explores the role of chronic inflammation as a biologically plausible mechanistic link between periodontal disease and risk of cancer. Furthermore, it highlights studies that have examined the potential importance of certain periodontal pathogens in this association.
Collapse
Affiliation(s)
- Ngozi Nwizu
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, USA.,School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, USA.,Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, USA
| | - Jean Wactawski-Wende
- School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, USA
| | - Robert J Genco
- Department of Oral Biology, University at Buffalo, The State University of New York, Buffalo, USA
| |
Collapse
|
64
|
Singh R, Dubey V, Wolfson D, Ahmad A, Butola A, Acharya G, Mehta DS, Basnet P, Ahluwalia BS. Quantitative assessment of morphology and sub-cellular changes in macrophages and trophoblasts during inflammation. BIOMEDICAL OPTICS EXPRESS 2020; 11:3733-3752. [PMID: 33014563 PMCID: PMC7510918 DOI: 10.1364/boe.389350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/14/2020] [Accepted: 05/21/2020] [Indexed: 05/06/2023]
Abstract
In pregnancy during an inflammatory condition, macrophages present at the feto-maternal junction release an increased amount of nitric oxide (NO) and pro-inflammatory cytokines such as TNF-α and INF-γ, which can disturb the trophoblast functions and pregnancy outcome. Measurement of the cellular and sub-cellular morphological modifications associated with inflammatory responses are important in order to quantify the extent of trophoblast dysfunction for clinical implication. With this motivation, we investigated morphological, cellular and sub-cellular changes in externally inflamed RAW264.7 (macrophage) and HTR-8/SVneo (trophoblast) using structured illumination microscopy (SIM) and quantitative phase microscopy (QPM). We monitored the production of NO, changes in cell membrane and mitochondrial structure of macrophages and trophoblasts when exposed to different concentrations of pro-inflammatory agents (LPS and TNF-α). In vitro NO production by LPS-induced macrophages increased 22-fold as compared to controls, whereas no significant NO production was seen after the TNF-α challenge. Under similar conditions as with macrophages, trophoblasts did not produce NO following either LPS or the TNF-α challenge. Super-resolution SIM imaging showed changes in the morphology of mitochondria and the plasma membrane in macrophages following the LPS challenge and in trophoblasts following the TNF-α challenge. Label-free QPM showed a decrease in the optical thickness of the LPS-challenged macrophages while TNF-α having no effect. The vice-versa is observed for the trophoblasts. We further exploited machine learning approaches on a QPM dataset to detect and to classify the inflammation with an accuracy of 99.9% for LPS-challenged macrophages and 98.3% for TNF-α-challenged trophoblasts. We believe that the multi-modal advanced microscopy methodologies coupled with machine learning approach could be a potential way for early detection of inflammation.
Collapse
Affiliation(s)
- Rajwinder Singh
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø 9037, Norway
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Author with equal contribution
| | - Vishesh Dubey
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø 9037, Norway
- Department of Physics, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Author with equal contribution
| | - Deanna Wolfson
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Azeem Ahmad
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Ankit Butola
- Department of Physics, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Ganesh Acharya
- Department of Clinical Science, Intervention and Technology Karolinska Univ. Hospital, Sweden
| | - Dalip Singh Mehta
- Department of Physics, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Purusotam Basnet
- Womeńs Health and Perinatology Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway and Department of Obstetrics and Gynecology, University Hospital of North Norway, Tromsø, Norway
| | - Balpreet Singh Ahluwalia
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø 9037, Norway
- Department of Clinical Science, Intervention and Technology Karolinska Univ. Hospital, Sweden
| |
Collapse
|
65
|
Dong SXM, Caballero R, Ali H, Roy DLF, Cassol E, Kumar A. Transfection of hard-to-transfect primary human macrophages with Bax siRNA to reverse Resveratrol-induced apoptosis. RNA Biol 2020; 17:755-764. [PMID: 32050839 PMCID: PMC7577235 DOI: 10.1080/15476286.2020.1730081] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 01/01/2023] Open
Abstract
Small interfering RNA (siRNA) is a critical loss-of-function tool for elucidating the role of genes in biomedical studies. The effective use of siRNA needs transfection technology that delivers siRNA into the correct location of target cells, especially those which are extremely difficult to transfect. Macrophages, which play an important role in the pathogenesis of many diseases, are known to be extremely hard to transfect. Thus, to elucidate the functions of genes in human macrophage biology, it is essential to devise technology for efficient siRNA transfection. However, a fast and efficient method for siRNA transfection in primary human macrophages has not been reported. The siRNA transfection is a tug-of-war between transfection rate and cytotoxicity. A higher transfection rate is generally accompanied with increased cytotoxicity, therefore, choosing a transfection reagent that limits cell death while maintain a desirable transfection rate is important. In this study, we employed auto-analysis function of the IncuCyte® to devise a fast and cost-saving technology for efficient transfection of adherent cells and particularly human macrophages. We show that DharmaFECT3 transfection reagent from Dharmacon was the most efficient in transfecting primary human monocyte-derived macrophages and PMA-differentiated U937 cells, whereas other transfection reagents tested were cytotoxic. This method exhibited approximately 85% transfection efficiency in human macrophages. Moreover, siRNA silencing of Bax with this technique effectively protected primary human macrophages and PMA-differentiated U937 cells against Resveratrol-induced cell death. In addition, this method inherently takes the balance between transfection rate and cytotoxicity of siRNA transfection reagents into consideration.
Collapse
Affiliation(s)
- Simon Xin Min Dong
- Apoptosis Research Center of Children’s Hospital of Eastern Ontario, Department of Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Ramon Caballero
- Apoptosis Research Center of Children’s Hospital of Eastern Ontario, Department of Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Hamza Ali
- Apoptosis Research Center of Children’s Hospital of Eastern Ontario, Department of Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | | | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Ashok Kumar
- Apoptosis Research Center of Children’s Hospital of Eastern Ontario, Department of Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
66
|
Deng S, Zhang L, Mo Y, Huang Y, Li W, Peng Q, Huang L, Ai Y. Mdivi-1 attenuates lipopolysaccharide-induced acute lung injury by inhibiting MAPKs, oxidative stress and apoptosis. Pulm Pharmacol Ther 2020; 62:101918. [PMID: 32251714 DOI: 10.1016/j.pupt.2020.101918] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/12/2020] [Accepted: 04/01/2020] [Indexed: 01/23/2023]
Abstract
Sepsis is among the most devastating events in intensive care units. As a complication of sepsis, acute lung injury (ALI) is common and highly associated with poor outcome. The present study demonstrated that abnormal mitochondrial dynamics play a pivotal role in lipopolysaccharide (LPS)-induced ALI. Inhibiting the mitochondrial fission with the specific inhibitor-1 (Mdivi-1) ameliorated ALI as assessed by hematoxylin and eosin (H&E) staining and wet/dry ratio. Furthermore, Mdivi-1 reduced mitogen-activated protein kinases (MAPKs) activation, oxidative stress and apoptosis in the lungs. Plasma pro-inflammation cytokines were also reduced significantly in Mdivi-1-treated mice. In vitro study revealed that Mdivi-1 protected the macrophages from LPS-induced MAPKs activation, oxidative stress and cell apoptosis. Mdivi-1 also inhibited the release of pro-inflammatory cytokines. Morphological analysis showed that Mdivi-1 rescued the macrophages from LPS-induced mitochondrial fragmentation. Moreover, LPS treatment induced significant phosphorylation of Drp1 at Ser616, dephosphorylation at Ser637 and translocation of Drp1 from the cytoplasm to mitochondria, while Mdivi-1 inhibited those effects. Thus, modification of fission to rebuild mitochondrial homeostasis may offer an innovative opportunity for developing therapeutic strategies against ALI.
Collapse
Affiliation(s)
- Songyun Deng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| | - Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| | - Yunan Mo
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| | - Yan Huang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| | - Wenchao Li
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| | - Qianyi Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| | - Li Huang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| | - Yuhang Ai
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, PR China.
| |
Collapse
|
67
|
Nikonova A, Khaitov M, Jackson DJ, Traub S, Trujillo-Torralbo MB, Kudlay DA, Dvornikov AS, Del-Rosario A, Valenta R, Stanciu LA, Khaitov R, Johnston SL. M1-like macrophages are potent producers of anti-viral interferons and M1-associated marker-positive lung macrophages are decreased during rhinovirus-induced asthma exacerbations. EBioMedicine 2020; 54:102734. [PMID: 32279057 PMCID: PMC7152663 DOI: 10.1016/j.ebiom.2020.102734] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Macrophages (Mф) can be M1/M2 polarized by Th1/2 signals, respectively. M2-like Mф are thought to be important in asthma pathogenesis, and M1-like in anti-infective immunity, however their roles in virus-induced asthma exacerbations are unknown. Our objectives were (i) to assess polarised Mф phenotype responses to rhinovirus (RV) infection in vitro and (ii) to assess Mф phenotypes in healthy subjects and people with asthma before and during experimental RV infection in vivo. METHODS We investigated characteristics of polarized/unpolarized human monocyte-derived Mф (MDM, from 3-6 independent donors) in vitro and evaluated frequencies of M1/M2-like bronchoalveolar lavage (BAL) Mф in experimental RV-induced asthma exacerbation in 7 healthy controls and 17 (at baseline) and 18 (at day 4 post infection) people with asthma. FINDINGS We observed in vitro: M1-like but not M2-like or unpolarized MDM are potent producers of type I and III interferons in response to RV infection (P<0.0001), and M1-like are more resistant to RV infection (P<0.05); compared to M1-like, M2-like MDM constitutively produced higher levels of CCL22/MDC (P = 0.007) and CCL17/TARC (P<0.0001); RV-infected M1-like MDM were characterized as CD14+CD80+CD197+ (P = 0.002 vs M2-like, P<0.0001 vs unpolarized MDM). In vivo we found reduced percentages of M1-like CD14+CD80+CD197+ BAL Mф in asthma during experimental RV16 infection compared to baseline (P = 0.024). INTERPRETATION Human M1-like BAL Mф are likely important contributors to anti-viral immunity and their numbers are reduced in patients with allergic asthma during RV-induced asthma exacerbations. This mechanism may be one explanation why RV-triggered clinical and pathologic outcomes are more severe in allergic patients than in healthy subjects. FUNDING ERC FP7 Advanced grant 233015, MRC Centre Grant G1000758, Asthma UK grant 08-048, NIHR Biomedical Research Centre funding scheme, NIHR BRC Centre grant P26095, the Predicta FP7 Collaborative Project grant 260895, RSF grant 19-15-00272, Megagrant No 14.W03.31.0024.
Collapse
Affiliation(s)
- Alexandra Nikonova
- National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom; MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Norfolk Place, London W2 1PG, United Kingdom; NRC Institute of Immunology FMBA, Kashirskoe shosse 24, 115478 Moscow, Russian Federation; Mechnikov Research Institute for Vaccines and Sera, M. Kazenny per., 5A, 105064 Moscow, Russian Federation.
| | - Musa Khaitov
- NRC Institute of Immunology FMBA, Kashirskoe shosse 24, 115478 Moscow, Russian Federation.
| | - David J Jackson
- National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom; Imperial College Healthcare NHS Trust, Norfolk Place, London W2 1PG, United Kingdom; MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Norfolk Place, London W2 1PG, United Kingdom.
| | - Stephanie Traub
- National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom; MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Norfolk Place, London W2 1PG, United Kingdom.
| | - Maria-Belen Trujillo-Torralbo
- National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom; Imperial College Healthcare NHS Trust, Norfolk Place, London W2 1PG, United Kingdom
| | - Dmitriy A Kudlay
- NRC Institute of Immunology FMBA, Kashirskoe shosse 24, 115478 Moscow, Russian Federation
| | - Anton S Dvornikov
- Pirogov Russian National Research Medical University, Ostrovitianov str. 1, 117513 Moscow, Russian Federation.
| | - Ajerico Del-Rosario
- National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom; Imperial College Healthcare NHS Trust, Norfolk Place, London W2 1PG, United Kingdom.
| | - Rudolf Valenta
- NRC Institute of Immunology FMBA, Kashirskoe shosse 24, 115478 Moscow, Russian Federation; Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | - Luminita A Stanciu
- National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom; MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Norfolk Place, London W2 1PG, United Kingdom.
| | - Rahim Khaitov
- NRC Institute of Immunology FMBA, Kashirskoe shosse 24, 115478 Moscow, Russian Federation.
| | - Sebastian L Johnston
- National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom; Imperial College Healthcare NHS Trust, Norfolk Place, London W2 1PG, United Kingdom; MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Norfolk Place, London W2 1PG, United Kingdom.
| |
Collapse
|
68
|
Radhakrishnan RK, Thandi RS, Tripathi D, Paidipally P, McAllister MK, Mulik S, Samten B, Vankayalapati R. BCG vaccination reduces the mortality of Mycobacterium tuberculosis-infected type 2 diabetes mellitus mice. JCI Insight 2020; 5:133788. [PMID: 32161191 DOI: 10.1172/jci.insight.133788] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetes is a significant risk factor for the development of active tuberculosis. In this study, we used a mouse model of type 2 diabetes mellitus (T2DM) to determine the effect of prior Bacillus Calmette-Guérin (BCG) vaccination on immune responses to Mycobacterium tuberculosis (Mtb) infection. We found that, at 6-7 months after Mtb infection, 90% of the Mtb-infected T2DM mice died, whereas only 50% of BCG-vaccinated T2DM-Mtb-infected mice died. Moreover, 40% of the PBS-treated uninfected T2DM mice and 30% of the uninfected BCG-vaccinated T2DM mice died, whereas all uninfected and infected nondiabetic mice survived. BCG vaccination was less effective in reducing the lung bacterial burden of Mtb-infected T2DM mice compared with Mtb-infected nondiabetic mice. BCG vaccination significantly reduced lung inflammation in Mtb-infected T2DM mice compared with that of unvaccinated T2DM mice infected with Mtb. Furthermore, reduced mortality of BCG-vaccinated Mtb-infected T2DM mice is associated with expansion of IL-13-producing CXCR3+ Tregs in the lungs of Mtb-infected T2DM mice. Recombinant IL-13 and Tregs from BCG-vaccinated Mtb-infected T2DM mice converted proinflammatory M1 macrophages to antiinflammatory M2 macrophages. Our findings suggest a potentially novel role for BCG in preventing excess inflammation and mortality in T2DM mice infected with Mtb.
Collapse
|
69
|
Use of Human Monocyte-Derived Macrophages to Study Neisseria gonorrhoeae Infection. Methods Mol Biol 2020. [PMID: 31119631 DOI: 10.1007/978-1-4939-9496-0_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Macrophages are critical cells in the innate immune response to microorganisms sensed in the tissues. During infections, the interaction between pathogens and macrophages leads to a macrophage response that includes cytokine production, antigen processing and presentation in the context of MHC molecules, expression of T cell costimulatory molecules and recruitment of innate defense effectors, which results in clearance of infection. However, Neisseria gonorrhoeae can suppress the protective immune response at this level, avoiding its detection and elimination. Studies addressed to develop the interactions between macrophages and Neisseria gonorrhoeae allow us to find potential targets to be exploited with vaccines and therapeutic drugs. In this chapter, we describe protocols to generate human monocyte-derived macrophages and assess their response to infection with Neisseria gonorrhoeae.
Collapse
|
70
|
Kong D, Guo H, Lu Z, Cui J. MicroRNA-21 Mediates the Inhibiting Effect of Praziquantel on NLRP3 Inflammasome in Schistosoma japonicum Infection. Front Vet Sci 2020; 6:517. [PMID: 32118052 PMCID: PMC7029728 DOI: 10.3389/fvets.2019.00517] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/31/2019] [Indexed: 12/21/2022] Open
Abstract
Praziquantel (PZQ), a traditional helminthicide drug, has been shown to exert an anti-inflammatory effect on splenomegaly induced by schistosomiasis via regulating macrophage polarization. Meanwhile, miR-21 has been demonstrated to control macrophage polarization. However, the role of miR-21 in the regulation of macrophage polarization by PZQ in schistosomiasis is still unclear. In the present study, we found that M1-type macrophages were the predominant splenic macrophages in chronic schistosomiasis and that NLRP3 inflammasome–related molecules were upregulated. PZQ inhibited NLRP3 inflammasome in M1 macrophages and reduced the expression of miR-21. Furthermore, using the methods of quantitative real-time PCR and transfection, the downregulation of NLRP3/IL-1β by PZQ in M1 macrophages were reversed by miR-21 overexpression. These results indicated that miR-21 was involved in the inhibiting effect of PZQ on activation of NLRP3 inflammasome. Moreover, miR-21 might target Smad7 to mediate the anti-inflammatory effect of PZQ in polarized macrophages. This study provides an in-depth mechanism of PZQ in the treatment of schistosomiasis.
Collapse
Affiliation(s)
- Delong Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Hongfei Guo
- Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Zhongkui Lu
- Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Jie Cui
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
71
|
Song R, Lin L. Glycoprotein Nonmetastatic Melanoma Protein B (GPNMB) Ameliorates the Inflammatory Response in Periodontal Disease. Inflammation 2020; 42:1170-1178. [PMID: 30793225 DOI: 10.1007/s10753-019-00977-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a type I transmembrane protein that can modulate osteoblasts and bone mineralization. Periodontal disease (PD) is characterized by gum inflammation, alveolar bone resorption, and tooth loss. In this study, we found that GPNMB is highly expressed in inflamed periodontal tissue through microarray and immunohistochemistry (IHC) assays. The role of GPNMB in the pathogenesis of PD was evaluated with primary human periodontal ligament cells (hPDLCs) treated with lipopolysaccharide (LPS) and a GPNMB-expressing lentivirus (lenti-GP). In the hPDLCs treated with LPS and lenti-GP, the expression of tumor necrosis factor (TNF)-α and interleukin (IL)-6 was suppressed and that of IL-10 was upregulated. GPNMB significantly decreased apoptosis in the hPDLCs treated with LPS. GPNMB could upregulate the expression of Jumonji domain-containing protein 3 (Jmjd3), a histone 3 lysine 27 (H3K27) demethylase that is linked to the modulation of the inflammatory response and apoptosis. Taken together, our data find that GPNMB is highly expressed in gum tissue with PD and may be an anti-inflammatory player in the pathogenesis of PD.
Collapse
Affiliation(s)
- Rong Song
- Department of Prosthodontics, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Department of Microbiology, Harbin Medical University, Harbin, 150081, China
| | - Lexun Lin
- Department of Microbiology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
72
|
Zhang X, Yang Y, Zhao Y. Macrophage phenotype and its relationship with renal function in human diabetic nephropathy. PLoS One 2019; 14:e0221991. [PMID: 31509552 PMCID: PMC6738594 DOI: 10.1371/journal.pone.0221991] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 05/30/2019] [Indexed: 12/30/2022] Open
Abstract
This study aimed to examine the macrophage phenotype and its relationship to renal function and histological changes in human DN and the effect of TREM-1 on high-glucose-induced macrophage activation. We observed that in renal tissue biopsies, the expression of CD68 and M1 was apparent in the glomeruli and interstitium, while accumulation of M2 and TREM-1 was primarily observed in the interstitium. The numbers of CD68, M1, and M2 macrophages infiltrating in the DN group were increased in a process-dependent manner compared with the control group, and the intensities of the infiltrates were proportional to the rate of subsequent decline in renal function. M1 macrophages were recruited into the kidney at an early stage (I+IIa) of DN. The M1-to-M2 macrophage ratio peaked at this time, whereas M2 macrophages predominated at later time points (III) when the percentage of M1/M2 macrophages was at its lowest level. In an in vitro study, we showed that under high glucose conditions, macrophages began to up-regulate their expression of TREM-1, M1, and marker iNOS and decreased the M2 marker MR. However, the above effects of high-glucose were abolished when TREM-1 expression was inhibited by TREM-1 siRNA. In conclusion, our study demonstrated that there was a positive correlation between the M1/M2 activation state and the progress of DN, and TREM-1 played an important role in high-glucose-induced macrophage phenotype transformation.
Collapse
Affiliation(s)
- Xiaoliang Zhang
- Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, China
- * E-mail:
| | - Ying Yang
- Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, China
| | - Yu Zhao
- Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
73
|
de Sousa JR, Da Costa Vasconcelos PF, Quaresma JAS. Functional aspects, phenotypic heterogeneity, and tissue immune response of macrophages in infectious diseases. Infect Drug Resist 2019; 12:2589-2611. [PMID: 31686866 PMCID: PMC6709804 DOI: 10.2147/idr.s208576] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/05/2019] [Indexed: 12/13/2022] Open
Abstract
Macrophages are a functionally heterogeneous group of cells with specialized functions depending not only on their subgroup but also on the function of the organ or tissue in which the cells are located. The concept of macrophage phenotypic heterogeneity has been investigated since the 1980s, and more recent studies have identified a diverse spectrum of phenotypic subpopulations. Several types of macrophages play a central role in the response to infectious agents and, along with other components of the immune system, determine the clinical outcome of major infectious diseases. Here, we review the functions of various macrophage phenotypic subpopulations, the concept of macrophage polarization, and the influence of these cells on the evolution of infections. In addition, we emphasize their role in the immune response in vivo and in situ, as well as the molecular effectors and signaling mechanisms used by these cells. Furthermore, we highlight the mechanisms of immune evasion triggered by infectious agents to counter the actions of macrophages and their consequences. Our aim here is to provide an overview of the role of macrophages in the pathogenesis of critical transmissible diseases and discuss how elucidation of this relationship could enhance our understanding of the host-pathogen association in organ-specific immune responses.
Collapse
Affiliation(s)
- Jorge Rodrigues de Sousa
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
| | - Pedro Fernando Da Costa Vasconcelos
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
| | - Juarez Antonio Simões Quaresma
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
- School of Medicine, São Paulo University, São Paulo, SP, Brazil
| |
Collapse
|
74
|
Chen H, Mao X, Meng X, Li Y, Feng J, Zhang L, Zhang Y, Wang Y, Yu Y, Xie K. Hydrogen alleviates mitochondrial dysfunction and organ damage via autophagy‑mediated NLRP3 inflammasome inactivation in sepsis. Int J Mol Med 2019; 44:1309-1324. [PMID: 31432098 PMCID: PMC6713420 DOI: 10.3892/ijmm.2019.4311] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 07/26/2019] [Indexed: 12/17/2022] Open
Abstract
Sepsis is a highly heterogeneous syndrome that is caused by a dysregulated host response to infection. The disproportionate inflammatory response to invasive infection is a triggering event inducing sepsis. The activation of inflammasomes in sepsis can amplify inflammatory responses. It has been reported that damaged mitochondria contribute to NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome-related sepsis. Our previous study revealed that hydrogen (H2) exerts anti-inflammatory effects in sepsis but the detailed mechanism remains to be elucidated. In the present study, septic mice induced by cecal ligation and puncture (CLP) and macrophages induced by lipopolysaccha-ride (LPS) were used as models of sepsis in vivo and in vitro, respectively. An inducer and inhibitor of autophagy and the NLRP3 inflammasome were administered to investigate the detailed mechanism of action of H2 treatment in sepsis. The results demonstrated that LPS and ATP led to NLRP3 inflammasome pathway activation, excessive cytokine release, mitochondrial dysfunction and the activation of autophagy. CLP induced organ injury and NLRP3 pathway activation. H2 treatment ameliorated vital organ damage, the inflammatory response, mitochondrial dysfunction and NLRP3 pathway activation, and promoted autophagy in macrophages induced by LPS and in CLP mice. However, the inhibitor of autophagy and the inducer of NLRP3 reversed the protective effect of H2 against organ damage, the inflammatory response and mitochondrial dysfunction in vivo and in vitro. Collectively, the results demonstrated that H2 alleviated mitochondrial dysfunction and cytokine release via autophagy-mediated NLRP3 inflammasome inactivation.
Collapse
Affiliation(s)
- Hongguang Chen
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Xing Mao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Xiaoyin Meng
- Department of Gynecology and Obstetrics, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Yuan Li
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Jingcheng Feng
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yang Zhang
- Department of Anesthesiology, Tianjin Fourth Center Hospital, Tianjin 300140, P.R. China
| | - Yaoqi Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
75
|
Abstract
Macrophages are cellular components of the immune system that are essential for responding to pathogens, initiating inflammation and maintaining tissue homeostasis. Isolation, culture, and functional characterization of bone marrow-derived macrophages from mice are exceptionally powerful in vitro techniques used to examine aspects of macrophage biology, including effector functions, such as phagocytosis, cytokine secretion, oxidative burst, migration, and antigen processing and presentation. These studies can be carried out using wild-type, gene-ablated, and/or transgenic mice. The quantity, purity, and ease of culture of these cells enhance their utility for primary cell cultures to understand macrophage biology. Mouse macrophages have become a cognate animal model for the study of human macrophage biology and disease. This chapter outlines protocols used to generate, polarize, quantitate, and functionally evaluate macrophages derived from bone marrow precursor cells.
Collapse
Affiliation(s)
- Beckley K Davis
- Department of Biology, Franklin & Marshall College, Lancaster, PA, USA.
| |
Collapse
|
76
|
Desruelle AV, Louge P, Richard S, Blatteau JE, Gaillard S, De Maistre S, David H, Risso JJ, Vallée N. Demonstration by Infra-Red Imaging of a Temperature Control Defect in a Decompression Sickness Model Testing Minocycline. Front Physiol 2019; 10:933. [PMID: 31396102 PMCID: PMC6668502 DOI: 10.3389/fphys.2019.00933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022] Open
Abstract
The prevention, prognosis and resolution of decompression sickness (DCS) are not satisfactory. The etiology of DCS has highlighted thrombotic and inflammatory phenomena that could cause severe neurological disorders or even death. Given the immunomodulatory effects described for minocycline, an antibiotic in widespread use, we have decided to explore its effects in an experimental model for decompression sickness. 40 control mice (Ctrl) and 40 mice treated orally with 90 mg/kg of minocycline (MINO) were subjected to a protocol in a hyperbaric chamber, compressed with air. The purpose was to mimic a scuba dive to a depth of 90 msw and its pathogenic decompression phase. Clinical examinations and blood counts were conducted after the return to the surface. For the first time they were completed by a simple infrared (IR) imaging technique in order to assess feasibility and its clinical advantage in differentiating the sick mice (DCS) from the healthy mice (NoDCS). In this tudy, exposure to the hyperbaric protocol provoked a reduction in the number of circulating leukocytes. DCS in mice, manifesting itself by paralysis or convulsion for example, is also associated with a fall in platelets count. Cold areas ( < 25°C) were detected by IR in the hind paws and tail with significant differences (p < 0.05) between DCS and NoDCS. Severe hypothermia was also shown in the DCS mice. The ROC analysis of the thermograms has made it possible to determine that an average tail temperature below 27.5°C allows us to consider the animals to be suffering from DCS (OR = 8; AUC = 0.754, p = 0.0018). Minocycline modulates blood analysis and it seems to limit the mobilization of monocytes and granulocytes after the provocative dive. While a higher proportion of mice treated with minocycline experienced DCS symptoms, there is no significant difference. The infrared imaging has made it possible to show severe hypothermia. It suggests an modification of thermregulation in DCS animals. Surveillance by infrared camera is fast and it can aid the prognosis in the case of decompression sickness in mice.
Collapse
Affiliation(s)
- Anne-Virginie Desruelle
- Unité Environnements Extrêmes, Département Environnement Opérationnel, Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle, Toulon, France
| | - Pierre Louge
- Service de Médecine Hyperbare et Expertise Plongée, Hôpital d'Instruction des Armées, Toulon, France
| | | | - Jean-Eric Blatteau
- Unité Environnements Extrêmes, Département Environnement Opérationnel, Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle, Toulon, France.,Service de Médecine Hyperbare et Expertise Plongée, Hôpital d'Instruction des Armées, Toulon, France
| | | | - Sébastien De Maistre
- Service de Médecine Hyperbare et Expertise Plongée, Hôpital d'Instruction des Armées, Toulon, France
| | - Hélène David
- Apricot Inhalotherapeutics, Saint-Laurent-de-l'Île-d'Orléans, QC, Canada
| | - Jean-Jacques Risso
- Unité Environnements Extrêmes, Département Environnement Opérationnel, Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle, Toulon, France
| | - Nicolas Vallée
- Unité Environnements Extrêmes, Département Environnement Opérationnel, Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle, Toulon, France
| |
Collapse
|
77
|
Mu Y, Patters BJ, Midde NM, He H, Kumar S, Cory TJ. Tobacco and Antiretrovirals Modulate Transporter, Metabolic Enzyme, and Antioxidant Enzyme Expression and Function in Polarized Macrophages. Curr HIV Res 2019; 16:354-363. [PMID: 30706821 PMCID: PMC6446460 DOI: 10.2174/1570162x17666190130114531] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/24/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cigarette smoking increases systemic oxidative stress, inflammation, and viral replication in individuals with HIV. Macrophages are infected during HIV infection and serve as an important reservoir throughout the process. Macrophages exist in two phenotypes, the classically activated M1 macrophage and alternatively activated M2 macrophage. The expression of drug efflux transporters and metabolic enzymes, which have direct effects on intracellular drug concentrations, differ between the pro-inflammatory M1 macrophage and the anti-inflammatory M2 macrophage. OBJECTIVE To further explain the role of tobacco use in worsened outcomes in the HIV + population receiving antiretroviral therapy. METHODS Western blotting was used to examine macrophage polarization and expression of drug efflux transporters, CYP enzymes, and antioxidant enzymes. The arginase assay was used to measure arginase activity. Cytokine production was measured using the human multiplex inflammatory cytokine assay kit. The 8-OHdG DNA Damage Quantification Direct Kit was used to quantify DNA damage. Viral replication under the influence of tobacco and antiretroviral drug use was measured by p24 Elisa. RESULTS We observed phenotypic shifts from M1 to M2 with both individual and combination treatments with cigarette smoke condensate and the protease inhibitor antiretroviral drug lopinavir. These shifts lead to changes in cytokine production, the expression of CYP enzymes, anti-oxidant enzymes, and drug efflux transporters, as well as changes in viral replication. CONCLUSION This data suggest a mechanism by which tobacco use impairs HIV antiretroviral therapy to increase intracellular drug concentrations in this important cellular reservoir.
Collapse
Affiliation(s)
- Ying Mu
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Benjamin J Patters
- Department of Pharmaceutical Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Narasimha M Midde
- Department of Pharmaceutical Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Hui He
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Santosh Kumar
- Department of Pharmaceutical Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Theodore J Cory
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| |
Collapse
|
78
|
Lu S, Li D, Xi L, Calderone R. Interplay of interferon-gamma and macrophage polarization during Talaromyces marneffei infection. Microb Pathog 2019; 134:103594. [PMID: 31199985 DOI: 10.1016/j.micpath.2019.103594] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/29/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022]
Abstract
Talaromyces marneffei is an increasingly destructive dimorphic fungal pathogen in clinical settings that can cause lethal Talaromycosis. The activation of macrophages is known to be important for host defenses against T. marneffei, and these macrophages are known to be activated in two ways (polarization), known as M1 and M2. We investigated the plasticity of these polarizations, in order to understand if cross-conversion of macrophages may be possible even after they have been programmed. We conducted in vitro experiments using a murine macrophage cell line to investigate the ability of T. marneffei to activate these polarizations. The pre-polarized (M0) macrophage subsets were challenged with LPS as a control, and the sets of M1 markers (iNOS and CD86) and M2 markers (Arg-1 and CD206) were assessed for a possible cross-conversion among M1, M2 and M0 (unstimulated) populations. We found that either conidia or yeast forms of T. marneffei initiate the repression of Arg-1 in M2 cells with no change in the M1 subtype marker molecule iNOS. However, an additional IFN-γ stimulus caused the three macrophage groups to fully exhibit an LPS-induced M2 suppression and a shift to M1 from M0 and M2. We conclude that the conversion of macrophages is required for maintenance of sufficient iNOS production against this organism in the host. The cytokine environment is the key factor that manipulates the plasticity changes among macrophage subtypes. Furthermore, IFN-γ is a crucial host defense factor against pathogenic T. marneffei that has significant therapeutic potential to promote an M1 polarization phenotype.
Collapse
Affiliation(s)
- Sha Lu
- Department of Dermatology and Venereology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Dongmei Li
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Liyan Xi
- Department of Dermatology and Venereology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Richard Calderone
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States.
| |
Collapse
|
79
|
Khan A, Singh VK, Hunter RL, Jagannath C. Macrophage heterogeneity and plasticity in tuberculosis. J Leukoc Biol 2019; 106:275-282. [PMID: 30938876 DOI: 10.1002/jlb.mr0318-095rr] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022] Open
Abstract
Macrophages are the primary host cells for Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), during its intracellular survival in humans. The pathogen has a remarkable capacity to survive within the hostile environment of macrophages. However, primary infection does not result in active TB disease in most individuals. The majority of individuals remain latently infected, wherein the bacteria are held in check by the host immune response. Nevertheless, such individuals can develop active TB later upon the decline in their immune status. In contrast, in a small fraction of infected individuals, the host immune response fails to control the growth of M. tuberculosis bacilli, and granulomatous TB develops progressively. Elucidating the molecular and phenotypic events that govern the outcome of the infection within macrophages is fundamental to understanding the key features of these cells that could be equally critical in infection control. The molecular details of the M. tuberculosis-macrophage interaction continue to be discerned, and emerging evidence suggests that macrophage population that participate in infection is heterogeneous. While the local environment and developmental origin could influence the phenotypic heterogeneity and functional plasticity of macrophages, M. tuberculosis has also been demonstrated to modulate the polarization of macrophages. In this review, we draw on work investigating specialized macrophage populations and their interactions with M. tuberculosis with respect to pathogenesis and specific immune responses. Understanding the mechanisms that control the repertoire of macrophage phenotypes and behaviors during infection may provide prospects for novel TB control strategies through modulation of immunobiological functions of macrophages.
Collapse
Affiliation(s)
- Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Vipul Kumar Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Robert L Hunter
- Department of Pathology and Laboratory Medicine, McGovern Medical School, University of Texas Health Sciences Center-Houston, Houston, Texas, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
80
|
Curto P, Santa C, Allen P, Manadas B, Simões I, Martinez JJ. A Pathogen and a Non-pathogen Spotted Fever Group Rickettsia Trigger Differential Proteome Signatures in Macrophages. Front Cell Infect Microbiol 2019; 9:43. [PMID: 30895174 PMCID: PMC6414445 DOI: 10.3389/fcimb.2019.00043] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/11/2019] [Indexed: 12/13/2022] Open
Abstract
We have previously reported that Rickettsia conorii and Rickettsia montanensis have distinct intracellular fates within THP-1 macrophages, suggesting that the ability to proliferate within macrophages may be a distinguishable factor between pathogenic and non-pathogenic Spotted fever group (SFG) members. To start unraveling the molecular mechanisms underlying the capacity (or not) of SFG Rickettsia to establish their replicative niche in macrophages, we have herein used quantitative proteomics by SWATH-MS to profile the alterations resulted by the challenge of THP-1 macrophages with R. conorii and R. montanensis. We show that the pathogenic, R. conorii, and the non-pathogenic, R. montanensis, member of SFG Rickettsia trigger differential proteomic signatures in macrophage-like cells upon infection. R. conorii specifically induced the accumulation of several enzymes of the tricarboxylic acid cycle, oxidative phosphorylation, fatty acid β-oxidation, and glutaminolysis, as well as of several inner and outer membrane mitochondrial transporters. These results suggest a profound metabolic rewriting of macrophages by R. conorii toward a metabolic signature of an M2-like, anti-inflammatory activation program. Moreover, several subunits forming the proteasome and immunoproteasome are found in lower abundance upon infection with both rickettsial species, which may help bacteria to escape immune surveillance. R. conorii-infection specifically induced the accumulation of several host proteins implicated in protein processing and quality control in ER, suggesting that this pathogenic Rickettsia may be able to increase the ER protein folding capacity. This work reveals novel aspects of macrophage-Rickettsia interactions, expanding our knowledge of how pathogenic rickettsiae explore host cells to their advantage.
Collapse
Affiliation(s)
- Pedro Curto
- PhD Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Cátia Santa
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Paige Allen
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Isaura Simões
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Juan J. Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
81
|
Wei H, Liu S, Lian R, Huang C, Li Y, Chen L, Zeng Y. Abnormal Expression of Indoleamine 2, 3-Dioxygenase in Human Recurrent Miscarriage. Reprod Sci 2019:1933719119833788. [PMID: 30832549 DOI: 10.1177/1933719119833788] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Indoleamine 2, 3-dioxygenase (IDO), an immunosuppressive enzyme that mediates the conversion of tryptophan to kynurenine, was shown to play a key role in placental development during normal pregnancy. However, little is known about the pattern of IDO expression in the endometrium and its attendant functional significance in pregnancies complicated with recurrent miscarriage (RM). Immunohistochemical studies of IDO, Foxp3, CD56, and CD163 expression were performed in endometrial samples from women with RM and healthy fertile controls. Our study found that IDO was localized in glandular epithelial cells, surface epithelial cells, and a small number of cells within the stromal compartment (including stromal cells and leukocytes) in endometrium. Indoleamine 2, 3-dioxygenase expression in the RM group was significantly lower than control group. The Foxp3 and CD56 expression were significantly increased with the elevated IDO expression in controls but not in RM. The percentage of Foxp3+ Tregs was significantly correlated with the level of IDO expression in the control group. Comparatively, no correlation was found between the percentage of CD56+ cells, CD163+ cells, and the level of IDO expression, no matter in controls and RM patients. This study demonstrated that the downregulation of IDO expression and noncoordinated association between IDO and other endometrial immune cells were associated with RM. Our findings provide insights into the contribution of IDO in immune regulation to maintain normal pregnancy, which could be used to develop potential therapeutic methods for RM.
Collapse
Affiliation(s)
- Hongxia Wei
- 1 Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
- These authors contributed equally to this work
| | - Su Liu
- 1 Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
- These authors contributed equally to this work
| | - Ruochun Lian
- 1 Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Chunyu Huang
- 1 Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Yuye Li
- 1 Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Lanna Chen
- 1 Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Yong Zeng
- 1 Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| |
Collapse
|
82
|
Pemmari A, Leppänen T, Paukkeri EL, Scotece M, Hämäläinen M, Moilanen E. Attenuating Effects of Nortrachelogenin on IL-4 and IL-13 Induced Alternative Macrophage Activation and on Bleomycin-Induced Dermal Fibrosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:13405-13413. [PMID: 30458613 DOI: 10.1021/acs.jafc.8b03023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Excessive alternative macrophage activation contributes to fibrosis. We studied the effects of nortrachelogenin, the major lignan component of Pinus sylvestris knot extract, on alternative (M2) macrophage activation. J774 murine and THP-1 human macrophages were cultured with IL-4+IL-13 to induce alternative activation, together with the extract and its components. Effects of nortrachelogenin were also studied in bleomycin-induced murine dermal fibrosis model. Knot extract significantly decreased the expression of alternative activation markers-arginase 1 in murine macrophages (97.4 ± 1.3% inhibition at 30 μg/mL) and CCL13 and PDGF in human macrophages-as did nortrachelogenin (94.9 ± 2.4% inhibition of arginase 1 at 10 μM). Nortrachelogenin also decreased PPARγ expression but had no effect on STAT6 phosphorylation. In vivo, nortrachelogenin reduced bleomycin-induced increase in skin thickness as well as the expression of collagens COL1A1, COL1A2, and COL3A1 (all by >50%). In conclusion, nortrachelogenin suppressed IL-4+IL-13-induced alternative macrophage activation and ameliorated bleomycin-induced fibrosis, indicating therapeutic potential in fibrosing conditions.
Collapse
Affiliation(s)
- Antti Pemmari
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology , Tampere University and Tampere University Hospital , Tampere , Finland
| | - Tiina Leppänen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology , Tampere University and Tampere University Hospital , Tampere , Finland
| | - Erja-Leena Paukkeri
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology , Tampere University and Tampere University Hospital , Tampere , Finland
| | - Morena Scotece
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology , Tampere University and Tampere University Hospital , Tampere , Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology , Tampere University and Tampere University Hospital , Tampere , Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology , Tampere University and Tampere University Hospital , Tampere , Finland
| |
Collapse
|
83
|
Escobar A, Rodas PI, Acuña-Castillo C. Macrophage- Neisseria gonorrhoeae Interactions: A Better Understanding of Pathogen Mechanisms of Immunomodulation. Front Immunol 2018; 9:3044. [PMID: 30627130 PMCID: PMC6309159 DOI: 10.3389/fimmu.2018.03044] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/10/2018] [Indexed: 12/19/2022] Open
Abstract
Neisseria gonorrhoeae is a significant health problem worldwide due to multi-drug resistance issues and absence of an effective vaccine. Patients infected with N. gonorrhoeae have not shown a better immune response in successive infections. This might be explained by the fact that N. gonorrhoeae possesses several mechanisms to evade the innate and adaptative immune responses at different levels. Macrophages are a key cellular component in the innate immune response against microorganisms. The current information suggests that gonococcus can hijack the host response by mechanisms that involve the control of macrophages activity. In this mini review, we intend to condense the recent knowledge on the macrophage–N. gonorrhoeae interactions with a focus on strategies developed by gonococcus to evade or to exploit immune response to establish a successful infection. Finally, we discuss the opportunities and challenges of therapeutics for controlling immune manipulation by N. gonorrhoeae.
Collapse
Affiliation(s)
- Alejandro Escobar
- Laboratorio Biología Celular y Molecular, Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Paula I Rodas
- Laboratorio de Microbiología Médica y Patogénesis, Facultad de Medicina, Universidad Andrés Bello, Concepción, Chile
| | - Claudio Acuña-Castillo
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
84
|
Karan D. Inflammasomes: Emerging Central Players in Cancer Immunology and Immunotherapy. Front Immunol 2018; 9:3028. [PMID: 30631327 PMCID: PMC6315184 DOI: 10.3389/fimmu.2018.03028] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/07/2018] [Indexed: 01/04/2023] Open
Abstract
Inflammation has an established role in cancer development and progression and is a key player in regulating the entry and exit of immune cells in the tumor microenvironment, mounting a significant impact on anti-tumor immunity. Recent studies have shed light on the role of inflammasomes in the regulation of inflammation with a focus on the subsequent effects on the immunobiology of tumors. To generate strong anti-tumor immunity, cross-talk between innate, and adaptive immune cells is necessary. Interestingly, inflammasome bridges both arms of the immune system representing a unique opportunity to manipulate the role of inflammation in favor of tumor suppression. In this review, we discuss the impact of inflammasomes on the regulation of the levels of inflammatory cytokines-chemokines and the efficacy of immunotherapy response in cancer treatment.
Collapse
Affiliation(s)
- Dev Karan
- Department of Pathology, MCW Cancer Center and Prostate Cancer Center of Excellence, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
85
|
Mezouar S, Chantran Y, Michel J, Fabre A, Dubus JC, Leone M, Sereme Y, Mège JL, Ranque S, Desnues B, Chanez P, Vitte J. Microbiome and the immune system: From a healthy steady-state to allergy associated disruption. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.humic.2018.10.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
86
|
Jiandong L, Yang Y, Peng J, Xiang M, Wang D, Xiong G, Li S. Trichosanthes kirilowii lectin ameliorates streptozocin-induced kidney injury via modulation of the balance between M1/M2 phenotype macrophage. Biomed Pharmacother 2018; 109:93-102. [PMID: 30396096 DOI: 10.1016/j.biopha.2018.10.060] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/21/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Macrophage polarization has been reported to induce podocyte injury, which is a typical characteristic of diabetic nephropathy (DN). Trichosanthes kirilowii is an herb showing renal protective effect as well as immune-regulating effect. Therefore, it was hypothesized that the renal protective effect of Trichosanthes kirilowii was associated with its modulation on macrophage polarization. In the current study, we tested the hypothesis by subjecting DN rats to treatment of Trichosanthes kirilowii lectin (TKL), an active component of Trichosanthes kirilowii. METHOD DN was induced using streptozocin (STZ) method, and after 3 days, treatments were performed with different doses of TKL for eight weeks. The effect of TKL on the renal function, structure, and inflammation was assessed. To explain the pathway mediating the effect of TKL on renal tissues, the expressions of markers involved in macrophage polarization, podocyte proliferation, and Notch signaling were determined. Moreover, the DN rats were further administrated with Notch signaling inhibitor, Dibenzazepine (DIB), to verify the key role of Notch signaling in the renal protective effect of TKL. RESULTS STZ induced damages in renal function and structure, which was attenuated by TKL of different doses. Moreover, STZ also increased the production of TNF-α and iNOS while suppressed the production of IL-10 and arginase-1 (Arg-1). The induced inflammation by STZ was inhibited by TKL. The polarization of macrophage into M1 type during the development of DN was blocked by TKL, contributing to the increased proliferation potential of podocytes. Regarding Notch signaling, TKL administration inhibited the activation of the pathway by suppressing the expression of Notch1, NICD1, and Hes1. The administration of DIB had similar effect to that of TKL administration on renal function and structure. CONCLUSIONS The study for the first time showed that TKL attenuated deterioration in renal structure and function by increasing M2 macrophage proportion via inhibition of Notch signaling.
Collapse
Affiliation(s)
- Lu Jiandong
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yilong Yang
- Geriatrics Department of Traditional Chinese Medicine, Shenzhen Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jinting Peng
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Min Xiang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Dongcai Wang
- Centers for Disease Early Treatment, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Guoliang Xiong
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Shunmin Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| |
Collapse
|
87
|
Bagheri N, Salimzadeh L, Shirzad H. The role of T helper 1-cell response in Helicobacter pylori-infection. Microb Pathog 2018; 123:1-8. [PMID: 29936093 DOI: 10.1016/j.micpath.2018.06.033] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
Helicobacter pylori (H. pylori) is a human pathogen affecting over 50% of the world population. This pathogen is usually associated with chronic inflammation of the gastric mucosa that can lead to peptic ulcer disease (PUD) and gastric cancer (GC), especially in susceptible individuals. These outcomes have been attributed to the interaction of several factors, including host genetic susceptibility, local innate and adaptive immune responses, virulence factors of H. pylori, and environmental factors. T helper (Th) cell subsets and their signature cytokines especially IFN-γ, contribute to anti-bacterial response, but at the mean time sustaining chronic inflammatory responses in the site of infection. It has been acknowledged that H. pylori-infection results in a Th1-dominant response and that inflammation of the gastric mucosa depends mainly on Th1 cell responses. But, the mechanism of the role of Th1 cell responses in H. pylori-infection has not yet been clearly explained. In this review, we will focus on the role of Th1 involved in H. pylori-infection, its interaction with Th17/Treg cells and its association with the clinical consequences of the infection.
Collapse
Affiliation(s)
- Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Loghman Salimzadeh
- Department of Microbiology and Immunology Programme, National University of Singapore, Singapore
| | - Hedayatollah Shirzad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
88
|
Lugo-Villarino G, Troegeler A, Balboa L, Lastrucci C, Duval C, Mercier I, Bénard A, Capilla F, Al Saati T, Poincloux R, Kondova I, Verreck FAW, Cougoule C, Maridonneau-Parini I, Sasiain MDC, Neyrolles O. The C-Type Lectin Receptor DC-SIGN Has an Anti-Inflammatory Role in Human M(IL-4) Macrophages in Response to Mycobacterium tuberculosis. Front Immunol 2018; 9:1123. [PMID: 29946317 PMCID: PMC6006465 DOI: 10.3389/fimmu.2018.01123] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 05/03/2018] [Indexed: 01/03/2023] Open
Abstract
DC-SIGN (CD209/CLEC4L) is a C-type lectin receptor (CLR) that serves as a reliable cell-surface marker of interleukin 4 (IL-4)-activated human macrophages [M(IL-4)], which historically represent the most studied subset within the M2 spectrum of macrophage activation. Although DC-SIGN plays important roles in Mycobacterium tuberculosis (Mtb) interactions with dendritic cells, its contribution to the Mtb–macrophage interaction remains poorly understood. Since high levels of IL-4 are correlated with tuberculosis (TB) susceptibility and progression, we investigated the role of DC-SIGN in M(IL-4) macrophages in the TB context. First, we demonstrate that DC-SIGN expression is present both in CD68+ macrophages found in tuberculous pulmonary lesions of non-human primates, and in the CD14+ cell population isolated from pleural effusions obtained from TB patients (TB-PE). Likewise, we show that DC-SIGN expression is accentuated in M(IL-4) macrophages derived from peripheral blood CD14+ monocytes isolated from TB patients, or in macrophages stimulated with acellular TB-PE, arguing for the pertinence of DC-SIGN-expressing macrophages in TB. Second, using a siRNA-mediated gene silencing approach, we performed a transcriptomic analysis of DC-SIGN-depleted M(IL-4) macrophages and revealed the upregulation of pro-inflammatory signals in response to challenge with Mtb, as compared to control cells. This pro-inflammatory gene signature was confirmed by RT-qPCR, cytokine/chemokine-based protein array, and ELISA analyses. We also found that inactivation of DC-SIGN renders M(IL-4) macrophages less permissive to Mtb intracellular growth compared to control cells, despite the equal level of bacteria uptake. Last, at the molecular level, we show that DC-SIGN interferes negatively with the pro-inflammatory response and control of Mtb intracellular growth mediated by another CLR, Dectin-1 (CLEC7A). Collectively, this study highlights a dual role for DC-SIGN as, on the one hand, being a host factor granting advantage for Mtb to parasitize macrophages and, on the other hand, representing a molecular switch to turn off the pro-inflammatory response in these cells to prevent potential immunopathology associated to TB.
Collapse
Affiliation(s)
- Geanncarlo Lugo-Villarino
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Buenos Aires, Argentina
- *Correspondence: Geanncarlo Lugo-Villarino,
| | - Anthony Troegeler
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Buenos Aires, Argentina
| | - Luciana Balboa
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Buenos Aires, Argentina
- IMEX-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Claire Lastrucci
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Buenos Aires, Argentina
- Centre for Genomic Regulation, Barcelona, Spain
| | - Carine Duval
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Ingrid Mercier
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Alan Bénard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- Department of Surgery, University, Hospital Erlangen, Friedrich-Alexander, University Erlangen-Nürnberg, Erlangen, Germany
| | | | | | - Renaud Poincloux
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | | | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Buenos Aires, Argentina
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Buenos Aires, Argentina
| | - Maria del Carmen Sasiain
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Buenos Aires, Argentina
- IMEX-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM–TB/HIV” (1167), Buenos Aires, Argentina
| |
Collapse
|
89
|
Li H, Jiang T, Li MQ, Zheng XL, Zhao GJ. Transcriptional Regulation of Macrophages Polarization by MicroRNAs. Front Immunol 2018; 9:1175. [PMID: 29892301 PMCID: PMC5985397 DOI: 10.3389/fimmu.2018.01175] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/11/2018] [Indexed: 01/26/2023] Open
Abstract
Diversity and plasticity are the hallmarks of cells from the monocyte-macrophage lineage. Macrophages undergo classical M1 or alternative M2 activation in response to the microenvironment signals. Several transcription factors, such as peroxisome proliferator-activated receptors, signal transducers and activators of transcription, CCAAT-enhancer-binding proteins, interferon regulatory factors, Kruppel-like factors, GATA binding protein 3, nuclear transcription factor-κB, and c-MYC, were found to promote the expression of specific genes, which dictate the functional polarization of macrophages. Importantly, these transcription factors can be regulated by microRNAs (miRNAs), a group of small non-coding RNAs, which regulate gene expression through translation repression or mRNA degradation. Recent studies have also revealed that miRNAs control macrophage polarization by regulating transcription factors in response to the microenvironment signals. This review will summarize recent progress of miRNAs in the transcriptional regulation of macrophage polarization and provide the insights into the development of macrophage-centered diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Heng Li
- The Clinic Medical College, Guilin Medical University, Guilin, Guangxi, China
| | - Ting Jiang
- Department of Practice Educational, Office of Academic Affairs, Guilin Medical University, Guilin, Guangxi, China
| | - Meng-Qi Li
- Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, Calgary, AB, Canada.,Key Laboratory of Molecular Targets and Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guo-Jun Zhao
- Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China.,Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, Calgary, AB, Canada
| |
Collapse
|
90
|
Shan G, Tang T, Qian H, Xia Y. Certain BCG-reactive responses are associated with bladder cancer prognosis. Cancer Immunol Immunother 2018; 67:797-803. [PMID: 29478100 PMCID: PMC11028363 DOI: 10.1007/s00262-018-2127-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 02/09/2018] [Indexed: 11/30/2022]
Abstract
A subset of bladder patients does not respond to BCG treatment effectively and the underlying reason behind this observation is currently unclear. CD4+ T cells are composed of various subsets that each expresses a distinctive set of cytokines and can potently shift the immune response toward various directions. In this study, we examined the CD4+ T-cell cytokine response in bladder cancer patients toward BCG stimulation. We found that bladder cancer patients presented a variety of responses toward BCG, with no uniform characteristics. Those patients with high IFN-γ and IL-21 expression in CD4+ T cells presented significantly better prognosis than patients with low cytokine secretion in CD4+ T cells. Tumor-infiltrating CD4+ T cells were significantly less potent in expressing IFN-γ, IL-4, and IL-17, and more potent in expressing IL-10 than circulating CD4+ T cells. In addition, we found no difference in CD80, CD86, or MHC II expression by macrophages from patients with different IFN-γ and IL-21 levels. However, the secretion of IL-12, a Th1-skewing cytokine, was released at significantly higher level by macrophages from patients with high IFN-γ or high IL-21 secretion. We also identified that modulating monocytes/macrophages by GM-CSF-mediated polarization resulted in significantly elevated expression of IFN-γ and IL-21 from CD4+ T cells. Overall, these results suggested that the specific types of responses mounted by CD4+ T cells were critical to the final outcome of bladder cancer patients and can be influenced by monocyte/macrophage polarization.
Collapse
Affiliation(s)
- Guang Shan
- Departments of Urology II, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei, China.
| | - Tian Tang
- Departments of Oncology II, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Huijun Qian
- Departments of Urology II, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei, China
| | - Yue Xia
- Departments of Urology II, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei, China
| |
Collapse
|
91
|
Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT, Sahebkar A. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol 2018; 233:6425-6440. [PMID: 29319160 DOI: 10.1002/jcp.26429] [Citation(s) in RCA: 3154] [Impact Index Per Article: 450.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022]
Abstract
Macrophages are heterogeneous and their phenotype and functions are regulated by the surrounding micro-environment. Macrophages commonly exist in two distinct subsets: 1) Classically activated or M1 macrophages, which are pro-inflammatory and polarized by lipopolysaccharide (LPS) either alone or in association with Th1 cytokines such as IFN-γ, GM-CSF, and produce pro-inflammatory cytokines such as interleukin-1β (IL-1β), IL-6, IL-12, IL-23, and TNF-α; and 2) Alternatively activated or M2 macrophages, which are anti-inflammatory and immunoregulatory and polarized by Th2 cytokines such as IL-4 and IL-13 and produce anti-inflammatory cytokines such as IL-10 and TGF-β. M1 and M2 macrophages have different functions and transcriptional profiles. They have unique abilities by destroying pathogens or repair the inflammation-associated injury. It is known that M1/M2 macrophage balance polarization governs the fate of an organ in inflammation or injury. When the infection or inflammation is severe enough to affect an organ, macrophages first exhibit the M1 phenotype to release TNF-α, IL-1β, IL-12, and IL-23 against the stimulus. But, if M1 phase continues, it can cause tissue damage. Therefore, M2 macrophages secrete high amounts of IL-10 and TGF-β to suppress the inflammation, contribute to tissue repair, remodeling, vasculogenesis, and retain homeostasis. In this review, we first discuss the basic biology of macrophages including origin, differentiation and activation, tissue distribution, plasticity and polarization, migration, antigen presentation capacity, cytokine and chemokine production, metabolism, and involvement of microRNAs in macrophage polarization and function. Secondly, we discuss the protective and pathogenic role of the macrophage subsets in normal and pathological pregnancy, anti-microbial defense, anti-tumor immunity, metabolic disease and obesity, asthma and allergy, atherosclerosis, fibrosis, wound healing, and autoimmunity.
Collapse
Affiliation(s)
- Abbas Shapouri-Moghaddam
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Mohammadian
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Vazini
- Nursing Department, Basic Sciences Faculty, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Mahdi Taghadosi
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed-Alireza Esmaeili
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mardani
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bita Seifi
- Department of Anatomy, Islamic Azad University, Mashhad Branch, Iran
| | - Asadollah Mohammadi
- Inflammation and Inflammatory Disease Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil T Afshari
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
92
|
Pahari S, Kaur G, Negi S, Aqdas M, Das DK, Bashir H, Singh S, Nagare M, Khan J, Agrewala JN. Reinforcing the Functionality of Mononuclear Phagocyte System to Control Tuberculosis. Front Immunol 2018; 9:193. [PMID: 29479353 PMCID: PMC5811511 DOI: 10.3389/fimmu.2018.00193] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022] Open
Abstract
The mononuclear phagocyte system (MPS) constitutes dendritic cells, monocytes, and macrophages. This system contributes to various functions that are essential for maintaining homeostasis, activation of innate immunity, and bridging it with the adaptive immunity. Consequently, MPS is highly important in bolstering immunity against the pathogens. However, MPS is the frontline cells in destroying Mycobacterium tuberculosis (Mtb), yet the bacterium prefers to reside in the hostile environment of macrophages. Therefore, it may be very interesting to study the struggle between Mtb and MPS to understand the outcome of the disease. In an event when MPS predominates Mtb, the host remains protected. By contrast, the situation becomes devastating when the pathogen tames and tunes the host MPS, which ultimately culminates into tuberculosis (TB). Hence, it becomes extremely crucial to reinvigorate MPS functionality to overwhelm Mtb and eliminate it. In this article, we discuss the strategies to bolster the function of MPS by exploiting the molecules associated with the innate immunity and highlight the mechanisms involved to overcome the Mtb-induced suppression of host immunity. In future, such approaches may provide an insight to develop immunotherapeutics to treat TB.
Collapse
Affiliation(s)
- Susanta Pahari
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Gurpreet Kaur
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Shikha Negi
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mohammad Aqdas
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Deepjyoti K Das
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Hilal Bashir
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sanpreet Singh
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mukta Nagare
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Junaid Khan
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
93
|
Mulder R, Banete A, Seaver K, Basta S. M(IL-4) Tissue Macrophages Support Efficient Interferon-Gamma Production in Antigen-Specific CD8 + T Cells with Reduced Proliferative Capacity. Front Immunol 2017; 8:1629. [PMID: 29250063 PMCID: PMC5714867 DOI: 10.3389/fimmu.2017.01629] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/09/2017] [Indexed: 12/31/2022] Open
Abstract
CD8+ cytotoxic T cell (CTL) responses are necessary for the lysis of virally infected cells and control of infection. CTLs are activated when their TCRs bind a major histocompatibility complex (MHC)-I/peptide complex on the surface of antigen presenting cells such as macrophages (MΦ). It is now apparent that MΦ display remarkable plasticity in response to environmental signals to polarize into classically activated M(LPS + IFN-γ) or alternatively activated M(IL-4). However, little is known about how MΦ activation status influences their antigen presentation function to CD8+ T cell in models of virus infection. Consequently, we tested how polarization of spleen-derived (Sp)-MΦ impacts direct presentation of viral antigens to influence effector and proliferative CD8+ T-cell responses. We show that M(IL-4) Sp-MΦ retain MHC-I surface expression and the ability to stimulate IFN-γ production by CTL following peptide stimulation and lymphocytic choriomeningitis virus infection to levels similar to M0 and M(LPS + IFN-γ) MΦ. However, memory CD8+ T cells cultured in the presence of M(IL-4) MΦ underwent significantly reduced proliferation and produced similar IFN-γ levels as coculturing with M0 or M(LPS + IFN-γ) cells. Thus, these results show a novel ability of polarized MΦ to regulate CD8+ T-cell proliferation and effector functions during virus infection.
Collapse
Affiliation(s)
- Rylend Mulder
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Andra Banete
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Kyle Seaver
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
94
|
de Sousa JR, Sotto MN, Simões Quaresma JA. Leprosy As a Complex Infection: Breakdown of the Th1 and Th2 Immune Paradigm in the Immunopathogenesis of the Disease. Front Immunol 2017; 8:1635. [PMID: 29234318 PMCID: PMC5712391 DOI: 10.3389/fimmu.2017.01635] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022] Open
Abstract
Leprosy is a chronic infectious disease whose evolution involves complex immune mechanisms of the host that influence the clinical presentation of the disease. For many years, the main interpretation of the host defense response was based on characterization of the established immune paradigm between T helper (Th) 1 and Th2 lymphocytes. However, with advances in the knowledge of immunology, new approaches have emerged along with the development of new immunological pathways that have changed the interpretation of the long-established paradigm of the polar forms of the disease, especially with the identification of new subtypes of T lymphocytes such as Th9, Th17, Th22, and Tregs. Thus, this review discusses the role of these new subtypes of T helper lymphocytes and how the development of the immune response of these cells modifies the pattern of the Th1/Th2 response in the immunopathogenesis of leprosy.
Collapse
Affiliation(s)
| | - Mirian Nacagami Sotto
- Faculty of Medicine, Department of Pathology, Sao Paulo University, São Paulo, Brazil
| | - Juarez Antonio Simões Quaresma
- Tropical Medicine Center, Federal University of Pará, Belém, Brazil.,Center of Biological and Health Sciences, State University of Pará, Belém, Brazil
| |
Collapse
|
95
|
Truflandier K, Beaumont E, Maghni K, De Marchie M, Charbonney E, Spahija J. Spinal cord injury modulates the lung inflammatory response in mechanically ventilated rats: a comparative animal study. Physiol Rep 2017; 4:4/24/e13009. [PMID: 28039398 PMCID: PMC5210386 DOI: 10.14814/phy2.13009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 09/25/2016] [Indexed: 12/26/2022] Open
Abstract
Mechanical ventilation (MV) is widely used in spinal injury patients to compensate for respiratory muscle failure. MV is known to induce lung inflammation, while spinal cord injury (SCI) is known to contribute to local inflammatory response. Interaction between MV and SCI was evaluated in order to assess the impact it may have on the pulmonary inflammatory profile. Sprague Dawley rats were anesthetized for 24 h and randomized to receive either MV or not. The MV group included C4-C5 SCI, T10 SCI and uninjured animals. The nonventilated (NV) group included T10 SCI and uninjured animals. Inflammatory cytokine profile, inflammation related to the SCI level, and oxidative stress mediators were measured in the bronchoalveolar lavage (BAL). The cytokine profile in BAL of MV animals showed increased levels of TNF-α, IL-1β, IL-6 and a decrease in IL-10 (P = 0.007) compared to the NV group. SCI did not modify IL-6 and IL-10 levels either in the MV or the NV groups, but cervical injury induced a decrease in IL-1β levels in MV animals. Cervical injury also reduced MV-induced pulmonary oxidative stress responses by decreasing isoprostane levels while increasing heme oxygenase-1 level. The thoracic SCI in NV animals increased M-CSF expression and promoted antioxidant pulmonary responses with low isoprostane and high heme oxygenase-1 levels. SCI shows a positive impact on MV-induced pulmonary inflammation, modulating specific lung immune and oxidative stress responses. Inflammation induced by MV and SCI interact closely and may have strong clinical implications since effective treatment of ventilated SCI patients may amplify pulmonary biotrauma.
Collapse
Affiliation(s)
- Karine Truflandier
- Research Center, Sacré-Cœur Hospital of Montreal, Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Eric Beaumont
- Research Center, Sacré-Cœur Hospital of Montreal, Department of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Karim Maghni
- Research Center, Sacré-Cœur Hospital of Montreal, Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Michel De Marchie
- Department of Adult Critical Care, Jewish General Hospital McGill University, Montréal, Quebec, Canada
| | - Emmanuel Charbonney
- Research Center, Sacré-Cœur Hospital of Montreal, Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Jadranka Spahija
- Research Center, Sacré-Cœur Hospital of Montreal, Department of Medicine, Université de Montréal, Montreal, Quebec, Canada .,School of Physical and Occupational Therapy, McGill University, Montreal, Quebec, Canada.,Center for Interdisciplinary Research in Rehabilitation in Montreal, Jewish Rehabilitation Hospital, Laval, Quebec, Canada
| |
Collapse
|
96
|
Zhao Y, Guo Y, Jiang Y, Zhu X, Liu Y, Zhang X. Mitophagy regulates macrophage phenotype in diabetic nephropathy rats. Biochem Biophys Res Commun 2017; 494:42-50. [PMID: 29061302 DOI: 10.1016/j.bbrc.2017.10.088] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/16/2017] [Indexed: 10/18/2022]
Abstract
Imbalance of M1/M2 macrophages phenotype activation is a key point in diabetic nephropathy (DN). Macrophages mainly exhibit M1 phenotype, which contributes to the inflammation and fibrosis in DN. Studies indicate that autophage plays an important role in M1/M2 activation. However, the effect of mitophage on M1/M2 macrophage phenotype transformation in DN is unknown. This study investigates the role of mitophage on macrophage polarization in DN. In vivo experiments show that macrophages are exhibited to M1 phenotype and display a lower level of mitophagy in the kidney of streptozocin (STZ)-induced diabetic rats. Additionally, inducible nitric oxide synthase (iNOS) expression is positive correlated with the P62 expression, while negative correlated with LC3. Electronic microscope analysis shows mitochondria swelling, crista decrease and lysosome reduction in DN rats compared with NC rats. In vitro, RAW264.7 macrophages switch to M1 phenotype under high glucose conditions. Mitophagy is downregulated in such high glucose induced M1 macrophages. Furthermore, macrophages tend to switch to the M1 phenotype, expressing higher iNOS and TNF-α when impair mitophagy by 3-MA. Rapamycin, an activator of mitophagy, signifcantly blocks high-glucose induced M1 makers (iNOS and TNF-α) expression, meanwhile enhances M2 makers (MR and Arg-1) expression. These results demonstrate that mitophage participates in the regulation of M1/M2 macrophage phenotype in diabetic nephropathy.
Collapse
Affiliation(s)
- Yu Zhao
- Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, 210009, China
| | - Yinfeng Guo
- Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, 210009, China
| | - Yuteng Jiang
- Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, 210009, China
| | - Xiaodong Zhu
- Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, 210009, China
| | - Yuqiu Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, 210009, China
| | - Xiaoliang Zhang
- Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
97
|
Deng SY, Zhang LM, Ai YH, Pan PH, Zhao SP, Su XL, Wu DD, Tan HY, Zhang LN, Tsung A. Role of interferon regulatory factor-1 in lipopolysaccharide-induced mitochondrial damage and oxidative stress responses in macrophages. Int J Mol Med 2017; 40:1261-1269. [PMID: 28849179 PMCID: PMC5593470 DOI: 10.3892/ijmm.2017.3110] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/04/2017] [Indexed: 01/20/2023] Open
Abstract
Sepsis causes many early deaths; both macrophage mitochondrial damage and oxidative stress responses are key factors in its pathogenesis. Although the exact mechanisms responsible for sepsis-induced mitochondrial damage are unknown, the nuclear transcription factor, interferon regulatory factor-1 (IRF-1) has been reported to cause mitochondrial damage in several diseases. Previously, we reported that in addition to promoting systemic inflammation, IRF-1 promoted the apoptosis of and inhibited autophagy in macrophages. In the present study, we hypothesized that lipopolysaccharide (LPS)-induced IRF-1 activation in macrophages may promote mitochondrial damage and oxidative stress. In vitro, LPS was found to promote IRF-1 activation, reactive oxygen species (ROS) production, adenosine triphosphate (ATP) depletion, superoxide dismutase (SOD) consumption, malondialdehyde (MDA) accumulation and mitochondrial depolarization in macrophages in a time- and dose-dependent manner. These effects were abrogated in cells in which IRF-1 was knocked down. Furthermore, IRF-1 overexpression increased LPS-induced oxidative stress responses and mitochondrial damage. In vivo, peritoneal macrophages obtained from IRF-1 knockout (KO) mice produced less ROS and had less mitochondrial depolarization and damage following the administration of LPS, when compared to their wild-type (WT) counterparts. In addition, IRF-1 KO mice exhibited a decreased release of mitochondrial DNA (mtDNA) following the administration of LPS. Thus, IRF-1 may be a critical factor in augmenting LPS-induced oxidative stress and mitochondrial damage in macrophages.
Collapse
Affiliation(s)
- Song-Yun Deng
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Le-Meng Zhang
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yu-Hang Ai
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Pin-Hua Pan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Shuang-Ping Zhao
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiao-Li Su
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Dong-Dong Wu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hong-Yi Tan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Li-Na Zhang
- Department of Intensive Care Unit, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
98
|
Nwizu NN, Marshall JR, Moysich K, Genco RJ, Hovey KM, Mai X, LaMonte MJ, Freudenheim JL, Wactawski-Wende J. Periodontal Disease and Incident Cancer Risk among Postmenopausal Women: Results from the Women's Health Initiative Observational Cohort. Cancer Epidemiol Biomarkers Prev 2017; 26:1255-1265. [PMID: 28765338 PMCID: PMC5551444 DOI: 10.1158/1055-9965.epi-17-0212] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/02/2017] [Accepted: 05/15/2017] [Indexed: 02/07/2023] Open
Abstract
Background: Periodontal pathogens have been isolated from precancerous and cancerous lesions and also shown to promote a procarcinogenic microenvironment. Few studies have examined periodontal disease as a risk factor for total cancer, and none have focused on older women. We examined whether periodontal disease is associated with incident cancer among postmenopausal women in the Women's Health Initiative Observational Study.Methods: Our prospective cohort study comprised 65,869 women, ages 54 to 86 years. Periodontal disease information was obtained via self-report questionnaires administered between 1999 and 2003, whereas ascertainment of cancer outcomes occurred through September 2013, with a maximum follow-up period of 15 years. Physician-adjudicated incident total cancers were the main outcomes and site-specific cancers were secondary outcomes. HRs and 95% confidence intervals (CI) were calculated using Cox proportional hazards regression. All analyses were conducted two-sided.Results: During a mean follow-up of 8.32 years, 7,149 cancers were identified. Periodontal disease history was associated with increased total cancer risk (multivariable-adjusted HR, 1.14; 95% CI, 1.08-1.20); findings were similar in analyses limited to 34,097 never-smokers (HR, 1.12; 95% CI, 1.04-1.22). Associations were observed for breast (HR, 1.13; 95% CI, 1.03-1.23), lung (HR, 1.31; 95% CI, 1.14-1.51), esophagus (HR, 3.28; 95% CI, 1.64-6.53), gallbladder (HR, 1.73; 95% CI, 1.01-2.95), and melanoma skin (HR, 1.23; 95% CI, 1.02-1.48) cancers. Stomach cancer was borderline (HR, 1.58; 95% CI, 0.94-2.67).Conclusions: Periodontal disease increases risk of total cancer among older women, irrespective of smoking, and certain anatomic sites appear to be vulnerable. Impact: Our findings support the need for further understanding of the effect of periodontal disease on cancer outcomes. Cancer Epidemiol Biomarkers Prev; 26(8); 1255-65. ©2017 AACR.
Collapse
Affiliation(s)
- Ngozi N Nwizu
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, Texas
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
- Department of Epidemiology and Environmental Health, University at Buffalo, State University of New York, Buffalo, New York
| | - James R Marshall
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
| | - Kirsten Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
| | - Robert J Genco
- Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, New York
| | - Kathleen M Hovey
- Department of Epidemiology and Environmental Health, University at Buffalo, State University of New York, Buffalo, New York
| | - Xiaodan Mai
- Department of Epidemiology and Environmental Health, University at Buffalo, State University of New York, Buffalo, New York
| | - Michael J LaMonte
- Department of Epidemiology and Environmental Health, University at Buffalo, State University of New York, Buffalo, New York
| | - Jo L Freudenheim
- Department of Epidemiology and Environmental Health, University at Buffalo, State University of New York, Buffalo, New York
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, State University of New York, Buffalo, New York.
- Department of Gynecology-Obstetrics, University at Buffalo, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
99
|
La Manna MP, Orlando V, Dieli F, Di Carlo P, Cascio A, Cuzzi G, Palmieri F, Goletti D, Caccamo N. Quantitative and qualitative profiles of circulating monocytes may help identifying tuberculosis infection and disease stages. PLoS One 2017; 12:e0171358. [PMID: 28208160 PMCID: PMC5313257 DOI: 10.1371/journal.pone.0171358] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/19/2017] [Indexed: 01/31/2023] Open
Abstract
Tuberculosis (TB) is one of the most important cause of morbidity and death among infectious diseases, and continuous efforts are needed to improve diagnostic tools and therapy. Previous published studies showed that the absolute cells number of monocytes or lymphocytes in peripheral blood or yet the ratio of monocytes to lymphocytes displayed the ability to predict the risk of active TB. In the present study we evaluated the ratio of monocytes to lymphocytes variation and we also analyzed the ex-vivo expression of CD64 on monocytes as tools to identify biomarkers for discriminating TB stages. Significant differences were found when the average ratio of monocytes to lymphocytes of active TB patients was compared with latent TB infection (LTBI) subjects, cured TB and healthy donors (HD). By the receiver operator characteristics (ROC) curve analysis the cut-off value of 0.285, allowed the discrimination of active TB from HD, with a sensitivity of 91.04% and a specificity of 93.55% (95% of confidence interval: 0.92-0.99). The ROC curve analysis comparing TB patients and LTBI groups, led to a sensitivity and the specificity of the assay of 85.07% and 85.71%, respectively (95% of confidence interval: 0.85 to 0.96). The upregulation of CD64 expression on circulating monocytes in active TB patients could represent an additional biomarker for diagnosis of active TB. In conclusion, we found that the ML ratio or monocyte absolute count or phenotypic measures show predictive value for active TB.
Collapse
Affiliation(s)
- Marco Pio La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research(CLADIBIOR), Azienda Universitaria Ospedaliera Policlinico P. Giaccone, Palermo, Italy
| | - Valentina Orlando
- Central Laboratory of Advanced Diagnosis and Biomedical Research(CLADIBIOR), Azienda Universitaria Ospedaliera Policlinico P. Giaccone, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research(CLADIBIOR), Azienda Universitaria Ospedaliera Policlinico P. Giaccone, Palermo, Italy
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Paola Di Carlo
- Department of Sciences for Health Promotion and Mother-Child Care “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Antonio Cascio
- Department of Sciences for Health Promotion and Mother-Child Care “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Gilda Cuzzi
- Translational Research Unit, National Institute for Infectious Diseases L. Spallanzani, Rome, Italy
| | - Fabrizio Palmieri
- Translational Research Unit, National Institute for Infectious Diseases L. Spallanzani, Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases L. Spallanzani, Rome, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research(CLADIBIOR), Azienda Universitaria Ospedaliera Policlinico P. Giaccone, Palermo, Italy
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| |
Collapse
|
100
|
Szebeni GJ, Vizler C, Nagy LI, Kitajka K, Puskas LG. Pro-Tumoral Inflammatory Myeloid Cells as Emerging Therapeutic Targets. Int J Mol Sci 2016; 17:ijms17111958. [PMID: 27886105 PMCID: PMC5133952 DOI: 10.3390/ijms17111958] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/01/2016] [Accepted: 11/16/2016] [Indexed: 12/29/2022] Open
Abstract
Since the observation of Virchow, it has long been known that the tumor microenvironment constitutes the soil for the infiltration of inflammatory cells and for the release of inflammatory mediators. Under certain circumstances, inflammation remains unresolved and promotes cancer development. Here, we review some of these indisputable experimental and clinical evidences of cancer related smouldering inflammation. The most common myeloid infiltrate in solid tumors is composed of myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs). These cells promote tumor growth by several mechanisms, including their inherent immunosuppressive activity, promotion of neoangiogenesis, mediation of epithelial-mesenchymal transition and alteration of cellular metabolism. The pro-tumoral functions of TAMs and MDSCs are further enhanced by their cross-talk offering a myriad of potential anti-cancer therapeutic targets. We highlight these main pro-tumoral mechanisms of myeloid cells and give a general overview of their phenotypical and functional diversity, offering examples of possible therapeutic targets. Pharmacological targeting of inflammatory cells and molecular mediators may result in therapies improving patient condition and prognosis. Here, we review experimental and clinical findings on cancer-related inflammation with a major focus on creating an inventory of current small molecule-based therapeutic interventions targeting cancer-related inflammatory cells: TAMs and MDSCs.
Collapse
Affiliation(s)
- Gabor J Szebeni
- Avidin Ltd., Also kikoto sor 11/D., H-6726 Szeged, Hungary.
- Synaptogenex Ltd., Őzsuta utca 20995/1, H-1037 Budapest, Hungary.
| | - Csaba Vizler
- Department of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Temesvari krt. 62., H-6726 Szeged, Hungary.
| | - Lajos I Nagy
- Avidin Ltd., Also kikoto sor 11/D., H-6726 Szeged, Hungary.
| | - Klara Kitajka
- Department of Genetics, Biological Research Center, Hungarian Academy of Sciences, Temesvari krt. 62., H-6726 Szeged, Hungary.
| | - Laszlo G Puskas
- Avidin Ltd., Also kikoto sor 11/D., H-6726 Szeged, Hungary.
- Department of Genetics, Biological Research Center, Hungarian Academy of Sciences, Temesvari krt. 62., H-6726 Szeged, Hungary.
| |
Collapse
|